101
|
Madsen MAJ, Wiggermann V, Bramow S, Christensen JR, Sellebjerg F, Siebner HR. Imaging cortical multiple sclerosis lesions with ultra-high field MRI. Neuroimage Clin 2021; 32:102847. [PMID: 34653837 PMCID: PMC8517925 DOI: 10.1016/j.nicl.2021.102847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cortical lesions are abundant in multiple sclerosis (MS), yet difficult to visualize in vivo. Ultra-high field (UHF) MRI at 7 T and above provides technological advances suited to optimize the detection of cortical lesions in MS. PURPOSE To provide a narrative and quantitative systematic review of the literature on UHF MRI of cortical lesions in MS. METHODS A systematic search of all literature on UHF MRI of cortical lesions in MS published before September 2020. Quantitative outcome measures included cortical lesion numbers reported using 3 T and 7 T MRI and between 7 T MRI sequences, along with sensitivity of UHF MRI towards cortical lesions verified by histopathology. RESULTS 7 T MRI detected on average 52 ± 26% (mean ± 95% confidence interval) more cortical lesions than the best performing image contrast at 3 T, with the largest increase in type II-IV intracortical lesion detection. Across all studies, the mean cortical lesion number was 17 ± 6 per patient. In progressive MS cohorts, approximately four times more cortical lesions were reported than in CIS/early RRMS, and RRMS. Yet, there was no difference in lesion type ratio between these MS subtypes. Furthermore, superiority of one MRI sequence over another could not be established from available data. Post-mortem lesion detection with UHF MRI agreed only modestly with pathological examinations. Mean pro- and retrospective sensitivity was 33 ± 6% and 71 ± 10%, respectively, with the highest sensitivity towards type I and type IV lesions. CONCLUSION UHF MRI improves cortical lesion detection in MS considerably compared to 3 T MRI, particularly for type II-IV lesions. Despite modest sensitivity, 7 T MRI is still capable of visualizing all aspects of cortical lesion pathology and could potentially aid clinicians in diagnosing and monitoring MS, and progressive MS in particular. However, standardization of acquisition and segmentation protocols is needed.
Collapse
Affiliation(s)
- Mads A J Madsen
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital - Amager & Hvidovre, Kettegard Allé 30, 2650 Hvidovre, Denmark.
| | - Vanessa Wiggermann
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital - Amager & Hvidovre, Kettegard Allé 30, 2650 Hvidovre, Denmark
| | - Stephan Bramow
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Valdemar Hansens Vej 1-23, 2600 Glostrup, Denmark
| | - Jeppe Romme Christensen
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Valdemar Hansens Vej 1-23, 2600 Glostrup, Denmark
| | - Finn Sellebjerg
- Danish Multiple Sclerosis Center, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Valdemar Hansens Vej 1-23, 2600 Glostrup, Denmark; Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Hartwig R Siebner
- Danish Research Centre for Magnetic Resonance, Copenhagen University Hospital - Amager & Hvidovre, Kettegard Allé 30, 2650 Hvidovre, Denmark; Department of Neurology, Copenhagen University Hospital - Bispebjerg, Bispebjerg Bakke 23, 2400 Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| |
Collapse
|
102
|
Kouchaki E, Dashti F, Mirazimi SMA, Alirezaei Z, Jafari SH, Hamblin MR, Mirzaei H. Neurofilament light chain as a biomarker for diagnosis of multiple sclerosis. EXCLI JOURNAL 2021; 20:1308-1325. [PMID: 34602928 PMCID: PMC8481790 DOI: 10.17179/excli2021-3973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
The treatments for multiple sclerosis (MS) have improved over the past 25 years, but now the main question for physicians is deciding who should receive treatment, for how long, and when to switch to other options. These decisions are typically based on treatment tolerance and a reasonable expectation of long-term efficacy. A significant unmet need is the lack of accurate laboratory measurements for diagnosis, and monitoring of treatment response, including deterioration and disease progression. There are few validated biomarkers for MS, and in practice, physicians employ two biomarkers discovered fifty years ago for MS diagnosis, often in combination with MRI scans. These biomarkers are intrathecal IgG and oligoclonal bands in the CSF (cerebrospinal fluid). Neurofilament light chain (NfL) is a relatively new biomarker for MS diagnosis and follow up. Neurofilaments are neuron-specific cytoskeleton proteins that can be measured in various body compartments. NfL is a new biomarker for MS that can be measured in serum samples, but this still needs further study to specify the laboratory cut-off values in clinical practice. In the present review we discuss the evidence for NfL as a reliable biomarker for the early detection and management of MS. Moreover, we highlight the correlation between MRI and NfL, and ask whether they can be combined.
Collapse
Affiliation(s)
- Ebrahim Kouchaki
- MS Fellowship, Department of Neurology, School of Medicine, Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Paramedical School, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran
| |
Collapse
|
103
|
Grothe M, Ellenberger D, von Podewils F, Stahmann A, Rommer PS, Zettl UK. Epilepsy as a predictor of disease progression in multiple sclerosis. Mult Scler 2021; 28:942-949. [PMID: 34595974 DOI: 10.1177/13524585211046739] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Epilepsy development during the course of multiple sclerosis (MS) is considered to be the result of cortical pathology. However, no long-term data exist on whether epilepsy in MS also leads to increasing disability over time. OBJECTIVE To examine if epilepsy leads to more rapid disease progression. METHODS We analyzed the data of 31,052 patients on the German Multiple Sclerosis Register in a case-control study. RESULTS Secondary progressive disease course (odds ratio (OR) = 2.23), age (OR = 1.12 per 10 years), and disability (OR = 1.29 per Expanded Disability Status Scale (EDSS) point) were associated with the 5-year prevalence of epilepsy. Patients who developed epilepsy during the course of the disease had a higher EDSS score at disease onset compared to matched control patients (EDSS 2.0 vs 1.5), progressed faster in each dimension, and consequently showed higher disability (EDSS 4.4 vs 3.4) and lower employment status (40% vs 65%) at final follow-up. After 15 years of MS, 64% of patients without compared to 54% of patients with epilepsy were not severely limited in walking distance. CONCLUSION This work highlights the association of epilepsy on disability progression in MS, and the need for additional data to further clarify the underlying mechanisms.
Collapse
Affiliation(s)
- Matthias Grothe
- Department of Neurology, University Medicine Greifswald, Ferdinand-Sauerbruchstraße, 17475 Greifswald, Germany.,Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - David Ellenberger
- German MS Register by the German MS Society, MS Research and Project Development gGmbH [MSFP], Hanover, Germany
| | - Felix von Podewils
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Stahmann
- German MS Register by the German MS Society, MS Research and Project Development gGmbH [MSFP], Hanover, Germany
| | - Paulus S Rommer
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany/Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Uwe K Zettl
- Department of Neurology, Neuroimmunological Section, University of Rostock, Rostock, Germany
| |
Collapse
|
104
|
Huiskamp M, Eijlers AJC, Broeders TAA, Pasteuning J, Dekker I, Uitdehaag BMJ, Barkhof F, Wink AM, Geurts JJG, Hulst HE, Schoonheim MM. Longitudinal Network Changes and Conversion to Cognitive Impairment in Multiple Sclerosis. Neurology 2021; 97:e794-e802. [PMID: 34099528 PMCID: PMC8397585 DOI: 10.1212/wnl.0000000000012341] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To characterize functional network changes related to conversion to cognitive impairment in a large sample of patients with multiple sclerosis (MS) over a period of 5 years. METHODS Two hundred twenty-seven patients with MS and 59 healthy controls of the Amsterdam MS cohort underwent neuropsychological testing and resting-state fMRI at 2 time points (time interval 4.9 ± 0.9 years). At both baseline and follow-up, patients were categorized as cognitively preserved (CP; n = 123), mildly impaired (MCI; z < -1.5 on ≥2 cognitive tests, n = 32), or impaired (CI; z < -2 on ≥2 tests, n = 72), and longitudinal conversion between groups was determined. Network function was quantified with eigenvector centrality, a measure of regional network importance, which was computed for individual resting-state networks at both time points. RESULTS Over time, 18.9% of patients converted to a worse phenotype; 22 of 123 patients who were CP (17.9%) converted from CP to MCI, 10 of 123 from CP to CI (8.1%), and 12 of 32 patients with MCI converted to CI (37.5%). At baseline, default-mode network (DMN) centrality was higher in CI individuals compared to controls (p = 0.05). Longitudinally, ventral attention network (VAN) importance increased in CP, driven by stable CP and CP-to-MCI converters (p < 0.05). CONCLUSIONS Of all patients, 19% worsened in their cognitive status over 5 years. Conversion from intact cognition to impairment is related to an initial disturbed functioning of the VAN, then shifting toward DMN dysfunction in CI. Because the VAN normally relays information to the DMN, these results could indicate that in MS normal processes crucial for maintaining overall network stability are progressively disrupted as patients clinically progress.
Collapse
Affiliation(s)
- Marijn Huiskamp
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK.
| | - Anand J C Eijlers
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Tommy A A Broeders
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Jasmin Pasteuning
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Iris Dekker
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Bernard M J Uitdehaag
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Frederik Barkhof
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Alle-Meije Wink
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Jeroen J G Geurts
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Hanneke E Hulst
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| | - Menno M Schoonheim
- From the Department of Anatomy and Neurosciences (M.H., A.J.C.E., T.A.A.B., J.P., J.J.G.G., H.E.H., M.M.S.), Department of Neurology (I.D., B.M.J.U.), and Department of Radiology and Nuclear Medicine (I.D., F.B., A.-M.W.), MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands; and UCL Institutes of Neurology and Healthcare Engineering (F.B.), London, UK
| |
Collapse
|
105
|
Tagge IJ, Anderson VC, Springer CS, Sammi MK, Bourdette DN, Spain RI, Rooney WD. Gray matter blood-brain barrier water exchange dynamics are reduced in progressive multiple sclerosis. J Neuroimaging 2021; 31:1111-1118. [PMID: 34355458 PMCID: PMC9291753 DOI: 10.1111/jon.12912] [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: 05/26/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose To compare transcapillary wall water exchange, a putative marker of cerebral metabolic health, in brain T2 white matter (WM) lesions and normal appearing white and gray matter (NAWM and NAGM, respectively) in individuals with progressive multiple sclerosis (PMS) and healthy controls (HC). Methods Dynamic‐contrast‐enhanced 7T MRI data were obtained from 19 HC and 23 PMS participants. High‐resolution pharmacokinetic parametric maps representing tissue microvascular and microstructural properties were created by shutter‐speed (SS) paradigm modeling to obtain estimates of blood volume fraction (vb), water molecule capillary efflux rate constant (kpo), and the water capillary wall permeability surface area product (PwS ≡ vb*kpo). Linear regression models were used to investigate differences in (i) kpo and PwS between groups in NAWM and NAGM, and (ii) between WM lesions and NAWM in PMS. Results High‐resolution parametric maps were produced to visualize tissue classes and resolve individual WM lesions. Normal‐appearing gray matter kpo and PwS were significantly decreased in PMS compared to HC (p ≤ .01). Twenty‐one T2 WM lesions were analyzed in 10 participants with PMS. kpo was significantly decreased in WM lesions compared to PMS NAWM (p < .0001). Conclusions Transcapillary water exchange is reduced in PMS NAGM compared to HC and is further reduced in PMS WM lesions, suggesting pathologically impaired brain metabolism. kpo provides a sensitive measure of cerebral metabolic activity and/or coupling, and can be mapped at higher spatial resolution than conventional imaging techniques assessing metabolic activity.
Collapse
Affiliation(s)
- Ian J Tagge
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Valerie C Anderson
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Charles S Springer
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Manoj K Sammi
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Dennis N Bourdette
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Rebecca I Spain
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA.,Neurology Division, Veterans Affairs Portland Health Care System, Portland, Oregon, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA.,Department of Neurology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
106
|
Sidoryk-Węgrzynowicz M, Strużyńska L. Astroglial and Microglial Purinergic P2X7 Receptor as a Major Contributor to Neuroinflammation during the Course of Multiple Sclerosis. Int J Mol Sci 2021; 22:8404. [PMID: 34445109 PMCID: PMC8395107 DOI: 10.3390/ijms22168404] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 02/08/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system that leads to the progressive disability of patients. A characteristic feature of the disease is the presence of focal demyelinating lesions accompanied by an inflammatory reaction. Interactions between autoreactive immune cells and glia cells are considered as a central mechanism underlying the pathology of MS. A glia-mediated inflammatory reaction followed by overproduction of free radicals and generation of glutamate-induced excitotoxicity promotes oligodendrocyte injury, contributing to demyelination and subsequent neurodegeneration. Activation of purinergic signaling, in particular P2X7 receptor-mediated signaling, in astrocytes and microglia is an important causative factor in these pathological processes. This review discusses the role of astroglial and microglial cells, and in particular glial P2X7 receptors, in inducing MS-related neuroinflammatory events, highlighting the importance of P2X7R-mediated molecular pathways in MS pathology and identifying these receptors as a potential therapeutic target.
Collapse
Affiliation(s)
- Marta Sidoryk-Węgrzynowicz
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, 02-106 Warsaw, Poland
| | - Lidia Strużyńska
- Laboratory of Pathoneurochemistry, Department of Neurochemistry, Mossakowski Medical Research Institute, 02-106 Warsaw, Poland
| |
Collapse
|
107
|
Pitteri M, Magliozzi R, Nicholas R, Ziccardi S, Pisani AI, Pezzini F, Marastoni D, Calabrese M. Cerebrospinal fluid inflammatory profile of cognitive impairment in newly diagnosed multiple sclerosis patients. Mult Scler 2021; 28:768-777. [PMID: 34328817 DOI: 10.1177/13524585211032510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The cerebrospinal fluid (CSF) molecular milieu is a marker of diffuse intrathecal inflammation in the meninges that, in turn, targets the grey matter (GM) in multiple sclerosis (MS). Cognitive impairment (CI) is associated with brain damage in MS and is often present early in people with MS (pwMS). OBJECTIVE To investigate whether a specific CSF inflammatory profile is associated with different degrees of CI in newly diagnosed pwMS. METHODS Sixty-nine pwMS and 43 healthy controls (HCs) underwent neuropsychological testing. The presence and levels of 57 inflammatory mediators in the CSF were assessed. RESULTS Apparently cognitively normal (ACN) pwMS had impaired executive functioning compared to HCs but performed better than pwMS with mild and severe CI (mCI and sCI) in all tests. CSF mediators involving innate immunity and immune activation and recruitment, differentiate ACN from pwMS with mCI, while CSF mediators related to B- and T-cell immunity and chemotaxis differentiate both ACN and mCI from those with sCI. CXCL13 was the only molecule that differentiated sCI from mCI pwMS. CONCLUSION Specific CSF molecular patterns, reflecting the involvement of both innate and adaptive immune responses, are associated with the severity of CI in newly diagnosed pwMS.
Collapse
Affiliation(s)
- Marco Pitteri
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Roberta Magliozzi
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy/Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Stefano Ziccardi
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna Isabella Pisani
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Francesco Pezzini
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Damiano Marastoni
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Massimiliano Calabrese
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
108
|
Imaging multiple sclerosis pathology at 160 μm isotropic resolution by human whole-brain ex vivo magnetic resonance imaging at 3 T. Sci Rep 2021; 11:15491. [PMID: 34326420 PMCID: PMC8322069 DOI: 10.1038/s41598-021-94891-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/13/2021] [Indexed: 11/09/2022] Open
Abstract
Postmortem magnetic resonance imaging (MRI) of the fixed healthy and diseased human brain facilitates spatial resolutions and image quality that is not achievable with in vivo MRI scans. Though challenging-and almost exclusively performed at 7 T field strength-depicting the tissue architecture of the entire brain in fine detail is invaluable since it enables the study of neuroanatomy and uncovers important pathological features in neurological disorders. The objectives of the present work were (1) to develop a 3D isotropic ultra-high-resolution imaging approach for human whole-brain ex vivo acquisitions working on a standard clinical 3 T MRI system; and (2) to explore the sensitivity and specificity of this concept for specific pathoanatomical features of multiple sclerosis. The reconstructed images demonstrate unprecedented resolution and soft tissue contrast of the diseased human brain at 3 T, thus allowing visualization of sub-millimetric lesions in the different cortical layers and in the cerebellar cortex, as well as unique cortical lesion characteristics such as the presence of incomplete/complete iron rims, and patterns of iron accumulation. Further details such as the subpial molecular layer, the line of Gennari, and some intrathalamic nuclei are also well distinguishable.
Collapse
|
109
|
MacDonald A, Lu B, Caron M, Caporicci-Dinucci N, Hatrock D, Petrecca K, Bourque G, Stratton JA. Single Cell Transcriptomics of Ependymal Cells Across Age, Region and Species Reveals Cilia-Related and Metal Ion Regulatory Roles as Major Conserved Ependymal Cell Functions. Front Cell Neurosci 2021; 15:703951. [PMID: 34335193 PMCID: PMC8319996 DOI: 10.3389/fncel.2021.703951] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/22/2021] [Indexed: 11/22/2022] Open
Abstract
Ependymal cells are ciliated-epithelial glial cells that develop from radial glia along the surface of the ventricles of the brain and the spinal canal. They play a critical role in cerebrospinal fluid (CSF) homeostasis, brain metabolism, and the clearance of waste from the brain. These cells have been implicated in disease across the lifespan including developmental disorders, cancer, and neurodegenerative disease. Despite this, ependymal cells remain largely understudied. Using single-cell RNA sequencing data extracted from publicly available datasets, we make key findings regarding the remarkable conservation of ependymal cell gene signatures across age, region, and species. Through this unbiased analysis, we have discovered that one of the most overrepresented ependymal cell functions that we observed relates to a critically understudied role in metal ion homeostasis. Our analysis also revealed distinct subtypes and states of ependymal cells across regions and ages of the nervous system. For example, neonatal ependymal cells maintained a gene signature consistent with developmental processes such as determination of left/right symmetry; while adult ventricular ependymal cells, not spinal canal ependymal cells, appeared to express genes involved in regulating cellular transport and inflammation. Together, these findings highlight underappreciated functions of ependymal cells, which will be important to investigate in order to better understand these cells in health and disease.
Collapse
Affiliation(s)
- Adam MacDonald
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Maxime Caron
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Nina Caporicci-Dinucci
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Dale Hatrock
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
110
|
Association between symbol digit modalities test and regional cortex thickness in young adults with relapsing-remitting multiple sclerosis. Clin Neurol Neurosurg 2021; 207:106805. [PMID: 34280674 DOI: 10.1016/j.clineuro.2021.106805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a demyelinating disease of the central nervous system, predominating within young adults. Cognitive disorders are common in MS and have are associated with several Magnetic Resonance Imaging (MRI) markers, especially brain atrophy. Many have found the symbol digit modalities test (SDMT) to be the most sensitive individual cognitive measure relevant to MS. However, the relationship between SDMT and regional brain cortex thickness in young adults with relapsing-remitting multiple sclerosis (YA-RRMS) has been little explored. The purpose of this study was to investigate the association between the SDMT and regional cortex thickness in YA-RRMS by FreeSurfer, which is an automatic brain structure segmentation method. METHOD Twenty-eight YA-RRMS patients (18-35 years old) were enrolled in the present study. Informed consent and information including gender, age, disease duration, number of relapses, annual relapse rate was collected from all patients. Clinical cognitive evaluations (SDMT and auditory verbal learning test (AVLT)) and daily performance: activities of daily living (ADL) were assessed in the present study. MRI scans were performed at the Institute of Neurosurgery of Tiantan Hospital. Twenty-eight matched healthy controls (HC) MRI data were obtained from Tiantan Hospital database. Data on thirty-four points of bilateral cortical structure thickness using statistically defined brain regions-of-interest from FreeSurfer were obtained from all participants. RESULTS Patients with RRMS exhibited extensively thinner cerebellar cortex compared with HC. SDMT scores were significantly correlated with AVLT subentries (IM, immediate memory; DRM, delayed recall memory; LTRM, long-term recognition memory) in YA-RRMS patients (P < 0.05). SDMT was strongly correlated with regional cortex thickness differences of the right temporal pole (r = 0.68) and bilateral parahippocampal areas (right r = 0.62; left r = 0.60), and moderately correlated with regional cortex thickness differences including the left superior temporal and right insula (r = 0.57 and 0.56, respectively) in YA-RRMS patients. CONCLUSION The present study has shown the SDMT is strongly correlated with selected cortex regions including the bilateral parahippocampal area and the right temporal pole which are involved in geometric structures processing.
Collapse
|
111
|
Smith JA, Nicaise AM, Ionescu RB, Hamel R, Peruzzotti-Jametti L, Pluchino S. Stem Cell Therapies for Progressive Multiple Sclerosis. Front Cell Dev Biol 2021; 9:696434. [PMID: 34307372 PMCID: PMC8299560 DOI: 10.3389/fcell.2021.696434] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system characterized by demyelination and axonal degeneration. MS patients typically present with a relapsing-remitting (RR) disease course, manifesting as sporadic attacks of neurological symptoms including ataxia, fatigue, and sensory impairment. While there are several effective disease-modifying therapies able to address the inflammatory relapses associated with RRMS, most patients will inevitably advance to a progressive disease course marked by a gradual and irreversible accrual of disabilities. Therapeutic intervention in progressive MS (PMS) suffers from a lack of well-characterized biological targets and, hence, a dearth of successful drugs. The few medications approved for the treatment of PMS are typically limited in their efficacy to active forms of the disease, have little impact on slowing degeneration, and fail to promote repair. In looking to address these unmet needs, the multifactorial therapeutic benefits of stem cell therapies are particularly compelling. Ostensibly providing neurotrophic support, immunomodulation and cell replacement, stem cell transplantation holds substantial promise in combatting the complex pathology of chronic neuroinflammation. Herein, we explore the current state of preclinical and clinical evidence supporting the use of stem cells in treating PMS and we discuss prospective hurdles impeding their translation into revolutionary regenerative medicines.
Collapse
Affiliation(s)
- Jayden A. Smith
- Cambridge Innovation Technologies Consulting (CITC) Limited, Cambridge, United Kingdom
| | - Alexandra M. Nicaise
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rosana-Bristena Ionescu
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Regan Hamel
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
112
|
Li J, Zhao YM. Magnetic Resonance Imaging and Clinical Features of the Demyelinating Degeneration of White Matter in Young Patients. Int J Gen Med 2021; 14:3177-3186. [PMID: 34262331 PMCID: PMC8274702 DOI: 10.2147/ijgm.s302587] [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: 01/18/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022] Open
Abstract
Objective Magnetic resonance imaging (MRI) of brain white matter demyelination often focuses on demyelinating disease, cerebral small vascular disease diagnosis, and follow-up of cognitive dysfunction for observation. This study explored MRI findings and clinical manifestations of demyelinating degeneration of white matter in young patients. Methods A total of ninety-four patients with white matter degeneration diagnosed with MRI were enrolled in this study from January 2014 to July 2018. These patients were divided into two groups: the demyelinating disease group (n = 43) and the non-demyelinating disease group (n = 51). The imaging findings and clinical manifestations of the two groups were analyzed. Results Compared with the non-demyelinating group, there were more female than male patients in the demyelinating group (P < 0.05). In addition, of the 45 patients with an imaging result of “demyelinating degeneration of white matter and multiple sclerosis,” 39 patients met the diagnosis of multiple sclerosis (86.7%). In comparison, of the 49 patients with an imaging result of “demyelinating degeneration of white matter,” only four patients met the diagnosis for demyelinating disease (8.2%). Conclusion In patients complaining of headaches, dizziness, vertigo, and other symptoms and in the case of an imaging result showing the demyelinating degeneration of white matter alone, the possibility of a clinical diagnosis of a demyelinating disease is minimal.
Collapse
Affiliation(s)
- Jian Li
- Department of Neurology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yi-Ming Zhao
- Center for Clinical Epidemiology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| |
Collapse
|
113
|
Cencioni MT, Mattoscio M, Magliozzi R, Bar-Or A, Muraro PA. B cells in multiple sclerosis - from targeted depletion to immune reconstitution therapies. Nat Rev Neurol 2021; 17:399-414. [PMID: 34075251 DOI: 10.1038/s41582-021-00498-5] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 02/04/2023]
Abstract
Increasing evidence indicates the involvement of B cells in the pathogenesis of multiple sclerosis (MS), but their precise roles are unclear. In this Review, we provide an overview of the development and physiological functions of B cells and the main mechanisms through which B cells are thought to contribute to CNS autoimmunity. In MS, abnormalities of B cell function include pro-inflammatory cytokine production, defective B cell regulatory function and the formation of tertiary lymphoid-like structures in the CNS, which are the likely source of abnormal immunoglobulin production detectable in the cerebrospinal fluid. We also consider the hypothesis that Epstein-Barr virus (EBV) is involved in the B cell overactivation that leads to inflammatory injury to the CNS in MS. We also review the immunological effects - with a focus on the effects on B cell subsets - of several successful therapeutic approaches in MS, including agents that selectively deplete B cells (rituximab, ocrelizumab and ofatumumab), agents that less specifically deplete lymphocytes (alemtuzumab and cladribine) and autologous haematopoietic stem cell transplantation, in which the immune system is unselectively ablated and reconstituted. We consider the insights that these effects on B cell populations provide and their potential to further our understanding and targeting of B cells in MS.
Collapse
Affiliation(s)
- Maria T Cencioni
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Miriam Mattoscio
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Roberta Magliozzi
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.,Department of Neurology, University of Verona, Verona, Italy
| | - Amit Bar-Or
- Center for Neuroinflammation and Experimental Therapeutics and Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paolo A Muraro
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
114
|
Church ME, Ceja G, McGeehan M, Miller MC, Farias P, Sánchez MD, Swain GP, Assenmacher CA, Stopa EG, Vite CH, Bar-Or A, Alvarez JI. Meningeal B Cell Clusters Correlate with Submeningeal Pathology in a Natural Model of Multiple Sclerosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:44-54. [PMID: 34162727 PMCID: PMC8695639 DOI: 10.4049/jimmunol.2000514] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 04/23/2021] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is an idiopathic demyelinating disease in which meningeal inflammation correlates with accelerated disease progression. The study of meningeal inflammation in MS has been limited because of constrained access to MS brain/spinal cord specimens and the lack of experimental models recapitulating progressive MS. Unlike induced models, a spontaneously occurring model would offer a unique opportunity to understand MS immunopathogenesis and provide a compelling framework for translational research. We propose granulomatous meningoencephalomyelitis (GME) as a natural model to study neuropathological aspects of MS. GME is an idiopathic, progressive neuroinflammatory disease of young dogs with a female bias. In the GME cases examined in this study, the meninges displayed focal and disseminated leptomeningeal enhancement on magnetic resonance imaging, which correlated with heavy leptomeningeal lymphocytic infiltration. These leptomeningeal infiltrates resembled tertiary lymphoid organs containing large B cell clusters that included few proliferating Ki67+ cells, plasma cells, follicular dendritic/reticular cells, and germinal center B cell-like cells. These B cell collections were confined in a specialized network of collagen fibers associated with the expression of the lympho-organogenic chemokines CXCL13 and CCL21. Although neuroparenchymal perivascular infiltrates contained B cells, they lacked the immune signature of aggregates in the meningeal compartment. Finally, meningeal B cell accumulation correlated significantly with cortical demyelination reflecting neuropathological similarities to MS. Hence, during chronic neuroinflammation, the meningeal microenvironment sustains B cell accumulation that is accompanied by underlying neuroparenchymal injury, indicating GME as a novel, naturally occurring model to study compartmentalized neuroinflammation and the associated pathology thought to contribute to progressive MS.
Collapse
Affiliation(s)
- Molly E Church
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Guadalupe Ceja
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Megan McGeehan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Miles C Miller
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Priscilla Farias
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Melissa D Sánchez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Gary P Swain
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Edward G Stopa
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI; and
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amit Bar-Or
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jorge I Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA;
| |
Collapse
|
115
|
Shahrampour S, Heholt J, Wang A, Vedaei F, Mohamed FB, Alizadeh M, Wang Z, Zabrecky G, Wintering N, Bazzan AJ, Leist TP, Monti DA, Newberg AB. N-acetyl cysteine administration affects cerebral blood flow as measured by arterial spin labeling MRI in patients with multiple sclerosis. Heliyon 2021; 7:e07615. [PMID: 34377857 PMCID: PMC8327674 DOI: 10.1016/j.heliyon.2021.e07615] [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: 01/18/2021] [Revised: 02/23/2021] [Accepted: 07/14/2021] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND The purpose of this study was to explore if administration of N-acetyl-cysteine (NAC) in patients with multiple sclerosis (MS) resulted in altered cerebral blood flow (CBF) based on Arterial Spin Labeling (ASL) magnetic resonance imaging (MRI). METHODS Twenty-three patients with mild to moderate MS, (17 relapsing remitting and 6 primary progressive) were randomized to either NAC plus standard of care (N = 11), or standard of care only (N = 12). The experimental group received NAC intravenously (50 mg/kg) once per week and orally (500mg 2x/day) the other six days. Patients in both groups were evaluated initially and after 2 months (of receiving the NAC or waitlist control) with ASL MRI to measure CBF. Clinical symptom questionnaires were also completed at both time points. RESULTS The CBF data showed significant differences in several brain regions including the pons, midbrain, left temporal and frontal lobe, left thalamus, right middle frontal lobe and right temporal/hippocampus (p < 0.001) in the MS group after treatment with NAC, when compared to the control group. Self-reported scores related to cognition and attention were also significantly improved in the NAC group as compared to the control group. CONCLUSIONS The results of this study suggest that NAC administration alters resting CBF in MS patients, and this is associated with qualitative improvements in cognition and attention. Given these findings, large scale efficacy studies will be of value to determine the potential clinical impact of NAC over the course of illness in patients with MS, as well as the most effective dosages and differential effects across subpopulations.
Collapse
Affiliation(s)
- Shiva Shahrampour
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Justin Heholt
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew Wang
- Charles E. Schmidt College of Medicine, Marcus Institute of Integrative Health at FAU Medicine, Florida Atlantic University, Boca Raton, FL USA
| | - Faezeh Vedaei
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Feroze B. Mohamed
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mahdi Alizadeh
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ze Wang
- Department of Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George Zabrecky
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nancy Wintering
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anthony J. Bazzan
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Thomas P. Leist
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Daniel A. Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew B. Newberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
116
|
Duan Y, Zhuo Z, Li H, Tian DC, Li Y, Yang L, Gao C, Zhang T, Zhang X, Shi FD, Barkhof F, Liu Y. Brain structural alterations in MOG antibody diseases: a comparative study with AQP4 seropositive NMOSD and MS. J Neurol Neurosurg Psychiatry 2021; 92:709-716. [PMID: 33687975 PMCID: PMC8223649 DOI: 10.1136/jnnp-2020-324826] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/07/2020] [Accepted: 02/01/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Brain structural alterations and their clinical significance of myelin oligodendrocyte glycoprotein antibody disease (MOGAD) have not been determined. METHODS We recruited 35 MOGAD, 38 aquaporin 4 antibody positive neuromyelitis optica spectrum diseases (AQP4+ NMOSD), 37 multiple sclerosis (MS) and 60 healthy controls (HC) who underwent multimodal brain MRI from two centres. Brain lesions, volumes of the whole brain parenchyma, cortical and subcortical grey matter (GM), brainstem, cerebellum and cerebral white matter (WM) and diffusion measures (fractional anisotropy, FA and mean diffusivity, MD) were compared among the groups. Associations between the MRI measurements and the clinical variables were assessed by partial correlations. Logistic regression was performed to differentiate MOGAD from AQP4+ NMOSD and MS. RESULTS In MOGAD, 19 (54%) patients had lesions on MRI, with cortical/juxtacortical (68%) as the most common location. MOGAD and MS showed lower cortical and subcortical GM volumes than HC, while AQP4+ NMOSD only demonstrated a decreased cortical GM volume. MS demonstrated a lower cerebellar volume, a lower FA and an increased MD than MOGAD and HC. The subcortical GM volume was negatively correlated with Expanded Disability Status Scale in MOGAD (R=-0.51; p=0.004). A combination of MRI and clinical measures could achieve an accuracy of 85% and 93% for the classification of MOGAD versus AQP4+ NMOSD and MOGAD versus MS, respectively. CONCLUSION MOGAD demonstrated cortical and subcortical atrophy without severe WM rarefaction. The subcortical GM volume correlated with clinical disability and a combination of MRI and clinical measures could separate MOGAD from AQP4+ NMOSD and MS.
Collapse
Affiliation(s)
- Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Tiantan Image Research Center, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Tiantan Image Research Center, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Haiqing Li
- Department of Radiology, Huashan Hospital Fudan University, Shanghai, China .,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - De-Cai Tian
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital Fudan University, Shanghai, China.,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Liqin Yang
- Department of Radiology, Huashan Hospital Fudan University, Shanghai, China.,Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Chenyang Gao
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tian Zhang
- Department of Radiology, Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, China
| | - Xinghu Zhang
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fu-Dong Shi
- Center for Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Translational Medicine Center, China National Clinical Research Center for Neurological Diseases, Beijing, China.,Department of Neurology and Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London, UK
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China .,Tiantan Image Research Center, China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
117
|
Audouard E, Rousselot L, Folcher M, Cartier N, Piguet F. Optimized Protocol for Subcutaneous Implantation of Encapsulated Cells Device and Evaluation of Biocompatibility. Front Bioeng Biotechnol 2021; 9:620967. [PMID: 34249877 PMCID: PMC8264370 DOI: 10.3389/fbioe.2021.620967] [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: 01/07/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
Improving a drug delivery system is critical to treat central nervous system disorders. Here we studied an innovative approach based on implantation of a wireless-powered cell-based device in mice. This device, coupling biologic material and electronics, is the first of its kind. The advantage of this technology is its ability to control the secretion of a therapeutic molecule and to switch the classical permanent delivery to activation on demand. In diseases with relapsing-remitting phases such as multiple sclerosis, such activation could be selectively achieved in relapsing phases. However, the safety (tolerance to biomaterials and surgical procedure) of such a clinical device needs to be verified. Therefore, the development of tools to assess the biocompatibility of the system in animal models is an essential step. We present the development of this new therapeutic approach, the challenges we encountered during the different steps of its development (such as cell loading in the chamber, surgery protocol for subcutaneous implantation of the device) and the tools we used to evaluate cell viability and biocompatibility of the device.
Collapse
Affiliation(s)
- Emilie Audouard
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Lisa Rousselot
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Marc Folcher
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zürich, Basel, Switzerland.,Institute of Molecular and Clinical Ophthalmology (IOB), Basel, Switzerland
| | - Nathalie Cartier
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| | - Françoise Piguet
- NeuroGenCell, Inserm U 1127, CNRS UMR 7225, ICM, Institut du Cerveau et de la Moelle Épinière, Sorbonne Université, Paris, France
| |
Collapse
|
118
|
Lipp I, Foster C, Stickland R, Sgarlata E, Tallantyre EC, Davidson AE, Robertson NP, Jones DK, Wise RG, Tomassini V. Predictors of training-related improvement in visuomotor performance in patients with multiple sclerosis: A behavioural and MRI study. Mult Scler 2021; 27:1088-1101. [PMID: 32749927 PMCID: PMC8151554 DOI: 10.1177/1352458520943788] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND The development of tailored recovery-oriented strategies in multiple sclerosis requires early identification of an individual's potential for functional recovery. OBJECTIVE To identify predictors of visuomotor performance improvements, a proxy of functional recovery, using a predictive statistical model that combines demographic, clinical and magnetic resonance imaging (MRI) data. METHODS Right-handed multiple sclerosis patients underwent baseline disability assessment and MRI of the brain structure, function and vascular health. They subsequently undertook 4 weeks of right upper limb visuomotor practice. Changes in performance with practice were our outcome measure. We identified predictors of improvement in a training set of patients using lasso regression; we calculated the best performing model in a validation set and applied this model to a test set. RESULTS Patients improved their visuomotor performance with practice. Younger age, better visuomotor abilities, less severe disease burden and concurrent use of preventive treatments predicted improvements. Neuroimaging localised outcome-relevant sensory motor regions, the microstructure and activity of which correlated with performance improvements. CONCLUSION Initial characteristics, including age, disease duration, visuo-spatial abilities, hand dexterity, self-evaluated disease impact and the presence of disease-modifying treatments, can predict functional recovery in individual patients, potentially improving their clinical management and stratification in clinical trials. MRI is a correlate of outcome, potentially supporting individual prognosis.
Collapse
Affiliation(s)
- Ilona Lipp
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK/Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Catherine Foster
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Rachael Stickland
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Eleonora Sgarlata
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Emma C Tallantyre
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Helen Durham Centre for Neuroinflammation, University Hospital of Wales, Cardiff, UK
| | - Alison E Davidson
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Neil P Robertson
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Helen Durham Centre for Neuroinflammation, University Hospital of Wales, Cardiff, UK
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Richard G Wise
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK/Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University ‘G. d’Annunzio’ of Chieti-Pescara, Chieti, Italy
| | - Valentina Tomassini
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK/Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK/Helen Durham Centre for Neuroinflammation, University Hospital of Wales, Cardiff, UK/Institute for Advanced Biomedical Technologies (ITAB), Department of Neurosciences, Imaging and Clinical Sciences, University ‘G. d’Annunzio’ of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
119
|
Piehl F. Current and emerging disease-modulatory therapies and treatment targets for multiple sclerosis. J Intern Med 2021; 289:771-791. [PMID: 33258193 PMCID: PMC8246813 DOI: 10.1111/joim.13215] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/08/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
The treatment of multiple sclerosis (MS), the most common chronic inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS), continues to transform. In recent years, a number of novel and increasingly effective disease-modulatory therapies (DMTs) have been approved, including oral fumarates and selective sphingosine 1-phosphate modulators, as well as cell-depleting therapies such as cladribine, anti-CD20 and anti-CD52 monoclonals. Amongst DMTs in clinical development, inhibitors of Bruton's tyrosine kinase represent an entirely new emerging drug class in MS, with three different drugs entering phase III trials. However, important remaining fields of improvement comprise tracking of long-term benefit-risk with existing DMTs and exploration of novel treatment targets relating to brain inherent disease processes underlying the progressive neurodegenerative aspect of MS, which accumulating evidence suggests start already early in the disease process. The aim here is to review current therapeutic options in relation to an improved understanding of the immunopathogenesis of MS, also highlighting examples where controlled trials have not generated the desired results. An additional aim is to review emerging therapies undergoing clinical development, including agents that interfere with disease processes believed to be important for neurodegeneration or aiming to enhance reparative responses. Notably, early trials now have shown initial evidence of enhanced remyelination both with small molecule compounds and biologicals. Finally, accumulating evidence from clinical trials and post-marketing real-world patient populations, which underscore the importance of early high effective therapy whilst maintaining acceptable tolerability, is discussed.
Collapse
Affiliation(s)
- F. Piehl
- From theDepartment of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
- The Karolinska University Hospital and Academic Specialist CentreStockholm Health ServicesStockholmSweden
| |
Collapse
|
120
|
Murdaca G, Greco M, Borro M, Gangemi S. Hygiene hypothesis and autoimmune diseases: A narrative review of clinical evidences and mechanisms. Autoimmun Rev 2021; 20:102845. [PMID: 33971339 DOI: 10.1016/j.autrev.2021.102845] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022]
Abstract
Since the start of the "modern era", characterized by the increase in urbanization, a progressive attention to hygiene and autoimmune conditions has considerably grown. Although these diseases are often multifactorial, it was demonstrated that environment factors such as pollution, diet and lifestyles may play a crucial role together with genetic signature. Our research, based on the newest and most significant literature of this topic, highlights that the progressive depletion of microbes and parasites due to increased socioeconomic improvement, may lead to a derangement of immunoregulatory mechanisms. Moreover, special attention was given to the complex interplay between microbial agents, as gut microbiome, diet and vitamin D supplementation with the aim of identifying promising future therapeutic options. In conclusion, autoimmunity cannot be limited to hygiene-hypothesis, but from the point of view of precision medicine, this theory represents a fundamental element together with the study of genomics, the microbiome and proteomics, in order to understand the complex functioning of the immune system.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Monica Greco
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Matteo Borro
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
121
|
Zhang Y, Li D, Zeng Q, Feng J, Fu H, Luo Z, Xiao B, Yang H, Wu M. LRRC4 functions as a neuron-protective role in experimental autoimmune encephalomyelitis. Mol Med 2021; 27:44. [PMID: 33932995 PMCID: PMC8088686 DOI: 10.1186/s10020-021-00304-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Leucine rich repeat containing 4 (LRRC4), also known as netrin-G ligand-2 (NGL-2), belongs to the superfamily of LRR proteins and serves as a receptor for netrin-G2. LRRC4 regulates the formation of excitatory synapses and promotes axon differentiation. Mutations in LRRC4 occur in Autism Spectrum Disorder (ASD) and intellectual disability. Multiple sclerosis (MS) is a chronic neuroinflammatory disease with spinal cords demyelination and neurodegeneration. Here, we sought to investigate whether LRRC4 is involved in spinal cords neuron-associated diseases. METHODS LRRC4 was detected in the CNS of experimental autoimmune encephalomyelitis (EAE) mice by the use of real-time PCR and western blotting. LRRC4-/- mice were created and immunized with myelin oligodendrocyte glycoprotein peptide (MOG)35-55. Pathological changes in spinal cords of LRRC4-/- and WT mice 15 days after immunization were examined by using hematoxylin and eosin (H&E), Luxol Fast Blue (LFB) staining and immunohistochemistry. The number of Th1/Th2/Th17/Treg cells in spleens and blood were measured with flow cytometry. Differential gene expression in the spinal cords from WT and LRRC4-/- mice was analyzed by using RNA sequencing (RNA-seq). Adeno-associated virus (AAV) vectors were used to overexpress LRRC4 (AAV-LRRC4) and were injected into EAE mice to assess the therapeutic effect of AAV-LRRC4 ectopic expression on EAE. RESULTS We report that LRRC4 is mainly expressed in neuron of spinal cords, and is decreased in the spinal cords of the EAE mice. Knockout of LRRC4 have a disease progression quickened and exacerbated with more severe myelin degeneration and infiltration of leukocytes into the spinal cords. We also first found that Rab7b is high expressed in EAE mice, and the deficiency of LRRC4 induces the elevated NF-κB p65 by up-regulating Rab7b, and up-regulation of IL-6, IFN-γ and down-regulation of TNF-α, results in more severe Th1 immune response in LRRC4-/- mice. Ectopic expression of LRRC4 alleviates the clinical symptoms of EAE mice and protects the neurons from immune damages. CONCLUSIONS We identified a neuroprotective role of LRRC4 in the progression of EAE, which may be used as a potential target for auxiliary support therapeutic treatment of MS.
Collapse
Affiliation(s)
- Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
- Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Di Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Qiuming Zeng
- Internal Medicine-Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Haijuan Fu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Zhaohui Luo
- Internal Medicine-Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Bo Xiao
- Internal Medicine-Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huan Yang
- Internal Medicine-Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
122
|
Collorone S, Prados F, Kanber B, Cawley NM, Tur C, Grussu F, Solanky BS, Yiannakas M, Davagnanam I, Wheeler-Kingshott CAMG, Barkhof F, Ciccarelli O, Toosy AT. Brain microstructural and metabolic alterations detected in vivo at onset of the first demyelinating event. Brain 2021; 144:1409-1421. [PMID: 33903905 PMCID: PMC8219367 DOI: 10.1093/brain/awab043] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/03/2020] [Accepted: 12/03/2020] [Indexed: 12/22/2022] Open
Abstract
In early multiple sclerosis, a clearer understanding of normal-brain tissue microstructural and metabolic abnormalities will provide valuable insights into its pathophysiology. We used multi-parametric quantitative MRI to detect alterations in brain tissues of patients with their first demyelinating episode. We acquired neurite orientation dispersion and density imaging [to investigate morphology of neurites (dendrites and axons)] and 23Na MRI (to estimate total sodium concentration, a reflection of underlying changes in metabolic function). In this cross-sectional study, we enrolled 42 patients diagnosed with clinically isolated syndrome or multiple sclerosis within 3 months of their first demyelinating event and 16 healthy controls. Physical and cognitive scales were assessed. At 3 T, we acquired brain and spinal cord structural scans, and neurite orientation dispersion and density imaging. Thirty-two patients and 13 healthy controls also underwent brain 23Na MRI. We measured neurite density and orientation dispersion indices and total sodium concentration in brain normal-appearing white matter, white matter lesions, and grey matter. We used linear regression models (adjusting for brain parenchymal fraction and lesion load) and Spearman correlation tests (significance level P ≤ 0.01). Patients showed higher orientation dispersion index in normal-appearing white matter, including the corpus callosum, where they also showed lower neurite density index and higher total sodium concentration, compared with healthy controls. In grey matter, compared with healthy controls, patients demonstrated: lower orientation dispersion index in frontal, parietal and temporal cortices; lower neurite density index in parietal, temporal and occipital cortices; and higher total sodium concentration in limbic and frontal cortices. Brain volumes did not differ between patients and controls. In patients, higher orientation dispersion index in corpus callosum was associated with worse performance on timed walk test (P = 0.009, B = 0.01, 99% confidence interval = 0.0001 to 0.02), independent of brain and lesion volumes. Higher total sodium concentration in left frontal middle gyrus was associated with higher disability on Expanded Disability Status Scale (rs = 0.5, P = 0.005). Increased axonal dispersion was found in normal-appearing white matter, particularly corpus callosum, where there was also axonal degeneration and total sodium accumulation. The association between increased axonal dispersion in the corpus callosum and worse walking performance implies that morphological and metabolic alterations in this structure could mechanistically contribute to disability in multiple sclerosis. As brain volumes were neither altered nor related to disability in patients, our findings suggest that these two advanced MRI techniques are more sensitive at detecting clinically relevant pathology in early multiple sclerosis.
Collapse
Affiliation(s)
- Sara Collorone
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Ferran Prados
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Universitat Oberta de Catalunya, Barcelona, Spain
| | - Baris Kanber
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Niamh M Cawley
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Carmen Tur
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Francesco Grussu
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Centre for Medical Image Computing (CMIC), Department of Computer Sciences, University College London, London, UK
| | - Bhavana S Solanky
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Marios Yiannakas
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| | - Indran Davagnanam
- Department of Brain Repair and Rehabilitation, University College London Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| | - Claudia A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Brain MRI 3T Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Frederik Barkhof
- Centre for Medical Image Computing (CMIC), Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Department of Brain Repair and Rehabilitation, University College London Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK.,Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Vrije Universiteit, The Netherlands.,National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
| | - Olga Ciccarelli
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK.,National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
| | - Ahmed T Toosy
- NMR Research Unit, Queen Square MS Centre, Department of Neuroinflammation, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, UK
| |
Collapse
|
123
|
Insights into the Pathophysiology of Psychiatric Symptoms in Central Nervous System Disorders: Implications for Early and Differential Diagnosis. Int J Mol Sci 2021; 22:ijms22094440. [PMID: 33922780 PMCID: PMC8123079 DOI: 10.3390/ijms22094440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Different psychopathological manifestations, such as affective, psychotic, obsessive-compulsive symptoms, and impulse control disturbances, may occur in most central nervous system (CNS) disorders including neurodegenerative and neuroinflammatory diseases. Psychiatric symptoms often represent the clinical onset of such disorders, thus potentially leading to misdiagnosis, delay in treatment, and a worse outcome. In this review, psychiatric symptoms observed along the course of several neurological diseases, namely Alzheimer’s disease, fronto-temporal dementia, Parkinson’s disease, Huntington’s disease, and multiple sclerosis, are discussed, as well as the involved brain circuits and molecular/synaptic alterations. Special attention has been paid to the emerging role of fluid biomarkers in early detection of these neurodegenerative diseases. The frequent occurrence of psychiatric symptoms in neurological diseases, even as the first clinical manifestations, should prompt neurologists and psychiatrists to share a common clinico-biological background and a coordinated diagnostic approach.
Collapse
|
124
|
Pardini M, Brown JWL, Magliozzi R, Reynolds R, Chard DT. Surface-in pathology in multiple sclerosis: a new view on pathogenesis? Brain 2021; 144:1646-1654. [PMID: 33876200 DOI: 10.1093/brain/awab025] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/03/2020] [Accepted: 11/17/2020] [Indexed: 11/12/2022] Open
Abstract
While multiple sclerosis can affect any part of the CNS, it does not do so evenly. In white matter it has long been recognized that lesions tend to occur around the ventricles, and grey matter lesions mainly accrue in the outermost (subpial) cortex. In cortical grey matter, neuronal loss is greater in the outermost layers. This cortical gradient has been replicated in vivo with magnetization transfer ratio and similar gradients in grey and white matter magnetization transfer ratio are seen around the ventricles, with the most severe abnormalities abutting the ventricular surface. The cause of these gradients remains uncertain, though soluble factors released from meningeal inflammation into the CSF has the most supporting evidence. In this Update, we review this 'surface-in' spatial distribution of multiple sclerosis abnormalities and consider the implications for understanding pathogenic mechanisms and treatments designed to slow or stop them.
Collapse
Affiliation(s)
- Matteo Pardini
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, and IRCCS AOU San Martino-IST, Genoa, Italy
| | - J William L Brown
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,Department of Clinical Neurosciences, University of Cambridge, Box 165, Cambridge Biomedical Campus, Cambridge, UK.,Clinical Outcomes Research Unit (CORe), University of Melbourne, Melbourne, Australia
| | - Roberta Magliozzi
- Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy.,Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Declan T Chard
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, University College London (UCL) Institute of Neurology, London, UK.,National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre, UK
| |
Collapse
|
125
|
Meca-Lallana V, Berenguer-Ruiz L, Carreres-Polo J, Eichau-Madueño S, Ferrer-Lozano J, Forero L, Higueras Y, Téllez Lara N, Vidal-Jordana A, Pérez-Miralles FC. Deciphering Multiple Sclerosis Progression. Front Neurol 2021; 12:608491. [PMID: 33897583 PMCID: PMC8058428 DOI: 10.3389/fneur.2021.608491] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is primarily an inflammatory and degenerative disease of the central nervous system, triggered by unknown environmental factors in patients with predisposing genetic risk profiles. The prevention of neurological disability is one of the essential goals to be achieved in a patient with MS. However, the pathogenic mechanisms driving the progressive phase of the disease remain unknown. It was described that the pathophysiological mechanisms associated with disease progression are present from disease onset. In daily practice, there is a lack of clinical, radiological, or biological markers that favor an early detection of the disease's progression. Different definitions of disability progression were used in clinical trials. According to the most descriptive, progression was defined as a minimum increase in the Expanded Disability Status Scale (EDSS) of 1.5, 1.0, or 0.5 from a baseline level of 0, 1.0–5.0, and 5.5, respectively. Nevertheless, the EDSS is not the most sensitive scale to assess progression, and there is no consensus regarding any specific diagnostic criteria for disability progression. This review document discusses the current pathophysiological concepts associated with MS progression, the different measurement strategies, the biomarkers associated with disability progression, and the available pharmacologic therapeutic approaches.
Collapse
Affiliation(s)
- Virginia Meca-Lallana
- Multiple Sclerosis Unit, Neurology Department, Fundación de Investigación Biomédica, Hospital Universitario de la Princesa, Madrid, Spain
| | | | - Joan Carreres-Polo
- Neuroradiology Section, Radiology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Sara Eichau-Madueño
- Multiple Sclerosis CSUR Unit, Neurology Department, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Jaime Ferrer-Lozano
- Department of Pathology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Lucía Forero
- Neurology Department, Hospital Puerta del Mar, Cádiz, Spain
| | - Yolanda Higueras
- Neurology Department, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Hospital Universitario Gregorio Marañón, Madrid, Spain.,Department of Experimental Psychology, Cognitive Processes and Speech Therapy, Universidad Complutense, Madrid, Spain
| | - Nieves Téllez Lara
- Neurology Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Angela Vidal-Jordana
- Neurology/Neuroimmunology Department, Centre d'Esclerosi Múltiple de Catalunya (Cemcat), Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Francisco Carlos Pérez-Miralles
- Neuroimmunology Unit, Neurology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain.,Department of Medicine, University of València, Valencia, Spain
| |
Collapse
|
126
|
Fujimori J, Fujihara K, Wattjes M, Nakashima I. Patterns of cortical grey matter thickness reduction in multiple sclerosis. Brain Behav 2021; 11:e02050. [PMID: 33506628 PMCID: PMC8035454 DOI: 10.1002/brb3.2050] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/07/2021] [Accepted: 01/17/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE To examine the patterns of cortical gray matter thickness in multiple sclerosis (MS) patients. METHODS Seventy-four MS patients-clinically isolated syndrome (4%), relapsing-remitting MS (79%), and progressive MS (17%)-and 21 healthy controls (HCs) underwent 1.5 Tesla T1-weighted 3D MRI examinations to measure brain cortical thickness in a total of 68 regions of interest. Using hierarchical cluster analysis with multivariate cortical thickness data, cortical thickness reduction patterns were cross-sectionally investigated in MS patients. RESULTS The MS patients were grouped into three major clusters (Clusters 1, 2, and 3). Most of the regional cortical thickness values were equivalent between the HCs and Cluster 1, but decreased in the order of Clusters 2 and 3. Only the thicknesses of the temporal lobe cortices (the bilateral superior and left middle temporal cortex, as well as the left fusiform cortex) were significantly different among Clusters 1, 2, and 3. In contrast, temporal pole thickness reduction was evident exclusively in Cluster 3, which was also characterized by increased lesion loads in the temporal pole and the adjacent juxtacortical white matter, dilatation of the inferior horn of the lateral ventricle, severe whole-brain volume reduction, and longer disease duration. Although cortical atrophy was significantly more common in the progressive phase, approximately half of the MS patients with the severe cortical atrophy pattern had relapsing-remitting disease. CONCLUSION Cortical thickness reduction patterns in MS are mostly characterized by the degree of temporal lobe cortical atrophy, which may start in the relapsing-remitting phase. Among the temporal lobe cortices, the neurodegenerative change may accelerate in the temporal pole in the progressive phase.
Collapse
Affiliation(s)
- Juichi Fujimori
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Multiple Sclerosis Therapeutics, Fukushima Medical University School of Medicine and Multiple Sclerosis and Neuromyelitis Optica Center, Southern Tohoku Research Institute for Neuroscience, Koriyama, Japan
| | - Mike Wattjes
- Department of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Ichiro Nakashima
- Division of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
127
|
De Meo E, Portaccio E, Giorgio A, Ruano L, Goretti B, Niccolai C, Patti F, Chisari CG, Gallo P, Grossi P, Ghezzi A, Roscio M, Mattioli F, Stampatori C, Simone M, Viterbo RG, Bonacchi R, Rocca MA, De Stefano N, Filippi M, Amato MP. Identifying the Distinct Cognitive Phenotypes in Multiple Sclerosis. JAMA Neurol 2021; 78:414-425. [PMID: 33393981 DOI: 10.1001/jamaneurol.2020.4920] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Cognitive impairment is a common and disabling feature of multiple sclerosis (MS), but a precise characterization of cognitive phenotypes in patients with MS is lacking. Objectives To identify cognitive phenotypes in a clinical cohort of patients with MS and to characterize their clinical and magnetic resonance imaging (MRI) features. Design, Setting, and Participants This multicenter cross-sectional study consecutively screened clinically stable patients with MS and healthy control individuals at 8 MS centers in Italy from January 1, 2010, to October 31, 2019. Patients with MS and healthy control individuals who were not using psychoactive drugs and had no history of other neurological or medical disorders, learning disability, severe head trauma, and alcohol or drug abuse were enrolled. Main Outcomes and Measures Participants underwent a neurological examination and a cognitive evaluation with the Rao Brief Repeatable Battery and Stroop Color and Word Test. A subgroup of participants also underwent a brain MRI examination. Latent profile analysis was used on cognitive test z scores to identify cognitive phenotypes. Linear regression and mixed-effects models were used to define clinical and MRI features of each phenotype. Results A total of 1212 patients with MS (mean [SD] age, 41.1 [11.1] years; 784 women [64.7%]) and 196 healthy control individuals (mean [SD] age, 40.4 [8.6] years; 130 women [66.3%]) were analyzed in this study. Five cognitive phenotypes were identified: preserved cognition (n = 235 patients [19.4%]), mild-verbal memory/semantic fluency (n = 362 patients [29.9%]), mild-multidomain (n = 236 patients [19.5%]), severe-executive/attention (n = 167 patients [13.8%]), and severe-multidomain (n = 212 patients [17.5%]) involvement. Patients with preserved cognition and mild-verbal memory/semantic fluency were younger (mean [SD] age, 36.5 [9.8] years and 38.2 [11.1] years) and had shorter disease duration (mean [SD] 8.0 [7.3] years and 8.3 [7.6] years) compared with patients with mild-multidomain (mean [SD] age, 42.6 [11.2] years; mean [SD] disease duration, 12.8 [9.6] years; P < .001), severe-executive/attention (mean [SD] age, 42.9 [11.7] years; mean [SD] disease duration, 12.2 [9.5] years; P < .001), and severe-multidomain (mean [SD] age, 44.0 [11.0] years; mean [SD] disease duration, 13.3 [10.2] years; P < .001) phenotypes. Severe cognitive phenotypes prevailed in patients with progressive MS. At MRI evaluation, compared with those with preserved cognition, patients with mild-verbal memory/semantic fluency exhibited decreased mean (SE) hippocampal volume (5.42 [0.68] mL vs 5.13 [0.68] mL; P = .04), patients with the mild-multidomain phenotype had decreased mean (SE) cortical gray matter volume (687.69 [35.40] mL vs 662.59 [35.48] mL; P = .02), patients with severe-executive/attention had higher mean (SE) T2-hyperintense lesion volume (51.33 [31.15] mL vs 99.69 [34.07] mL; P = .04), and patients with the severe-multidomain phenotype had extensive brain damage, with decreased volume in all the brain structures explored, except for nucleus pallidus, amygdala and caudate nucleus. Conclusions and Relevance This study found that by defining homogeneous and clinically meaningful phenotypes, the limitations of the traditional dichotomous classification in MS can be overcome. These phenotypes can represent a more meaningful measure of the cognitive status of patients with MS and can help define clinical disability, support clinicians in treatment choices, and tailor cognitive rehabilitation strategies.
Collapse
Affiliation(s)
- Ermelinda De Meo
- Neuroimaging Research Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Institute of Experimental Neurology, Vita-Salute San Raffaele University, Milan, Italy.,Section Neurosciences, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, University of Florence, Florence, Italy
| | - Emilio Portaccio
- Department of Neurology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Department of Neurorehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Antonio Giorgio
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Luis Ruano
- EPIUnit, Instituto de Saúde Pública de Universidade do Porto, Porto, Portugal.,Neurology Department, Centro Hospitalar de Entre Douro e Vouga, Santa Maria da Feira, Portugal
| | - Benedetta Goretti
- Section Neurosciences, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, University of Florence, Florence, Italy
| | - Claudia Niccolai
- Department of Neurorehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Francesco Patti
- Department of Neurology, University of Catania, Catania, Italy
| | | | - Paolo Gallo
- Department of Neurology, University of Padova, Padova, Italy
| | - Paola Grossi
- Neuroimmunology Center, Cardiocerebrovascular, Azienda Socio Sanitaria Territoriale (ASST) of Crema, Crema, Italy
| | | | | | - Flavia Mattioli
- Neuropsychology Unit, ASST Spedali Civili Brescia, Brescia, Italy
| | | | - Marta Simone
- Child and Adolescence Neuropsychiatry Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs University Aldo Moro Bari, Bari, Italy
| | - Rosa Gemma Viterbo
- Child and Adolescence Neuropsychiatry Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs University Aldo Moro Bari, Bari, Italy
| | - Raffaello Bonacchi
- Neuroimaging Research Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Institute of Experimental Neurology, Vita-Salute San Raffaele University, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy.,Institute of Experimental Neurology, Vita-Salute San Raffaele University, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Amato
- Section Neurosciences, Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, University of Florence, Florence, Italy.,Department of Neurorehabilitation, IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
128
|
Picon C, Jayaraman A, James R, Beck C, Gallego P, Witte ME, van Horssen J, Mazarakis ND, Reynolds R. Neuron-specific activation of necroptosis signaling in multiple sclerosis cortical grey matter. Acta Neuropathol 2021; 141:585-604. [PMID: 33569629 PMCID: PMC7952371 DOI: 10.1007/s00401-021-02274-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/06/2021] [Accepted: 01/21/2021] [Indexed: 01/01/2023]
Abstract
Sustained exposure to pro-inflammatory cytokines in the leptomeninges is thought to play a major role in the pathogenetic mechanisms leading to cortical pathology in multiple sclerosis (MS). Although the molecular mechanisms underlying neurodegeneration in the grey matter remain unclear, several lines of evidence suggest a prominent role for tumour necrosis factor (TNF). Using cortical grey matter tissue blocks from post-mortem brains from 28 secondary progressive MS subjects and ten non-neurological controls, we describe an increase in expression of multiple steps in the TNF/TNF receptor 1 signaling pathway leading to necroptosis, including the key proteins TNFR1, FADD, RIPK1, RIPK3 and MLKL. Activation of this pathway was indicated by the phosphorylation of RIPK3 and MLKL and the formation of protein oligomers characteristic of necrosomes. In contrast, caspase-8 dependent apoptotic signaling was decreased. Upregulation of necroptotic signaling occurred predominantly in macroneurons in cortical layers II–III, with little expression in other cell types. The presence of activated necroptotic proteins in neurons was increased in MS cases with prominent meningeal inflammation, with a 30-fold increase in phosphoMLKL+ neurons in layers I–III. The density of phosphoMLKL+ neurons correlated inversely with age at death, age at progression and disease duration. In vivo induction of chronically elevated TNF and INFγ levels in the CSF in a rat model via lentiviral transduction in the meninges, triggered inflammation and neurodegeneration in the underlying cortical grey matter that was associated with increased neuronal expression of TNFR1 and activated necroptotic signaling proteins. Exposure of cultured primary rat cortical neurons to TNF induced necroptosis when apoptosis was inhibited. Our data suggest that neurons in the MS cortex are dying via TNF/TNFR1 stimulated necroptosis rather than apoptosis, possibly initiated in part by chronic meningeal inflammation. Neuronal necroptosis represents a pathogenetic mechanism that is amenable to therapeutic intervention at several points in the signaling pathway.
Collapse
|
129
|
Preziosa P, Storelli L, Meani A, Moiola L, Rodegher M, Filippi M, Rocca MA. Effects of Fingolimod and Natalizumab on Brain T1-/T2-Weighted and Magnetization Transfer Ratios: a 2-Year Study. Neurotherapeutics 2021; 18:878-888. [PMID: 33483938 PMCID: PMC8423925 DOI: 10.1007/s13311-020-00997-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 11/26/2022] Open
Abstract
Fingolimod and natalizumab significantly reduce disease activity in relapsing-remitting multiple sclerosis (RRMS) and could promote tissue repair and neuroprotection. The ratio between conventional T1- and T2-weighted sequences (T1w/T2w-ratio) and magnetization transfer ratio (MTR) allow to quantify brain microstructural tissue abnormalities. Here, we compared fingolimod and natalizumab effects on brain T1w/T2w-ratio and MTR in RRMS over 2 years of treatment. RRMS patients starting fingolimod (n = 25) or natalizumab (n = 30) underwent 3T brain MRI scans at baseline (T0), month 6 (M6), month 12 (M12), and month 24 (M24). White matter (WM) lesions, normal-appearing (NA) WM, and gray matter (GM) T1w/T2w-ratio and MTR were estimated and compared between groups using linear mixed models. No baseline demographic, clinical, and MRI difference was found between groups. In natalizumab patients, lesion T1w/T2w-ratio and MTR significantly increased at M6 vs. T0 (p ≤ 0.035) and decreased at subsequent timepoints (p ≤ 0.037). In fingolimod patients, lesion T1w/T2w-ratio increased at M12 vs. T0 (p = 0.010), while MTR gradually increased at subsequent timepoints vs. T0 (p ≤ 0.027). Natalizumab stabilized NAWM and GM T1w/T2w-ratio and MTR. In fingolimod patients, NAWM T1w/T2w-ratio and MTR significantly increased at M24 vs. M12 (p ≤ 0.001). A significant GM T1w/T2w-ratio decrease at M6 vs. T0 (p = 0.014) and increase at M24 vs. M6 (p = 0.008) occurred, whereas GM MTR was significantly higher at M24 vs. previous timepoints (p ≤ 0.017) with significant between-group differences (p ≤ 0.034). Natalizumab may promote an early recovery of lesional damage and prevent microstructural damage accumulation in NAWM and GM during the first 2 years of treatment. Fingolimod enhances tissue damage recovery being visible after 6 months in lesions and after 2 years in NAWM and GM.
Collapse
Affiliation(s)
- Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Meani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Via Olgettina, 60, 20132, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - 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, Via Olgettina, 60, 20132, Milan, Italy.
| |
Collapse
|
130
|
Ramasamy R, Smith PP. Animal modeling of lower urinary tract dysfunction associated with multiple sclerosis: Part I: Justification of the mouse model for MS research. Neurourol Urodyn 2021; 40:950-957. [PMID: 33719097 DOI: 10.1002/nau.24649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 01/16/2021] [Indexed: 12/20/2022]
Abstract
Lower urinary tract symptoms and dysfunction (LUTS/LUTD) contribute to loss of quality of life, morbidity, and need for medical intervention in most patients with multiple sclerosis (MS). Although MS is an inflammatory neurodegenerative disease, clinical manifestations including continence control disorders have traditionally been attributed to the loss of neural signaling due to neurodegeneration. Clinical approaches to MS-LUTS/LUTD have focused on addressing symptoms in the context of urodynamic dysfunctions as pathophysiologic understandings are incomplete. The mouse model provides a useful research platform for the discovery of more detailed molecular, cellular, and tissue-level knowledge of the disease and its clinical manifestations. The aim of this two-part review is to provide a state-of-the-art update on the use of the mouse model for MS research, with a focus on lower urinary tract symptoms. Part I presents a summary of the current understanding of MS pathophysiology, the impact on lower urinary tract symptoms, and briefly introduces the types of mouse models available to study MS. Part II presents the common animal models that are currently available to study MS, their mechanism, relevance to MS-LUTS/LUTD and their urinary pathophysiology, advantages, and disadvantages.
Collapse
Affiliation(s)
- Ramalakshmi Ramasamy
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA.,Department of Neuroscience, University of Connecticut Graduate School, Farmington, Connecticut, USA.,CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA
| | - Phillip P Smith
- UConn Center on Aging, UConn Health, Farmington, Connecticut, USA.,Department of Neuroscience, University of Connecticut Graduate School, Farmington, Connecticut, USA.,CT Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, Connecticut, USA.,Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
131
|
Halder SK, Milner R. Hypoxia in multiple sclerosis; is it the chicken or the egg? Brain 2021; 144:402-410. [PMID: 33351069 PMCID: PMC8453297 DOI: 10.1093/brain/awaa427] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/03/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Over the past 50 years, intense research effort has taught us a great deal about multiple sclerosis. We know that it is the most common neurological disease affecting the young-middle aged, that it affects two to three times more females than males, and that it is characterized as an autoimmune disease, in which autoreactive T lymphocytes cross the blood-brain barrier, resulting in demyelinating lesions. But despite all the knowledge gained, a key question still remains; what is the initial event that triggers the inflammatory demyelinating process? While most research effort to date has focused on the immune system, more recently, another potential candidate has emerged: hypoxia. Specifically, a growing number of studies have described the presence of hypoxia (both 'virtual' and real) at an early stage of demyelinating lesions, and several groups, including our own, have begun to investigate how manipulation of inspired oxygen levels impacts disease progression. In this review we summarize the findings of these hypoxia studies, and in particular, address three main questions: (i) is the hypoxia found in demyelinating lesions 'virtual' or real; (ii) what causes this hypoxia; and (iii) how does manipulation of inspired oxygen impact disease progression?
Collapse
Affiliation(s)
- Sebok K Halder
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA 92121, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, 10865 Road to the Cure, Suite 100, San Diego, CA 92121, USA
| |
Collapse
|
132
|
Schirmer L, Schafer DP, Bartels T, Rowitch DH, Calabresi PA. Diversity and Function of Glial Cell Types in Multiple Sclerosis. Trends Immunol 2021; 42:228-247. [PMID: 33593693 PMCID: PMC7914214 DOI: 10.1016/j.it.2021.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 12/25/2022]
Abstract
Glial subtype diversity is an emerging topic in neurobiology and immune-mediated neurological diseases such as multiple sclerosis (MS). We discuss recent conceptual and technological advances that allow a better understanding of the transcriptomic and functional heterogeneity of oligodendrocytes (OLs), astrocytes, and microglial cells under inflammatory-demyelinating conditions. Recent single cell transcriptomic studies suggest the occurrence of novel homeostatic and reactive glial subtypes and provide insight into the molecular events during disease progression. Multiplexed RNA in situ hybridization has enabled 'mapping back' dysregulated gene expression to glial subtypes within the MS lesion microenvironment. These findings suggest novel homeostatic and reactive glial-cell-type functions both in immune-related processes and neuroprotection relevant to understanding the pathology of MS.
Collapse
Affiliation(s)
- Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| | - Dorothy P Schafer
- Department of Neurobiology and the Brudnik Neuropsychiatric Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Theresa Bartels
- Department of Paediatrics, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - David H Rowitch
- Department of Paediatrics, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Departments of Pediatrics and Neurosurgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
133
|
Silva BA, Miglietta E, Ferrari CC. Insights into the role of B cells in the cortical pathology of Multiple sclerosis: evidence from animal models and patients. Mult Scler Relat Disord 2021; 50:102845. [PMID: 33636613 DOI: 10.1016/j.msard.2021.102845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 02/03/2021] [Accepted: 02/13/2021] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system (CNS) that affects both white and gray matter. Although it has been traditionally considered as a T cell mediated disease, the role of B cell in MS pathology has become a topic of great research interest. Cortical lesions, key feature of the progressive forms of MS, are involved in cognitive impairment and worsening of the patients' outcome. These lesions present pathognomonic hallmarks, such as: absence of blood-brain barrier (BBB) disruption, limited inflammatory events, reactive microglia, neurodegeneration, demyelination and meningeal inflammation. B cells located in the meninges, either as part of diffuse inflammation or as part of follicle-like structures, are strongly associated with cortical damage. The function of CD20-expressing B cells in MS is further highlighted by the success of specific therapies using anti-CD20 antibodies. The possible roles of B cells in pathology go beyond their ability to produce antibodies, as they also present antigens to T cells, secrete cytokines (both pathogenic and protective) within the CNS to modulate T and myeloid cell functions, and are involved in meningeal inflammation. Here, we will review the contributions of B cells to the pathogenesis of meningeal inflammation and cortical lesions in MS patients as well as in preclinical animal models.
Collapse
Affiliation(s)
- Berenice Anabel Silva
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina; Centro Universitario de Esclerosis Múltiple, División Neurología, Hospital JM Ramos Mejía, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Esteban Miglietta
- Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina
| | - Carina Cintia Ferrari
- Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET, Buenos Aires, Argentina; Leloir Institute Foundation, Institute for Biochemical Investigations, IIBBA, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
134
|
Zrzavy T, Schwaiger C, Wimmer I, Berger T, Bauer J, Butovsky O, Schwab JM, Lassmann H, Höftberger R. Acute and non-resolving inflammation associate with oxidative injury after human spinal cord injury. Brain 2021; 144:144-161. [PMID: 33578421 PMCID: PMC7880675 DOI: 10.1093/brain/awaa360] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/08/2020] [Accepted: 08/11/2020] [Indexed: 12/25/2022] Open
Abstract
Traumatic spinal cord injury is a devastating insult followed by progressive cord atrophy and neurodegeneration. Dysregulated or non-resolving inflammatory processes can disturb neuronal homeostasis and drive neurodegeneration. Here, we provide an in-depth characterization of innate and adaptive inflammatory responses as well as oxidative tissue injury in human traumatic spinal cord injury lesions compared to non-traumatic control cords. In the lesion core, microglia were rapidly lost while intermediate (co-expressing pro- as well as anti-inflammatory molecules) blood-borne macrophages dominated. In contrast, in the surrounding rim, TMEM119+ microglia numbers were maintained through local proliferation and demonstrated a predominantly pro-inflammatory phenotype. Lymphocyte numbers were low and mainly consisted of CD8+ T cells. Only in a subpopulation of patients, CD138+/IgG+ plasma cells were detected, which could serve as candidate cellular sources for a developing humoral immunity. Oxidative neuronal cell body and axonal injury was visualized by intracellular accumulation of amyloid precursor protein (APP) and oxidized phospholipids (e06) and occurred early within the lesion core and declined over time. In contrast, within the surrounding rim, pronounced APP+/e06+ axon-dendritic injury of neurons was detected, which remained significantly elevated up to months/years, thus providing mechanistic evidence for ongoing neuronal damage long after initial trauma. Dynamic and sustained neurotoxicity after human spinal cord injury might be a substantial contributor to (i) an impaired response to rehabilitation; (ii) overall failure of recovery; or (iii) late loss of recovered function (neuro-worsening/degeneration).
Collapse
Affiliation(s)
- Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Carmen Schwaiger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Isabella Wimmer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Jan Bauer
- Center for Brain Research, Medical University of Vienna, Austria
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Womeńs Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jan M Schwab
- The Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH 43210, USA
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
- Department of Physical Medicine & Rehabilitation, The Ohio State University, Columbus, OH 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
135
|
Bagnato F, Gauthier SA, Laule C, Moore GRW, Bove R, Cai Z, Cohen-Adad J, Harrison DM, Klawiter EC, Morrow SA, Öz G, Rooney WD, Smith SA, Calabresi PA, Henry RG, Oh J, Ontaneda D, Pelletier D, Reich DS, Shinohara RT, Sicotte NL. Imaging Mechanisms of Disease Progression in Multiple Sclerosis: Beyond Brain Atrophy. J Neuroimaging 2021; 30:251-266. [PMID: 32418324 DOI: 10.1111/jon.12700] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinicians involved with different aspects of the care of persons with multiple sclerosis (MS) and scientists with expertise on clinical and imaging techniques convened in Dallas, TX, USA on February 27, 2019 at a North American Imaging in Multiple Sclerosis Cooperative workshop meeting. The aim of the workshop was to discuss cardinal pathobiological mechanisms implicated in the progression of MS and novel imaging techniques, beyond brain atrophy, to unravel these pathologies. Indeed, although brain volume assessment demonstrates changes linked to disease progression, identifying the biological mechanisms leading up to that volume loss are key for understanding disease mechanisms. To this end, the workshop focused on the application of advanced magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging techniques to assess and measure disease progression in both the brain and the spinal cord. Clinical translation of quantitative MRI was recognized as of vital importance, although the need to maintain a relatively short acquisition time mandated by most radiology departments remains the major obstacle toward this effort. Regarding PET, the panel agreed upon its utility to identify ongoing pathological processes. However, due to costs, required expertise, and the use of ionizing radiation, PET was not considered to be a viable option for ongoing care of persons with MS. Collaborative efforts fostering robust study designs and imaging technique standardization across scanners and centers are needed to unravel disease mechanisms leading to progression and discovering medications halting neurodegeneration and/or promoting repair.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan A Gauthier
- Judith Jaffe Multiple Sclerosis Center, Department of Neurology, Feil Family Brain and Mind Institute, and Department of Radiology, Weill Cornell Medicine, New York, NY
| | - Cornelia Laule
- Department of Radiology, Pathology, and Laboratory Medicine, Department of Physics and Astronomy, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - George R Wayne Moore
- Department of Pathology and Laboratory Medicine, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Riley Bove
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University, New Haven, CT
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal and Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD
| | - Eric C Klawiter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN
| | - William D Rooney
- Advanced Imaging Research Center, Departments of Biomedical Engineering, Neurology, and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Seth A Smith
- Radiology and Radiological Sciences and Vanderbilt University Imaging Institute, Vanderbilt University Medical Center, and Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roland G Henry
- Departments of Neurology, Radiology and Biomedical Imaging, and the UC San Francisco & Berkeley Bioengineering Graduate Group, University of California San Francisco, San Francisco, CA
| | - Jiwon Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Neurology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel Pelletier
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| | -
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
136
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
137
|
Abstract
PURPOSE OF REVIEW In multiple sclerosis, currently approved disease-modifying treatments are effective in modulating peripheral immunity, and coherently, in reducing clinical/radiological relapses, but still, they perform poorly in preventing disease progression and overall disability accrual. This review provides an up-to-date overview of the neuropathology of progressive multiple sclerosis, including a summary of the main mechanisms of disease progression. RECENT FINDINGS Clinical progression in multiple sclerosis is likely related to the accumulation of neuro-axonal loss in a lifelong inflammatory CNS environment (both adaptive and innate) and relative un-balance between damage, repair and brain functional reserve. A critical driver appears to be the T-cell and B-cell-mediated compartmentalized inflammation within the leptomeninges and within the parenchyma. Recent perspective highlighted also the role of the glial response to such lifelong inflammatory injury as the critical player for both pathological and clinical outcomes. SUMMARY The neuropathological and biological understanding of disease progression in multiple sclerosis have progressed in the last few years. As a consequence, new therapeutic approaches are emerging outside the modulation of T-cell activity and/or the depletion of B cells.
Collapse
|
138
|
Magliozzi R, Pitteri M, Ziccardi S, Pisani AI, Montibeller L, Marastoni D, Rossi S, Mazziotti V, Guandalini M, Dapor C, Schiavi G, Tamanti A, Nicholas R, Reynolds R, Calabrese M. CSF parvalbumin levels reflect interneuron loss linked with cortical pathology in multiple sclerosis. Ann Clin Transl Neurol 2021; 8:534-547. [PMID: 33484486 PMCID: PMC7951111 DOI: 10.1002/acn3.51298] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction and methods In order to verify whether parvalbumin (PVALB), a protein specifically expressed by GABAergic interneurons, could be a MS‐specific marker of grey matter neurodegeneration, we performed neuropathology/molecular analysis of PVALB expression in motor cortex of 40 post‐mortem progressive MS cases, with/without meningeal inflammation, and 10 control cases, in combination with cerebrospinal fluid (CSF) assessment. Analysis of CSF PVALB and neurofilaments (Nf‐L) levels combined with physical/cognitive/3TMRI assessment was performed in 110 naïve MS patients and in 32 controls at time of diagnosis. Results PVALB gene expression was downregulated in MS (fold change = 3.7 ± 1.2, P < 0.001 compared to controls) reflecting the significant reduction of PVALB+ cell density in cortical lesions, to a greater extent in MS patients with high meningeal inflammation (51.8, P < 0.001). Likewise, post‐mortem CSF‐PVALB levels were higher in MS compared to controls (fold change = 196 ± 36, P < 0.001) and correlated with decreased PVALB+ cell density (r = −0.64, P < 0.001) and increased MHC‐II+ microglia density (r = 0.74, P < 0.01), as well as with early age of onset (r = −0.69, P < 0.05), shorter time to wheelchair (r = −0.49, P < 0.05) and early age of death (r = −0.65, P < 0.01). Increased CSF‐PVALB levels were detected in MS patients at diagnosis compared to controls (P = 0.002). Significant correlation was found between CSF‐PVALB levels and cortical lesion number on MRI (R = 0.28, P = 0.006) and global cortical thickness (R = −0.46, P < 0.001), better than Nf‐L levels. CSF‐PVALB levels increased in MS patients with severe cognitive impairment (mean ± SEM:25.2 ± 7.5 ng/mL) compared to both cognitively normal (10.9 ± 2.4, P = 0.049) and mild cognitive impaired (10.1 ± 2.9, P = 0.024) patients. Conclusions CSF‐PVALB levels reflect loss of cortical interneurons in MS patients with more severe disease course and might represent an early, new MS‐specific biomarker of cortical neurodegeneration, atrophy, and cognitive decline.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.,Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Marco Pitteri
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Stefano Ziccardi
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Anna Isabella Pisani
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Luigi Montibeller
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Damiano Marastoni
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Stefania Rossi
- Department of Oncology and Molecular Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Mazziotti
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Maddalena Guandalini
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Caterina Dapor
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Gianmarco Schiavi
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Agnese Tamanti
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Richard Nicholas
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Massimiliano Calabrese
- Neurology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
139
|
Fransen NL, Hsiao CC, van der Poel M, Engelenburg HJ, Verdaasdonk K, Vincenten MCJ, Remmerswaal EBM, Kuhlmann T, Mason MRJ, Hamann J, Smolders J, Huitinga I. Tissue-resident memory T cells invade the brain parenchyma in multiple sclerosis white matter lesions. Brain 2021; 143:1714-1730. [PMID: 32400866 DOI: 10.1093/brain/awaa117] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/10/2020] [Accepted: 02/28/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis is a chronic inflammatory, demyelinating disease, although it has been suggested that in the progressive late phase, inflammatory lesion activity declines. We recently showed in the Netherlands Brain Bank multiple sclerosis-autopsy cohort considerable ongoing inflammatory lesion activity also at the end stage of the disease, based on microglia/macrophage activity. We have now studied the role of T cells in this ongoing inflammatory lesion activity in chronic multiple sclerosis autopsy cases. We quantified T cells and perivascular T-cell cuffing at a standardized location in the medulla oblongata in 146 multiple sclerosis, 20 neurodegenerative control and 20 non-neurological control brain donors. In addition, we quantified CD3+, CD4+, and CD8+ T cells in 140 subcortical white matter lesions. The location of CD8+ T cells in either the perivascular space or the brain parenchyma was determined using CD8/laminin staining and confocal imaging. Finally, we analysed CD8+ T cells, isolated from fresh autopsy tissues from subcortical multiple sclerosis white matter lesions (n = 8), multiple sclerosis normal-appearing white matter (n = 7), and control white matter (n = 10), by flow cytometry. In normal-appearing white matter, the number of T cells was increased compared to control white matter. In active and mixed active/inactive lesions, the number of T cells was further augmented compared to normal-appearing white matter. Active and mixed active/inactive lesions were enriched for both CD4+ and CD8+ T cells, the latter being more abundant in all lesion types. Perivascular clustering of T cells in the medulla oblongata was only found in cases with a progressive disease course and correlated with a higher percentage of mixed active/inactive lesions and a higher lesion load compared to cases without perivascular clusters in the medulla oblongata. In all white matter samples, CD8+ T cells were located mostly in the perivascular space, whereas in mixed active/inactive lesions, 16.3% of the CD8+ T cells were encountered in the brain parenchyma. CD8+ T cells from mixed active/inactive lesions showed a tissue-resident memory phenotype with expression of CD69, CD103, CD44, CD49a, and PD-1 and absence of S1P1. They upregulated markers for homing (CXCR6), reactivation (Ki-67), and cytotoxicity (GPR56), yet lacked the cytolytic enzyme granzyme B. These data show that in chronic progressive multiple sclerosis cases, inflammatory lesion activity and demyelinated lesion load is associated with an increased number of T cells clustering in the perivascular space. Inflammatory active multiple sclerosis lesions are populated by CD8+ tissue-resident memory T cells, which show signs of reactivation and infiltration of the brain parenchyma.
Collapse
Affiliation(s)
- Nina L Fransen
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Cheng-Chih Hsiao
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marlijn van der Poel
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Hendrik J Engelenburg
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Kim Verdaasdonk
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Maria C J Vincenten
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands.,Renal Transplant Unit, Department of Internal Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Tanja Kuhlmann
- Institute for Neuropathology, University Hospital Münster, Münster, Germany
| | - Matthew R J Mason
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Jörg Hamann
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Joost Smolders
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.,MS center ErasMS, Departments of Neurology and Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
140
|
Morgan BP, Gommerman JL, Ramaglia V. An "Outside-In" and "Inside-Out" Consideration of Complement in the Multiple Sclerosis Brain: Lessons From Development and Neurodegenerative Diseases. Front Cell Neurosci 2021; 14:600656. [PMID: 33488361 PMCID: PMC7817777 DOI: 10.3389/fncel.2020.600656] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
The last 15 years have seen an explosion of new findings on the role of complement, a major arm of the immune system, in the central nervous system (CNS) compartment including contributions to cell migration, elimination of synapse during development, aberrant synapse pruning in neurologic disorders, damage to nerve cells in autoimmune diseases, and traumatic injury. Activation of the complement system in multiple sclerosis (MS) is typically thought to occur as part of a primary (auto)immune response from the periphery (the outside) against CNS antigens (the inside). However, evidence of local complement production from CNS-resident cells, intracellular complement functions, and the more recently discovered role of early complement components in shaping synaptic circuits in the absence of inflammation opens up the possibility that complement-related sequelae may start and finish within the brain itself. In this review, the complement system will be introduced, followed by evidence that implicates complement in shaping the developing, adult, and normal aging CNS as well as its contribution to pathology in neurodegenerative conditions. Discussion of data supporting "outside-in" vs. "inside-out" roles of complement in MS will be presented, concluded by thoughts on potential approaches to therapies targeting specific elements of the complement system.
Collapse
Affiliation(s)
- B. Paul Morgan
- UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
141
|
Beltrán E, Paunovic M, Gebert D, Cesur E, Jeitler M, Höftberger R, Malotka J, Mader S, Kawakami N, Meinl E, Bradl M, Dornmair K, Lassmann H. Archeological neuroimmunology: resurrection of a pathogenic immune response from a historical case sheds light on human autoimmune encephalomyelitis and multiple sclerosis. Acta Neuropathol 2021; 141:67-83. [PMID: 33242149 PMCID: PMC7785560 DOI: 10.1007/s00401-020-02239-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022]
Abstract
Aim of our study was to identify the target auto-antigen in the central nervous system recognized by the immune system of a unique patient, who died more than 60 years ago from a disease with pathological changes closely resembling multiple sclerosis (MS), following a misguided immunization with lyophilized calf brain tissue. Total mRNA was isolated from formaldehyde fixed and paraffin embedded archival brain tissue containing chronic active inflammatory demyelinating lesions with inflammatory infiltrates rich in B-lymphocytes and plasma cells. Analysis of the transcriptome by next generation sequencing and reconstruction of the dominant antibody by bioinformatic tools revealed the presence of one strongly expanded B-cell clone, producing an autoantibody against a conformational epitope of myelin oligodendrocytes glycoprotein (MOG), similar to that recognized by the well characterized monoclonal anti-MOG antibody 8-18C5. The reconstructed antibody induced demyelination after systemic or intrathecal injection into animals with T-cell mediated encephalomyelitis. Our study suggests that immunization with bovine brain tissue in humans may-in a small subset of patients-induce a disease with an intermediate clinical and pathological presentation between MS and MOG-antibody associated inflammatory demyelinating disease (MOGAD).
Collapse
Affiliation(s)
- Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Manuela Paunovic
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - David Gebert
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Emine Cesur
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Markus Jeitler
- Core Facility Genomics, Medical University Vienna, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Joachim Malotka
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Simone Mader
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Naoto Kawakami
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Klaus Dornmair
- Institute of Clinical Neuroimmunology, University Hospital and Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hans Lassmann
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
142
|
Kerbrat A, Gros C, Badji A, Bannier E, Galassi F, Combès B, Chouteau R, Labauge P, Ayrignac X, Carra-Dalliere C, Maranzano J, Granberg T, Ouellette R, Stawiarz L, Hillert J, Talbott J, Tachibana Y, Hori M, Kamiya K, Chougar L, Lefeuvre J, Reich DS, Nair G, Valsasina P, Rocca MA, Filippi M, Chu R, Bakshi R, Callot V, Pelletier J, Audoin B, Maarouf A, Collongues N, De Seze J, Edan G, Cohen-Adad J. Multiple sclerosis lesions in motor tracts from brain to cervical cord: spatial distribution and correlation with disability. Brain 2020; 143:2089-2105. [PMID: 32572488 DOI: 10.1093/brain/awaa162] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/27/2020] [Accepted: 04/02/2020] [Indexed: 11/12/2022] Open
Abstract
Despite important efforts to solve the clinico-radiological paradox, correlation between lesion load and physical disability in patients with multiple sclerosis remains modest. One hypothesis could be that lesion location in corticospinal tracts plays a key role in explaining motor impairment. In this study, we describe the distribution of lesions along the corticospinal tracts from the cortex to the cervical spinal cord in patients with various disease phenotypes and disability status. We also assess the link between lesion load and location within corticospinal tracts, and disability at baseline and 2-year follow-up. We retrospectively included 290 patients (22 clinically isolated syndrome, 198 relapsing remitting, 39 secondary progressive, 31 primary progressive multiple sclerosis) from eight sites. Lesions were segmented on both brain (T2-FLAIR or T2-weighted) and cervical (axial T2- or T2*-weighted) MRI scans. Data were processed using an automated and publicly available pipeline. Brain, brainstem and spinal cord portions of the corticospinal tracts were identified using probabilistic atlases to measure the lesion volume fraction. Lesion frequency maps were produced for each phenotype and disability scores assessed with Expanded Disability Status Scale score and pyramidal functional system score. Results show that lesions were not homogeneously distributed along the corticospinal tracts, with the highest lesion frequency in the corona radiata and between C2 and C4 vertebral levels. The lesion volume fraction in the corticospinal tracts was higher in secondary and primary progressive patients (mean = 3.6 ± 2.7% and 2.9 ± 2.4%), compared to relapsing-remitting patients (1.6 ± 2.1%, both P < 0.0001). Voxel-wise analyses confirmed that lesion frequency was higher in progressive compared to relapsing-remitting patients, with significant bilateral clusters in the spinal cord corticospinal tracts (P < 0.01). The baseline Expanded Disability Status Scale score was associated with lesion volume fraction within the brain (r = 0.31, P < 0.0001), brainstem (r = 0.45, P < 0.0001) and spinal cord (r = 0.57, P < 0.0001) corticospinal tracts. The spinal cord corticospinal tracts lesion volume fraction remained the strongest factor in the multiple linear regression model, independently from cord atrophy. Baseline spinal cord corticospinal tracts lesion volume fraction was also associated with disability progression at 2-year follow-up (P = 0.003). Our results suggest a cumulative effect of lesions within the corticospinal tracts along the brain, brainstem and spinal cord portions to explain physical disability in multiple sclerosis patients, with a predominant impact of intramedullary lesions.
Collapse
Affiliation(s)
- Anne Kerbrat
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Canada.,CHU Rennes, Neurology department, Empenn U 1128 Inserm, CIC1414 Inserm, Rennes, France
| | - Charley Gros
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Canada
| | - Atef Badji
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada
| | - Elise Bannier
- CHU Rennes, Radiology department, Rennes, France.,Univ Rennes, Inria, CNRS, Inserm, IRISA UMR 6074, Empenn U1128, Rennes, France
| | - Francesca Galassi
- Univ Rennes, Inria, CNRS, Inserm, IRISA UMR 6074, Empenn U1128, Rennes, France
| | - Benoit Combès
- Univ Rennes, Inria, CNRS, Inserm, IRISA UMR 6074, Empenn U1128, Rennes, France
| | - Raphaël Chouteau
- CHU Rennes, Neurology department, Empenn U 1128 Inserm, CIC1414 Inserm, Rennes, France
| | - Pierre Labauge
- MS Unit, Department of Neurology, CHU Montpellier, Montpellier, France
| | - Xavier Ayrignac
- MS Unit, Department of Neurology, CHU Montpellier, Montpellier, France
| | | | - Josefina Maranzano
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada.,University of Quebec in Trois-Rivieres, Quebec, Canada
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Russell Ouellette
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Leszek Stawiarz
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Hillert
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jason Talbott
- Department of Radiology and Biomedical Imaging, Zuckerberg San Francisco General Hospital, University of California, San Francisco, CA, USA
| | | | - Masaaki Hori
- Toho University Omori Medical Center, Tokyo, Japan
| | | | - Lydia Chougar
- Department of Neuroradiology, La Pitié Salpêtrière Hospital, Paris, France
| | - Jennifer Lefeuvre
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Maryland, USA
| | - Daniel S Reich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Maryland, USA
| | - Govind Nair
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Maryland, USA
| | - Paola Valsasina
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, and Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, and Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, and Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Renxin Chu
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Rohit Bakshi
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Virginie Callot
- AP-HM, Pôle d'imagerie médicale, Hôpital de la Timone, CEMEREM, Marseille, France.,Aix-Marseille Univ, CNRS, CRMBM, Marseille, France
| | - Jean Pelletier
- Aix-Marseille Univ, CNRS, CRMBM, Marseille, France.,AP-HM, CHU Timone, Pôle de Neurosciences Cliniques, Department of Neurology, Marseille, France
| | - Bertrand Audoin
- Aix-Marseille Univ, CNRS, CRMBM, Marseille, France.,AP-HM, CHU Timone, Pôle de Neurosciences Cliniques, Department of Neurology, Marseille, France
| | - Adil Maarouf
- Aix-Marseille Univ, CNRS, CRMBM, Marseille, France.,AP-HM, CHU Timone, Pôle de Neurosciences Cliniques, Department of Neurology, Marseille, France
| | - Nicolas Collongues
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 67 000 Strasbourg, France.,Département de Neurologie, Centre Hospitalier Universitaire de Strasbourg, 67200 Strasbourg, France.,Centre d'investigation Clinique, INSERM U1434, Centre Hospitalier Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Jérôme De Seze
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment 3 de la Faculté de Médecine, 67 000 Strasbourg, France.,Département de Neurologie, Centre Hospitalier Universitaire de Strasbourg, 67200 Strasbourg, France.,Centre d'investigation Clinique, INSERM U1434, Centre Hospitalier Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Gilles Edan
- CHU Rennes, Neurology department, Empenn U 1128 Inserm, CIC1414 Inserm, Rennes, France
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, Canada.,Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Canada
| |
Collapse
|
143
|
Kleerekooper I, Houston S, Dubis AM, Trip SA, Petzold A. Optical Coherence Tomography Angiography (OCTA) in Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder. Front Neurol 2020; 11:604049. [PMID: 33362705 PMCID: PMC7758345 DOI: 10.3389/fneur.2020.604049] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/17/2020] [Indexed: 12/29/2022] Open
Abstract
Vascular changes are increasingly recognized as important factors in the pathophysiology of neuroinflammatory disease, especially in multiple sclerosis (MS). The relatively novel technology of optical coherence tomography angiography (OCTA) images the retinal and choroidal vasculature non-invasively and in a depth-resolved manner. OCTA provides an alternative quantitative measure of retinal damage, by measuring vascular density instead of structural atrophy. Preliminary results suggest OCTA is sensitive to retinal damage in early disease stages, while also having less of a "floor-effect" compared with commonly used OCT metrics, meaning it can pick up further damage in a severely atrophied retina in later stages of disease. Furthermore, it may serve as a surrogate marker for vascular pathology in the central nervous system. Data to date consistently reveal lower densities of the retinal microvasculature in both MS and neuromyelitis optica spectrum disorder (NMOSD) compared with healthy controls, even in the absence of prior optic neuritis. Exploring the timing of vascular changes relative to structural atrophy may help answer important questions about the role of hypoperfusion in the pathophysiology of neuroinflammatory disease. Finally, qualitative characteristics of retinal microvasculature may help discriminate between different neuroinflammatory disorders. There are however still issues regarding image quality and development of standardized analysis methods before OCTA can be fully incorporated into clinical practice.
Collapse
Affiliation(s)
- Iris Kleerekooper
- Department of Neuro-Ophthalmology, Moorfields Eye Hospital, London, United Kingdom.,Queen Square MS Centre, UCL Institute of Neurology and National Hospital for Neurology & Neurosurgery, London, United Kingdom
| | - Sarah Houston
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Adam M Dubis
- National Institute for Health Research, Biomedical Resource Centre at University College London, Institute of Ophthalmology and Moorfields Eye Hospital National Health Service Trust, London, United Kingdom
| | - S Anand Trip
- Queen Square MS Centre, UCL Institute of Neurology and National Hospital for Neurology & Neurosurgery, London, United Kingdom
| | - Axel Petzold
- Department of Neuro-Ophthalmology, Moorfields Eye Hospital, London, United Kingdom.,Queen Square MS Centre, UCL Institute of Neurology and National Hospital for Neurology & Neurosurgery, London, United Kingdom.,Dutch Expertise Centre of Neuro-Ophthalmology, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
144
|
Sekera ER, Saraswat D, Zemaitis KJ, Sim FJ, Wood TD. MALDI Mass Spectrometry Imaging in a Primary Demyelination Model of Murine Spinal Cord. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2462-2468. [PMID: 32926612 PMCID: PMC8628303 DOI: 10.1021/jasms.0c00187] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Destruction of myelin, or demyelination, is a characteristic of traumatic spinal cord injury and pathognomonic for primary demyelinating pathologies such as multiple sclerosis (MS). The regenerative process known as remyelination, which can occur following demyelination, fails as MS progresses. Models of focal demyelination by local injection of gliotoxins have provided important biological insights into the demyelination/remyelination process. Here, injection of lysolecithin to induce spinal cord demyelination is investigated using matrix-assisted laser desorption/ionization mass spectrometry imaging. A segmentation analysis revealed changes to the lipid composition during lysolecithin-induced demyelination at the lesion site and subsequent remyelination over time. The results of this study can be utilized to identify potential myelin-repair mechanisms and in the design of therapeutic strategies to enhance myelin repair.
Collapse
Affiliation(s)
- Emily R Sekera
- Department of Chemistry, Natural Sciences Complex, University at Buffalo State University of New York, Buffalo, New York 14260-3000, United States
| | - Darpan Saraswat
- Department of Pharmacology & Toxicology, Jacobs School of Medicine and Biomedical Sciences, 955 Main Street, University at Buffalo State University of New York, Buffalo, New York 14203, United States
| | - Kevin J Zemaitis
- Department of Chemistry, Natural Sciences Complex, University at Buffalo State University of New York, Buffalo, New York 14260-3000, United States
| | - Fraser J Sim
- Department of Pharmacology & Toxicology, Jacobs School of Medicine and Biomedical Sciences, 955 Main Street, University at Buffalo State University of New York, Buffalo, New York 14203, United States
| | - Troy D Wood
- Department of Chemistry, Natural Sciences Complex, University at Buffalo State University of New York, Buffalo, New York 14260-3000, United States
| |
Collapse
|
145
|
Hauser SL, Cree BAC. Treatment of Multiple Sclerosis: A Review. Am J Med 2020; 133:1380-1390.e2. [PMID: 32682869 PMCID: PMC7704606 DOI: 10.1016/j.amjmed.2020.05.049] [Citation(s) in RCA: 392] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 10/23/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune demyelinating and neurodegenerative disease of the central nervous system, and the leading cause of nontraumatic neurological disability in young adults. Effective management requires a multifaceted approach to control acute attacks, manage progressive worsening, and remediate bothersome or disabling symptoms associated with this illness. Remarkable advances in treatment of all forms of MS, and especially for relapsing disease, have favorably changed the long-term outlook for many patients. There also has been a conceptual shift in understanding the immune pathology of MS, away from a purely T-cell-mediated model to recognition that B cells have a key role in pathogenesis. The emergence of higher-efficacy drugs requiring less frequent administration have made these preferred options in terms of tolerability and adherence. Many experts now recommend use of these as first-line treatment for many patients with early disease, before permanent disability is evident.
Collapse
Affiliation(s)
- Stephen L Hauser
- UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco.
| | - Bruce A C Cree
- UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco
| |
Collapse
|
146
|
Weeda MM, Pruis IJ, Westerveld ASR, Brouwer I, Bellenberg B, Barkhof F, Vrenken H, Lukas C, Schneider R, Pouwels PJW. Damage in the Thalamocortical Tracts is Associated With Subsequent Thalamus Atrophy in Early Multiple Sclerosis. Front Neurol 2020; 11:575611. [PMID: 33281710 PMCID: PMC7705066 DOI: 10.3389/fneur.2020.575611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/05/2020] [Indexed: 01/01/2023] Open
Abstract
Background: In early multiple sclerosis (MS), thalamus atrophy and decreased integrity of the thalamocortical white matter (WM) tracts have been observed. Objective: To investigate the temporal association between thalamus volume and WM damage in the thalamocortical tract in subjects with early MS. Methods: At two time points, 72 subjects with early MS underwent T1, FLAIR and diffusion tensor imaging. Thalamocortical tracts were identified with probabilistic tractography using left and right thalamus as seed regions. Regression analysis was performed to identify predictors of annual percentage change in both thalamus volumes and integrity of the connected tracts. Results: Significant atrophy was seen in left and right thalamus (p < 0.001) over the follow-up period (13.7 ± 4.8 months), whereas fractional anisotropy (FA) and mean diffusivity (MD) changes of the left and right thalamus tracts were not significant, although large inter-subject variability was seen. Annual percentage change in left thalamus volume was significantly predicted by baseline FA of the left thalamus tracts F(1.71) = 4.284, p = 0.042; while no such relation was found for the right thalamus. Annual percentage change in FA or MD of the thalamus tracts was not predicted by thalamus volume or any of the demographic parameters. Conclusion: Over a short follow-up time, thalamus atrophy could be predicted by decreased integrity of the thalamic tracts, but changes in the integrity of the thalamic tracts could not be predicted by thalamus volume. This is the first study showing directionality in the association between thalamus atrophy and connected WM tract damage. These results need to be verified over longer follow-up periods.
Collapse
Affiliation(s)
- Merlin M Weeda
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| | - Ilanah J Pruis
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| | - Aimee S R Westerveld
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| | - Iman Brouwer
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| | - Barbara Bellenberg
- Institute of Neuroradiology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands.,Institutes of Neurology and Healthcare Engineering, University College London, London, United Kingdom
| | - Hugo Vrenken
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| | - Carsten Lukas
- Institute of Neuroradiology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Ruth Schneider
- Institute of Neuroradiology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany.,Department of Neurology, St. Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Petra J W Pouwels
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-Location VUmc, Amsterdam, Netherlands
| |
Collapse
|
147
|
Beutel T, Dzimiera J, Kapell H, Engelhardt M, Gass A, Schirmer L. Cortical projection neurons as a therapeutic target in multiple sclerosis. Expert Opin Ther Targets 2020; 24:1211-1224. [PMID: 33103501 DOI: 10.1080/14728222.2020.1842358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory-demyelinating disease of the central nervous system associated with lesions of the cortical gray matter and subcortical white matter. Recently, cortical lesions have become a major focus of research because cortical pathology and neuronal damage are critical determinants of irreversible clinical progression. Recent transcriptomic studies point toward cell type-specific changes in cortical neurons in MS with a selective vulnerability of excitatory projection neuron subtypes. AREAS COVERED We discuss the cortical mapping and the molecular properties of excitatory projection neurons and their role in MS lesion pathology while placing an emphasis on their subtype-specific transcriptomic changes and levels of vulnerability. We also examine the latest magnetic resonance imaging techniques to study cortical MS pathology as a key tool for monitoring disease progression and treatment efficacy. Finally, we consider possible therapeutic avenues and novel strategies to protect excitatory cortical projection neurons. Literature search methodology: PubMed articles from 2000-2020. EXPERT OPINION Excitatory cortical projection neurons are an emerging therapeutic target in the treatment of progressive MS. Understanding neuron subtype-specific molecular pathologies and their exact spatial mapping will help establish starting points for the development of novel cell type-specific therapies and biomarkers in MS.
Collapse
Affiliation(s)
- Tatjana Beutel
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Julia Dzimiera
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Hannah Kapell
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Maren Engelhardt
- Institute of Neuroanatomy, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| | - Achim Gass
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University , Mannheim, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University , Heidelberg, Germany
| |
Collapse
|
148
|
Lommers E, Guillemin C, Reuter G, Fouarge E, Delrue G, Collette F, Degueldre C, Balteau E, Maquet P, Phillips C. Voxel-Based quantitative MRI reveals spatial patterns of grey matter alteration in multiple sclerosis. Hum Brain Mapp 2020; 42:1003-1012. [PMID: 33155763 PMCID: PMC7856642 DOI: 10.1002/hbm.25274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/10/2020] [Accepted: 10/22/2020] [Indexed: 12/22/2022] Open
Abstract
Despite robust postmortem evidence and potential clinical importance of gray matter (GM) pathology in multiple sclerosis (MS), assessing GM damage by conventional magnetic resonance imaging (MRI) remains challenging. This prospective cross‐sectional study aimed at characterizing the topography of GM microstructural and volumetric alteration in MS using, in addition to brain atrophy measures, three quantitative MRI (qMRI) parameters—magnetization transfer (MT) saturation, longitudinal (R1), and effective transverse (R2*) relaxation rates, derived from data acquired during a single scanning session. Our study involved 35 MS patients (14 relapsing–remitting MS; 21 primary or secondary progressive MS) and 36 age‐matched healthy controls (HC). The qMRI maps were computed and segmented in different tissue classes. Voxel‐based quantification (VBQ) and voxel‐based morphometry (VBM) statistical analyses were carried out using multiple linear regression models. In MS patients compared with HC, three configurations of GM microstructural/volumetric alterations were identified. (a) Co‐localization of GM atrophy with significant reduction of MT, R1, and/or R2*, usually observed in primary cortices. (b) Microstructural modifications without significant GM loss: hippocampus and paralimbic cortices, showing reduced MT and/or R1 values without significant atrophy. (c) Atrophy without significant change in microstructure, identified in deep GM nuclei. In conclusion, this quantitative multiparametric voxel‐based approach reveals three different spatially‐segregated combinations of GM microstructural/volumetric alterations in MS that might be associated with different neuropathology.
Collapse
Affiliation(s)
- Emilie Lommers
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,Clinical Neuroimmunology Unit, Neurology Department, CHU Liège, Liège, Belgium
| | - Camille Guillemin
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,Psychology and Neuroscience of Cognition Research Unit, University of Liège, Liège, Belgium
| | - Gilles Reuter
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,Neurosurgery Department, CHU Liège, Liège, Belgium
| | - Eve Fouarge
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium
| | - Gaël Delrue
- Clinical Neuroimmunology Unit, Neurology Department, CHU Liège, Liège, Belgium
| | - Fabienne Collette
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,Psychology and Neuroscience of Cognition Research Unit, University of Liège, Liège, Belgium
| | | | - Evelyne Balteau
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium
| | - Pierre Maquet
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,Clinical Neuroimmunology Unit, Neurology Department, CHU Liège, Liège, Belgium
| | - Christophe Phillips
- GIGA - CRC in vivo imaging, University of Liège, Liège, Belgium.,GIGA - in silico medicine, University of Liège, Liège, Belgium
| |
Collapse
|
149
|
Sacco S, Caverzasi E, Papinutto N, Cordano C, Bischof A, Gundel T, Cheng S, Asteggiano C, Kirkish G, Mallott J, Stern WA, Bastianello S, Bove RM, Gelfand JM, Goodin DS, Green AJ, Waubant E, Wilson MR, Zamvil SS, Cree BA, Hauser SL, Henry RG. Neurite Orientation Dispersion and Density Imaging for Assessing Acute Inflammation and Lesion Evolution in MS. AJNR Am J Neuroradiol 2020; 41:2219-2226. [PMID: 33154077 DOI: 10.3174/ajnr.a6862] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/29/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND PURPOSE MR imaging is essential for MS diagnosis and management, yet it has limitations in assessing axonal damage and remyelination. Gadolinium-based contrast agents add value by pinpointing acute inflammation and blood-brain barrier leakage, but with drawbacks in safety and cost. Neurite orientation dispersion and density imaging (NODDI) assesses microstructural features of neurites contributing to diffusion imaging signals. This approach may resolve the components of MS pathology, overcoming conventional MR imaging limitations. MATERIALS AND METHODS Twenty-one subjects with MS underwent serial enhanced MRIs (12.6 ± 9 months apart) including NODDI, whose key metrics are the neurite density and orientation dispersion index. Twenty-one age- and sex-matched healthy controls underwent unenhanced MR imaging with the same protocol. Fifty-eight gadolinium-enhancing and non-gadolinium-enhancing lesions were semiautomatically segmented at baseline and follow-up. Normal-appearing WM masks were generated by subtracting lesions and dirty-appearing WM from the whole WM. RESULTS The orientation dispersion index was higher in gadolinium-enhancing compared with non-gadolinium-enhancing lesions; logistic regression indicated discrimination, with an area under the curve of 0.73. At follow-up, in the 58 previously enhancing lesions, we identified 2 subgroups based on the neurite density index change across time: Type 1 lesions showed increased neurite density values, whereas type 2 lesions showed decreased values. Type 1 lesions showed greater reduction in size with time compared with type 2 lesions. CONCLUSIONS NODDI is a promising tool with the potential to detect acute MS inflammation. The observed heterogeneity among lesions may correspond to gradients in severity and clinical recovery after the acute phase.
Collapse
Affiliation(s)
- S Sacco
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California.,Institute of Radiology (S.S., C.A.), Department of Clinical Surgical Diagnostic and Pediatric Sciences
| | - E Caverzasi
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - N Papinutto
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - C Cordano
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - A Bischof
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - T Gundel
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - S Cheng
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - C Asteggiano
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California.,Institute of Radiology (S.S., C.A.), Department of Clinical Surgical Diagnostic and Pediatric Sciences
| | - G Kirkish
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - J Mallott
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - W A Stern
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - S Bastianello
- Department of Brain and Behavioral Sciences (S.B.), University of Pavia, Pavia, Italy.,Neuroradiology Department (S.B.), Istituto Di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia, Italy
| | - R M Bove
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - J M Gelfand
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - D S Goodin
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - A J Green
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - E Waubant
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - M R Wilson
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - S S Zamvil
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - B A Cree
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - S L Hauser
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | - R G Henry
- From the Department of Neurology (S.S., E.C., N.P., C.C., A.B., T.G., S.C., C.A., G.K., J.M., W.A.S., R.M.B., J.M.G., D.S.G., A.J.G., E.W., M.R.W., S.S.Z, B.A.C., S.L.H., and R.G.H.), University of California, San Francisco Weill Institute for Neurosciences, University of California, San Francisco, California
| | | |
Collapse
|
150
|
Monaco S, Nicholas R, Reynolds R, Magliozzi R. Intrathecal Inflammation in Progressive Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21218217. [PMID: 33153042 PMCID: PMC7663229 DOI: 10.3390/ijms21218217] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 01/05/2023] Open
Abstract
Progressive forms of multiple sclerosis (MS) are associated with chronic demyelination, axonal loss, neurodegeneration, cortical and deep gray matter damage, and atrophy. These changes are strictly associated with compartmentalized sustained inflammation within the brain parenchyma, the leptomeninges, and the cerebrospinal fluid. In progressive MS, molecular mechanisms underlying active demyelination differ from processes that drive neurodegeneration at cortical and subcortical locations. The widespread pattern of neurodegeneration is consistent with mechanisms associated with the inflammatory molecular load of the cerebrospinal fluid. This is at variance with gray matter demyelination that typically occurs at focal subpial sites, in the proximity of ectopic meningeal lymphoid follicles. Accordingly, it is possible that variations in the extent and location of neurodegeneration may be accounted for by individual differences in CSF flow, and by the composition of soluble inflammatory factors and their clearance. In addition, “double hit” damage may occur at sites allowing a bidirectional exchange between interstitial fluid and CSF, such as the Virchow–Robin spaces and the periventricular ependymal barrier. An important aspect of CSF inflammation and deep gray matter damage in MS involves dysfunction of the blood–cerebrospinal fluid barrier and inflammation in the choroid plexus. Here, we provide a comprehensive review on the role of intrathecal inflammation compartmentalized to CNS and non-neural tissues in progressive MS.
Collapse
Affiliation(s)
- Salvatore Monaco
- Department of Neurosciences, Biomedicine and Movements Sciences, University of Verona, 37134 Verona, Italy
- Correspondence: (S.M.); (R.M.)
| | - Richard Nicholas
- Department of Brain Sciences, Imperial College, Faculty of Medicine, London W12 ONN, UK; (R.N.); (R.R.)
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College, Faculty of Medicine, London W12 ONN, UK; (R.N.); (R.R.)
| | - Roberta Magliozzi
- Department of Neurosciences, Biomedicine and Movements Sciences, University of Verona, 37134 Verona, Italy
- Department of Brain Sciences, Imperial College, Faculty of Medicine, London W12 ONN, UK; (R.N.); (R.R.)
- Correspondence: (S.M.); (R.M.)
| |
Collapse
|