1
|
Ravano V, Andelova M, Piredda GF, Sommer S, Caneschi S, Roccaro L, Krasensky J, Kudrna M, Uher T, Corredor-Jerez RA, Disselhorst JA, Maréchal B, Hilbert T, Thiran JP, Richiardi J, Horakova D, Vaneckova M, Kober T. Microstructural characterization of multiple sclerosis lesion phenotypes using multiparametric longitudinal analysis. J Neurol 2024; 271:5944-5957. [PMID: 39003428 PMCID: PMC11377637 DOI: 10.1007/s00415-024-12568-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND AND OBJECTIVES In multiple sclerosis (MS), slowly expanding lesions were shown to be associated with worse disability and prognosis. Their timely detection from cross-sectional data at early disease stages could be clinically relevant to inform treatment planning. Here, we propose to use multiparametric, quantitative MRI to allow a better cross-sectional characterization of lesions with different longitudinal phenotypes. METHODS We analysed T1 and T2 relaxometry maps from a longitudinal cohort of MS patients. Lesions were classified as enlarging, shrinking, new or stable based on their longitudinal volumetric change using a newly developed automated technique. Voxelwise deviations were computed as z-scores by comparing individual patient data to T1, T2 and T2/T1 normative values from healthy subjects. We studied the distribution of microstructural properties inside lesions and within perilesional tissue. RESULTS AND CONCLUSIONS Stable lesions exhibited the highest T1 and T2 z-scores in lesion tissue, while the lowest values were observed for new lesions. Shrinking lesions presented the highest T1 z-scores in the first perilesional ring while enlarging lesions showed the highest T2 z-scores in the same region. Finally, a classification model was trained to predict the longitudinal lesion type based on microstructural metrics and feature importance was assessed. Z-scores estimated in lesion and perilesional tissue from T1, T2 and T2/T1 quantitative maps carry discriminative and complementary information to classify longitudinal lesion phenotypes, hence suggesting that multiparametric MRI approaches are essential for a better understanding of the pathophysiological mechanisms underlying disease activity in MS lesions.
Collapse
Affiliation(s)
- Veronica Ravano
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland.
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Michaela Andelova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University, Prague, Czech Republic
| | - Gian Franco Piredda
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
| | - Stefan Sommer
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Swiss Center for Muscoloskeletal Imaging (SCMI) Balgrist Campus, Zurich, Switzerland
| | - Samuele Caneschi
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lucia Roccaro
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jan Krasensky
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Matej Kudrna
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomas Uher
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University, Prague, Czech Republic
| | - Ricardo A Corredor-Jerez
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jonathan A Disselhorst
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bénédicte Maréchal
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tom Hilbert
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Jonas Richiardi
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tobias Kober
- Advanced Clinical Imaging Technology, Siemens Healthineers International AG, Lausanne, Geneva and Zurich, Switzerland
- Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- LTS5, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
2
|
Würtemberger U, Rau A, Diebold M, Becker L, Hohenhaus M, Beck J, Reinacher PC, Erny D, Reisert M, Urbach H, Demerath T. Advanced diffusion MRI provides evidence for altered axonal microstructure and gradual peritumoral infiltration in GBM in comparison to brain metastases. Clin Neuroradiol 2024; 34:703-711. [PMID: 38683350 PMCID: PMC11339137 DOI: 10.1007/s00062-024-01416-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE In contrast to peritumoral edema in metastases, GBM is histopathologically characterized by infiltrating tumor cells within the T2 signal alterations. We hypothesized that depending on the distance from the outline of the contrast-enhancing tumor we might reveal imaging evidence of gradual peritumoral infiltration in GBM and predominantly vasogenic edema around metastases. We thus investigated the gradual change of advanced diffusion metrics with the peritumoral zone in metastases and GBM. METHODS In 30 patients with GBM and 28 with brain metastases, peritumoral T2 hyperintensity was segmented in 33% partitions based on the total volume beginning at the enhancing tumor margin and divided into inner, middle and outer zones. Diffusion Tensor Imaging (DTI)-derived fractional anisotropy and mean diffusivity as well as Diffusion Microstructure Imaging (DMI)-based parameters Dax-intra, Dax-extra, V‑CSF and V-intra were employed to assess group-wise differences between inner and outer zones as well as within-group gradients between the inner and outer zones. RESULTS In metastases, fractional anisotropy and Dax-extra were significantly reduced in the inner zone compared to the outer zone (FA p = 0.01; Dax-extra p = 0.03). In GBM, we noted a reduced Dax-extra and significantly lower intraaxonal volume fraction (Dax-extra p = 0.008, V‑intra p = 0.006) accompanied by elevated axial intraaxonal diffusivity in the inner zone (p = 0.035). Between-group comparison of the outer to the inner zones revealed significantly higher gradients in metastases over GBM for FA (p = 0.04) as well as the axial diffusivity in the intra- (p = 0.02) and extraaxonal compartment (p < 0.001). CONCLUSION Our findings provide evidence of gradual alterations within the peritumoral zone of brain tumors. These are compatible with predominant (vasogenic) edema formation in metastases, whereas our findings in GBM are in line with an axonal destructive component in the immediate peritumoral area and evidence of tumor cell infiltration with accentuation in the tumor's vicinity.
Collapse
Affiliation(s)
- U Würtemberger
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.
- Dept. of Neuroradiology, University Medical Center Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| | - A Rau
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Department of Diagnostic and Interventional Radiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - M Diebold
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - L Becker
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - M Hohenhaus
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - J Beck
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - P C Reinacher
- Department of Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Fraunhofer Institute for Laser Technology, 52074, Aachen, Germany
| | - D Erny
- Institute of Neuropathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - M Reisert
- Department of Medical Physics, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - H Urbach
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - T Demerath
- Department of Neuroradiology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| |
Collapse
|
3
|
Singh V, Zheng Y, Ontaneda D, Mahajan KR, Holloman J, Fox RJ, Nakamura K, Trapp BD. Disability independent of cerebral white matter demyelination in progressive multiple sclerosis. Acta Neuropathol 2024; 148:34. [PMID: 39217272 PMCID: PMC11365858 DOI: 10.1007/s00401-024-02796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
The pathogenic mechanisms contributing to neurological disability in progressive multiple sclerosis (PMS) are poorly understood. Cortical neuronal loss independent of cerebral white matter (WM) demyelination in myelocortical MS (MCMS) and identification of MS patients with widespread cortical atrophy and disability progression independent of relapse activity (PIRA) support pathogenic mechanisms other than cerebral WM demyelination. The three-dimensional distribution and underlying pathology of myelinated T2 lesions were investigated in postmortem MCMS brains. Postmortem brain slices from previously characterized MCMS (10 cases) and typical MS (TMS) cases (12 cases) were co-registered with in situ postmortem T2 hyperintensities and T1 hypointensities. T1 intensity thresholds were used to establish a classifier that differentiates MCMS from TMS. The classifier was validated in 36 uncharacterized postmortem brains and applied to baseline MRIs from 255 living PMS participants enrolled in SPRINT-MS. Myelinated T2 hyperintensities in postmortem MCMS brains have a contiguous periventricular distribution that expands at the occipital poles of the lateral ventricles where a surface-in gradient of myelinated axonal degeneration was observed. The MRI classifier distinguished pathologically confirmed postmortem MCMS and TMS cases with an accuracy of 94%. For SPRINT-MS patients, the MRI classifier identified 78% as TMS, 10% as MCMS, and 12% with a paucity of cerebral T1 and T2 intensities. In SPRINT-MS, expanded disability status scale and brain atrophy measures were similar in MCMS and TMS cohorts. A paucity of cerebral WM demyelination in 22% of living PMS patients raises questions regarding a primary role for cerebral WM demyelination in disability progression in all MS patients and has implications for clinical management of MS patients and clinical trial outcomes in PMS. Periventricular myelinated fiber degeneration provides additional support for surface-in gradients of neurodegeneration in MS.
Collapse
Affiliation(s)
- Vikas Singh
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Yufan Zheng
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel Ontaneda
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Kedar R Mahajan
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Jameson Holloman
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J Fox
- Mellen Center for Treatment and Research in MS, Cleveland Clinic, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, NC30, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
4
|
Nakamura K, Zheng Y, Mahajan KR, Cohen JA, Fox RJ, Ontaneda D. Effect of ibudilast on thalamic magnetization transfer ratio and volume in progressive multiple sclerosis. Mult Scler 2023; 29:1257-1265. [PMID: 37537928 PMCID: PMC11130979 DOI: 10.1177/13524585231187289] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
BACKGROUND Thalamic volume (TV) is a sensitive biomarker of disease burden of injury in multiple sclerosis (MS) and appears to reflect overall lesion loads. Ibudilast showed significant treatment effect on brain atrophy and magnetization transfer ratio (MTR) of normal-appearing brain tissue but not in new/enlarging T2 lesion in the SPRINT-MS randomized clinical trial. OBJECTIVE To evaluate the effect of ibudilast on thalamic tissue integrity and volume in the SPRINT-MS. METHODS A total of 255 participants with progressive MS were randomized to oral ibudilast or placebo, and thalamic MTR and normalized TV over 96 weeks were quantified. Mixed-effect modeling assessed treatment effects on the thalamic MTR and TV, separately. Similarly, the measures were compared between the participants with confirmed disability progression (CDP). RESULTS Ibudilast's treatment effect was observed compared to placebo for thalamic MTR (p = 0.03) but not for TV (p = 0.68) while TV correlated with T2 lesion volume (p < 0.001). CDP associated with thalamic MTR (p = 0.04) but not with TV (p = 0.7). CONCLUSION Ibudilast showed an effect on thalamic MTR, which was associated with CDP, suggesting a clinically relevant effect on thalamic tissue integrity. However, the treatment effect was not observed in TV, suggesting that thalamic atrophy is more closely associated with global inflammatory activity than local tissue integrity. CLINICALTRIALS.GOV NCT01982942.
Collapse
|
5
|
Jakimovski D, Qureshi F, Ramanathan M, Gehman V, Keshavan A, Leyden K, Dwyer MG, Bergsland N, Weinstock-Guttman B, Zivadinov R. Proteomics and relationship with axonal pathology in multiple sclerosis: 5-year diffusion tensor imaging study. Brain Commun 2023; 5:fcad183. [PMID: 37361716 PMCID: PMC10288551 DOI: 10.1093/braincomms/fcad183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/08/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
Blood-based biomarkers can be economic and easily accessible tools for monitoring and predicting disease activity in multiple sclerosis. The objective of this study was to determine the predictive value of a multivariate proteomic assay for concurrent and future microstructural/axonal brain pathology in a longitudinal study of a heterogeneous group of people with multiple sclerosis. A proteomic analysis was obtained on serum samples from 202 people with multiple sclerosis (148 relapsing-remitting and 54 progressive) at baseline and 5-year follow-up. The concentration of 21 proteins related to multiple pathways of multiple sclerosis pathophysiology was derived using Proximity Extension Assay on the Olink platform. Patients were imaged on the same 3T MRI scanner at both timepoints. Тhe rate of whole brain, white matter and grey matter atrophy over the 5-year follow-up was determined using the multi-timepoint Structural Image Evaluation, using Normalisation, of Atrophy algorithms. Lesion burden measures were also assessed. The severity of microstructural axonal brain pathology was quantified using diffusion tensor imaging. Fractional anisotropy and mean diffusivity of normal-appearing brain tissue, normal-appearing white matter, grey matter, T2 and T1 lesions were calculated. Age, sex and body mass index-adjusted step-wise regression models were used. Glial fibrillary acidic protein was the most common and highest-ranked proteomic biomarker associated with greater concurrent microstructural central nervous system alterations (P < 0.001). The rate of whole brain atrophy was associated with baseline levels of glial fibrillary acidic protein, protogenin precursor, neurofilament light chain and myelin oligodendrocyte (P < 0.009), whereas grey matter atrophy was associated with higher baseline neurofilament light chain, higher osteopontin and lower protogenin precursor levels (P < 0.016). Higher baseline glial fibrillary acidic protein level was a significant predictor of future severity of the microstructural CNS alterations as measured by normal-appearing brain tissue fractional anisotropy and mean diffusivity (standardized β = -0.397/0.327, P < 0.001), normal-appearing white matter fractional anisotropy (standardized β = -0.466, P < 0.0012), grey matter mean diffusivity (standardized β = 0.346, P < 0.011) and T2 lesion mean diffusivity (standardized β = 0.416, P < 0.001) at the 5-year follow-up. Serum levels of myelin-oligodendrocyte glycoprotein, neurofilament light chain, contactin-2 and osteopontin proteins were additionally and independently associated with worse concomitant and future axonal pathology. Higher glial fibrillary acidic protein levels were associated with future disability progression (Exp(B) = 8.65, P = 0.004). Multiple proteomic biomarkers are independently associated with greater severity of axonal brain pathology as measured by diffusion tensor imaging in multiple sclerosis. Baseline serum glial fibrillary acidic protein levels can predict future disability progression.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | | | - Murali Ramanathan
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | | | | | | | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- IRCCS, Fondazione Don Carlo Gnocchi, Milan 20113, Italy
| | - Bianca Weinstock-Guttman
- Jacobs Comprehensive MS Treatment and Research Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Robert Zivadinov
- Correspondence to: Robert Zivadinov, MD, PhD Department of Neurology, Jacobs School of Medicine and Biomedical Sciences Buffalo Neuroimaging Analysis Center, Center for Biomedical Imaging at Clinical Translational Science Institute University at Buffalo, 100 High St., Buffalo, NY 14203, USA E-mail:
| |
Collapse
|
6
|
Khalifa A, Ibrahim HIM, Sheikh A, Khalil HE. Probiotic-Fermented Camel Milk Attenuates Neurodegenerative Symptoms via SOX5/miR-218 Axis Orchestration in Mouse Models. Pharmaceuticals (Basel) 2023; 16:357. [PMID: 36986457 PMCID: PMC10059028 DOI: 10.3390/ph16030357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Multiple sclerosis is an autoimmune-mediated myelin damage disorder in the central nervous system that is widespread among neurological patients. It has been demonstrated that several genetic and epigenetic factors control autoimmune encephalomyelitis (EAE), a murine model of MS, through CD4+ T-cell population quantity. Alterations in the gut microbiota influence neuroprotectiveness via unexplored mechanisms. In this study, the ameliorative effect of Bacillus amyloliquefaciens fermented in camel milk (BEY) on an autoimmune-mediated neurodegenerative model using myelin oligodendrocyte glycoprotein/complete fraud adjuvant/pertussis toxin (MCP)-immunized C57BL6j mice is investigated. Anti-inflammatory activity was confirmed in the in vitro cell model, and inflammatory cytokines interleukins IL17 (from EAE 311 to BEY 227 pg/mL), IL6 (from EAE 103 to BEY 65 pg/mL), IFNγ (from EAE 423 to BEY 243 pg/mL) and TGFβ (from EAE 74 to BEY 133 pg/mL) were significantly reduced in BEY-treated mice. The epigenetic factor miR-218-5P was identified and confirmed its mRNA target SOX-5 using in silico tools and expression techniques, suggesting SOX5/miR-218-5p could serve as an exclusive diagnostic marker for MS. Furthermore, BEY improved the short-chain fatty acids, in particular butyrate (from 0.57 to 0.85 µM) and caproic (from 0.64 to 1.33 µM) acids, in the MCP mouse group. BEY treatment significantly regulated the expression of inflammatory transcripts in EAE mice and upregulated neuroprotective markers such as neurexin (from 0.65- to 1.22-fold) (p < 0.05), vascular endothelial adhesion molecules (from 0.41- to 0.76-fold) and myelin-binding protein (from 0.46- to 0.89-fold) (p < 0.03). These findings suggest that BEY could be a promising clinical approach for the curative treatment of neurodegenerative diseases and could promote the use of probiotic food as medicine.
Collapse
Affiliation(s)
- Ashraf Khalifa
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Hairul Islam Mohamed Ibrahim
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
- Molecular Biology Division, Pondicherry Centre for Biological Sciences and Educational Trust, Pondicherry 605004, India
| | - Abdullah Sheikh
- Camel Research Center, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
| | - Hany Ezzat Khalil
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| |
Collapse
|
7
|
Bacillus amyloliquifaciens-Supplemented Camel Milk Suppresses Neuroinflammation of Autoimmune Encephalomyelitis in a Mouse Model by Regulating Inflammatory Markers. Nutrients 2023; 15:nu15030550. [PMID: 36771257 PMCID: PMC9921734 DOI: 10.3390/nu15030550] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Multiple sclerosis (MS), a distinct autoimmune neuroinflammatory disorder, affects millions of people worldwide, including Saudi Arabia. Changes in the gut microbiome are linked to the development of neuroinflammation via mechanisms that are not fully understood. Prebiotics and probiotics in camel milk that has been fermented have a variety of health benefits. In this study, Bacillus amyloliquefaciens-supplemented camel milk (BASY) was used to assess its preventive effect on MS symptoms in a myelin oligodendrocyte glycoprotein (MOG)-immunized C57BL6J mice model. To this end, MOG-induced experimental autoimmune encephalomyelitis (EAE) was established and the level of disease index, pathological scores, and anti-inflammatory markers of BASY-treated mice using macroscopic and microscopic examinations, qPCR and immunoblot were investigated. The results demonstrate that BASY significantly reduced the EAE disease index, increased total microbial load (2.5 fold), and improved the levels of the short-chain fatty acids propionic, butyric and caproic acids in the diseased mice group. Additionally, myeloperoxidase (MPO) proinflammatory cytokines (IL-1β, IL-6, IL-17, TNF-α) and anti-inflammatory cytokines (TGF-β) were regulated by BASY treatment. Significant suppression of MPO and VCAM levels were noticed in the BASY-treated group (from 168 to 111 µM and from 34 to 27 pg/mL, respectively), in comparison to the EAE group. BASY treatment significantly reduced the expression of inflammatory cytokines, inflammatory progression related transcripts, and inflammatory progression protein markers. In conclusion, BASY significantly reduced the symptoms of EAE mice and may be used to develop a probiotic-based diet to promote host gut health. The cumulative findings of this study confirm the significant neuroprotection of BASY in the MOG-induced mice model. They could also suggest a novel approach to the treatment of MS-associated disorders.
Collapse
|
8
|
Alvarez-Sanchez N, Dunn SE. Potential biological contributers to the sex difference in multiple sclerosis progression. Front Immunol 2023; 14:1175874. [PMID: 37122747 PMCID: PMC10140530 DOI: 10.3389/fimmu.2023.1175874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Multiple sclerosis (MS) is an immune-mediated disease that targets the myelin sheath of central nervous system (CNS) neurons leading to axon injury, neuronal death, and neurological progression. Though women are more highly susceptible to developing MS, men that develop this disease exhibit greater cognitive impairment and accumulate disability more rapidly than women. Magnetic resonance imaging and pathology studies have revealed that the greater neurological progression seen in males correlates with chronic immune activation and increased iron accumulation at the rims of chronic white matter lesions as well as more intensive whole brain and grey matter atrophy and axon loss. Studies in humans and in animal models of MS suggest that male aged microglia do not have a higher propensity for inflammation, but may become more re-active at the rim of white matter lesions as a result of the presence of pro-inflammatory T cells, greater astrocyte activation or iron release from oligodendrocytes in the males. There is also evidence that remyelination is more efficient in aged female than aged male rodents and that male neurons are more susceptible to oxidative and nitrosative stress. Both sex chromosome complement and sex hormones contribute to these sex differences in biology.
Collapse
Affiliation(s)
- Nuria Alvarez-Sanchez
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
| | - Shannon E. Dunn
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
- Department of Immunology, 1 King’s College Circle, Toronto, ON, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON, Canada
- *Correspondence: Shannon E. Dunn,
| |
Collapse
|
9
|
Volumetric assessment and longitudinal changes of subcortical structures in formalinized Beagle brains. PLoS One 2022; 17:e0261484. [PMID: 36206292 PMCID: PMC9543981 DOI: 10.1371/journal.pone.0261484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 08/02/2022] [Indexed: 11/07/2022] Open
Abstract
High field MRI is an advanced technique for diagnostic and research purposes on animal models, such as the Beagle dog. In this context, studies on neuroscience applications, e.g. aging and neuro-pathologies, are currently increasing. This led to a need for reference values, in terms of volumetric assessment, for the structures typically involved. Nowadays, several canine brain MRI atlases have been provided. However, no reports are available regarding the measurements’ reproducibility and little is known about the effect of formalin on MRI segmentation. Here, we assessed the segmentation variability of selected structures among operators (two operators segmented the same data) in a sample of 11 Beagle dogs. Then, we analyzed, for one Beagle dog, the longitudinal volumetric changes of these structures. We considered four conditions: in vivo, post mortem (after euthanasia), ex vivo (brain extracted and studied after 1 month in formalin, and after 12 months). The MRI data were collected with a 3 T scanner. Our findings suggest that the segmentation procedure was overall reproducible since only slight statistical differences were detected. In the post mortem/ ex vivo comparison, most structures showed a higher contrast, thereby leading to greater reproducibility between operators. We observed a net increase in the volume of the studied structures. This could be justified by the intrinsic relaxation time changes observed because of the formalin fixation. This led to an improvement in brain structure visualization and segmentation. To conclude, MRI-based segmentation seems to be a useful and accurate tool that allows longitudinal studies on formalin-fixed brains.
Collapse
|
10
|
Zheng Y, Dudman J, Chen JT, Mahajan KR, Herman D, Fox RJ, Ontaneda D, Trapp BD, Nakamura K. Sensitivity of T1/T2-weighted ratio in detection of cortical demyelination is similar to magnetization transfer ratio using post-mortem MRI. Mult Scler 2022; 28:198-205. [PMID: 34014144 PMCID: PMC8605037 DOI: 10.1177/13524585211014760] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Detecting cortical demyelination using magnetic resonance imaging (MRI) in multiple sclerosis (MS) remains a challenge. Magnetization transfer ratio (MTR), T1-weighted/T2-weighted ratio (T1T2R), and T2-weighted (T2w) signal are sensitive to cortical demyelination, but their accuracy is unknown. OBJECTIVES To quantify the sensitivity, specificity, and accuracy of postmortem T1T2R, MTR, and T2w in detecting cortical demyelination. METHODS In situ postmortem MRIs from 9 patients were used to measure T1T2R, MTR, and T2w along the midline of cortical gray matter and classified as normal or abnormal. MRIs were co-registered and compared to hemispheric myelin staining. The sensitivity, specificity, and accuracy of T1T2R, MTR, and T2w in detecting cortical demyelination were measured. RESULTS The mean age (standard deviation) at death was 64.7 (+/-13.7) years with a disease duration of 23.8 (+/-10.5) years. The sensitivity was 78% for MTR, 75% for T1T2R, and 63% for T2w. The specificity was 46% (T2w), 13% (T1T2R), and 29% (MTR). The accuracy was 71% (T2w), 39% (MTR), and 42% (T1T2R). There were no significant differences between different MRI measures in cortical demyelination or intracortical/subpial lesion detection. CONCLUSIONS Although somewhat sensitive, the modest specificity of conventional MRI modalities for cortical demyelination indicates that they are influenced by cortical changes other than demyelination. Improved acquisition and post-processing are needed to reliably measure cortical lesion load.
Collapse
Affiliation(s)
- Yufan Zheng
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jessica Dudman
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacqueline T Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kedar R Mahajan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA/Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Danielle Herman
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
11
|
Vanderdonckt P, Aloisi F, Comi G, de Bruyn A, Hartung HP, Huitinga I, Kuhlmann T, Lucchinetti CF, Metz I, Reynolds R, Lassmann H. OUP accepted manuscript. Brain Commun 2022; 4:fcac094. [PMID: 35480225 PMCID: PMC9039502 DOI: 10.1093/braincomms/fcac094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/04/2022] [Accepted: 04/13/2022] [Indexed: 12/05/2022] Open
Abstract
Although major progress in multiple sclerosis research has been made during the last decades, key questions related to the cause and the mechanisms of brain and spinal cord pathology remain unresolved. These cover a broad range of topics, including disease aetiology, antigenic triggers of the immune response inside and/or outside the CNS and mechanisms of inflammation, demyelination neurodegeneration and tissue repair. Most of these questions can be addressed with novel molecular technologies in the injured CNS. Access to brain and spinal cord tissue from multiple sclerosis patients is, therefore, of critical importance. High-quality tissue is provided in part by the existing brain banks. However, material from early and highly active disease stages is limited. An initiative, realized under the patronage of the European Charcot Foundation, gathered together experts from different disciplines to analyse the current state of multiple sclerosis tissues collected post-mortem or as biopsies. Here, we present an account of what material is currently available and where it can be accessed. We also provide recommendations on how tissue donation from patients in early disease stages could be potentially increased and for procedures of tissue sampling and preservation. We also suggest to create a registry of the available tissues that, depending on the source (autopsy versus biopsy), could be made accessible to clinicians and researchers.
Collapse
Affiliation(s)
| | - Francesca Aloisi
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | - Giancarlo Comi
- Centro Sclerosi Multipla Ospedale Gallarate and European Charcot Foundation, San Rafaele Scientific Institute, Milano, Italy
| | | | - Hans-Peter Hartung
- Department of Neurology UKD, Germany Medical Faculty, Heinrich Heine Universität, Düsseldorf, Germany
- Brain and Mind Center, University of Sydney, Camperdown, Australia
- Department of Neurology, University of Vienna, Wien, Austria
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Tanja Kuhlmann
- Institut für Neuropathologie, Universitätsklinikum Münster/UKM, Münster, Germany
| | | | - Imke Metz
- Institute of Neuropathology, University Medical Center, Göttingen, Germany
| | | | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Wien, Austria
- Correspondence to: Hans Lassmann Center for Brain Research Medical University of Vienna Spitalgasse 4, A-1090 Wien, Austria E-mail:
| |
Collapse
|
12
|
Positron emission tomography in multiple sclerosis - straight to the target. Nat Rev Neurol 2021; 17:663-675. [PMID: 34545219 DOI: 10.1038/s41582-021-00537-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 02/08/2023]
Abstract
Following the impressive progress in the treatment of relapsing-remitting multiple sclerosis (MS), the major challenge ahead is the development of treatments to prevent or delay the irreversible accumulation of clinical disability in progressive forms of the disease. The substrate of clinical progression is neuro-axonal degeneration, and a deep understanding of the mechanisms that underlie this process is a precondition for the development of therapies for progressive MS. PET imaging involves the use of radiolabelled compounds that bind to specific cellular and metabolic targets, thereby enabling direct in vivo measurement of several pathological processes. This approach can provide key insights into the clinical relevance of these processes and their chronological sequence during the disease course. In this Review, we focus on the contribution that PET is making to our understanding of extraneuronal and intraneuronal mechanisms that are involved in the pathogenesis of irreversible neuro-axonal damage in MS. We consider the major challenges with the use of PET in MS and the steps necessary to realize clinical benefits of the technique. In addition, we discuss the potential of emerging PET tracers and future applications of existing compounds to facilitate the identification of effective neuroprotective treatments for patients with MS.
Collapse
|
13
|
Kocsis K, Szabó N, Tóth E, Király A, Faragó P, Kincses B, Veréb D, Bozsik B, Boross K, Katona M, Bodnár P, László NG, Vécsei L, Klivényi P, Bencsik K, Kincses ZT. Two Classes of T1 Hypointense Lesions in Multiple Sclerosis With Different Clinical Relevance. Front Neurol 2021; 12:619135. [PMID: 33746876 PMCID: PMC7966518 DOI: 10.3389/fneur.2021.619135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/14/2021] [Indexed: 12/04/2022] Open
Abstract
Background: Hypointense lesions on T1-weighted images have important clinical relevance in multiple sclerosis patients. Traditionally, spin-echo (SE) sequences are used to assess these lesions (termed black holes), but Fast Spoiled Gradient-Echo (FSPGR) sequences provide an excellent alternative. Objective: To determine whether the contrast difference between T1 hypointense lesions and the surrounding normal white matter is similar on the two sequences, whether different lesion types could be identified, and whether the clinical relevance of these lesions types are different. Methods: Seventy-nine multiple sclerosis patients' lesions were manually segmented, then registered to T1 sequences. Median intensity values of lesions were identified on all sequences, then K-means clustering was applied to assess whether distinct clusters of lesions can be defined based on intensity values on SE, FSPGR, and FLAIR sequences. The standardized intensity of the lesions in each cluster was compared to the intensity of the normal appearing white matter in order to see if lesions stand out from the white matter on a given sequence. Results: 100% of lesions on FSPGR images and 69% on SE sequence in cluster #1 exceeded a standardized lesion distance of Z = 2.3 (p < 0.05). In cluster #2, 78.7% of lesions on FSPGR and only 17.7% of lesions on SE sequence were above this cutoff value, meaning that these lesions were not easily seen on SE images. Lesion count in the second cluster (lesions less identifiable on SE) significantly correlated with the Expanded Disability Status Scale (EDSS) (R: 0.30, p ≤ 0.006) and with disease duration (R: 0.33, p ≤ 0.002). Conclusion: We showed that black holes can be separated into two distinct clusters based on their intensity values on various sequences, only one of which is related to clinical parameters. This emphasizes the joint role of FSPGR and SE sequences in the monitoring of MS patients and provides insight into the role of black holes in MS.
Collapse
Affiliation(s)
- Krisztián Kocsis
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Nikoletta Szabó
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Eszter Tóth
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - András Király
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Péter Faragó
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Bálint Kincses
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Dániel Veréb
- Department of Radiology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Bence Bozsik
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Katalin Boross
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Melinda Katona
- Department of Image Processing and Computer Graphics, University of Szeged, Szeged, Hungary
| | - Péter Bodnár
- Department of Image Processing and Computer Graphics, University of Szeged, Szeged, Hungary
| | - Nyúl Gábor László
- Department of Image Processing and Computer Graphics, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,Magyar Tudományos Akadémia-Szegedi Tudományegyetem (MTA-SZTE) Neuroscience Research Group, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Krisztina Bencsik
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Zsigmond Tamás Kincses
- Department of Neurology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,Department of Radiology, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| |
Collapse
|
14
|
Peixoto-Santos JE, Velasco TR, Carlotti CG, Assirati JA, Rezende GHDSE, Kobow K, Coras R, Blümcke I, Salmon CEG, Santos ACD, Leite JP. Histological correlates of hippocampal magnetization transfer images in drug-resistant temporal lobe epilepsy patients. NEUROIMAGE-CLINICAL 2020; 28:102463. [PMID: 33395959 PMCID: PMC7586233 DOI: 10.1016/j.nicl.2020.102463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/01/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Temporal lobe epilepsy patients (TLE) often present with hippocampal atrophy, increased T2 relaxation, and reduced magnetization transfer ratio (MTR) in magnetic resonance images (MRI). The histological correlates of the reduced hippocampal MTR are so far unknown. Since MTR is dependent on the tissue's macromolecules, our aim was to evaluate the correlations between cellular populations, extracellular matrix molecules and the MTR in TLE patients. METHODS Patients with TLE (n = 26) and voluntaries (=20) were scanned in a 3 Tesla MRI scanner, and MTR images were calculated from 3DT1 sequences with magnetization pulse on resonance. Immunohistochemistry for neurons, reactive astrocytes, activated microglia, and extracellular matrix chondroitin sulfate were performed in formalin fixed, paraffin embedded tissues of TLE and autopsy controls (n = 10). Results were considered significant with adjusted p < 0.05. RESULTS Compared to the respective controls, TLE patients had reduced hippocampal MTR, increased reactive astrocytes and activated microglia, increased extracellular chondroitin sulfate, and reduced neuron density, compares to controls. MTR correlated positively with neuron density in CA3 and with chondroitin sulfate in CA3 and CA1. Multiple linear regressions reinforced the correlations between chondroitin sulfate and MTR. SIGNIFICANCE Our data indicate that extracellular matrix molecules are the most significant histological correlates of magnetization transfer ratio in the hippocampus of TLE patients.
Collapse
Affiliation(s)
- Jose Eduardo Peixoto-Santos
- Department of Neurology and Neurosurgery, Paulista Medical School, UNIFESP, Sao Paulo, Brazil; Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Tonicarlo R Velasco
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Carlos Gilberto Carlotti
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Joao Alberto Assirati
- Department of Surgery and Anatomy, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Gustavo Henrique de Souza E Rezende
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Katja Kobow
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany.
| | - Roland Coras
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany.
| | - Ingmar Blümcke
- Department of Neuropathology, University Hospital Erlangen, Erlangen, Germany.
| | - Carlos Ernesto Garrido Salmon
- Department of Physics and Mathematics, Faculty of Philosophy, Science and Languages of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Antonio Carlos Dos Santos
- Department of Internal Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| | - Joao Pereira Leite
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.
| |
Collapse
|
15
|
Moccia M, van de Pavert S, Eshaghi A, Haider L, Pichat J, Yiannakas M, Ourselin S, Wang Y, Wheeler-Kingshott C, Thompson A, Barkhof F, Ciccarelli O. Pathologic correlates of the magnetization transfer ratio in multiple sclerosis. Neurology 2020; 95:e2965-e2976. [PMID: 32938787 DOI: 10.1212/wnl.0000000000010909] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/22/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify pathologic correlates of magnetization transfer ratio (MTR) in multiple sclerosis (MS) in an MRI-pathology study. METHODS We acquired MTR maps at 3T from 16 fixed MS brains and 4 controls, and immunostained 100 tissue blocks for neuronal neurofilaments, myelin (SMI94), tissue macrophages (CD68), microglia (IBA1), B-lymphocytes, T-lymphocytes, cytotoxic T-lymphocytes, astrocytes (glial fibrillary acidic protein), and mitochondrial damage (COX4, VDAC). We defined regions of interest in lesions, normal-appearing white matter (NAWM), and cortical normal-appearing gray matter (NAGM). Associations between MTR and immunostaining intensities were explored using linear mixed-effects models (with cassettes nested within patients) and interaction terms (for differences between regions of interest and between cases and controls); a multivariate linear mixed-effects model identified the best pathologic correlates of MTR. RESULTS MTR was the lowest in white matter (WM) lesions (23.4 ± 9.4%) and the highest in NAWM (38.1 ± 8.7%). In MS brains, lower MTR was associated with lower immunostaining intensity for myelin (coefficient 0.31; 95% confidence interval [CI] 0.07-0.55), macrophages (coefficient 0.03; 95% CI 0.01-0.07), and astrocytes (coefficient 0.51; 95% CI 0.02-1.00), and with greater mitochondrial damage (coefficient 0.31; 95% CI 0.07-0.55). Based on interaction terms, MTR was more strongly associated with myelin in WM (coefficient 1.58; 95% CI 1.09-2.08) and gray matter (GM) lesions (coefficient 0.66; 95% CI 0.13-1.20), and with macrophages (coefficient 1.40; 95% CI 0.56-2.25), astrocytes (coefficient 2.66; 95% CI 1.31-4.01), and mitochondrial damage (coefficient -12.59; 95% CI -23.16 to -2.02) in MS brains than controls. In the multivariate model, myelin immunostaining intensity was the best correlate of MTR (coefficient 0.31; 95% CI 0.09-0.52; p = 0.004). CONCLUSIONS Myelin was the strongest correlate of MTR, especially in WM and cortical GM lesions, but additional correlates should be kept in mind when designing and interpreting MTR observational and experimental studies in MS.
Collapse
Affiliation(s)
- Marcello Moccia
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Steven van de Pavert
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Arman Eshaghi
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Lukas Haider
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Jonas Pichat
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Marios Yiannakas
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Sebastien Ourselin
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Yi Wang
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Claudia Wheeler-Kingshott
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Alan Thompson
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Frederik Barkhof
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK
| | - Olga Ciccarelli
- From the Department of Neuroinflammation, Queen Square MS Centre, NMR Research Unit, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences (M.M., S.v.d.P., A.E., L.H., M.Y., Y.W., C.W.-K., A.T., F.B., O.C.), Centre for Medical Image Computing, Department of Medical Physics and Bioengineering (J.P., S.O.), and Translational Imaging Group, UCL Institute of Healthcare Engineering (F.B.), University College London, UK; Multiple Sclerosis Clinical Care and Research Centre, Department of Neurosciences (M.M.), Federico II University, Naples, Italy; Department of Radiology and Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands; and National Institute for Health Research University College London Hospitals Biomedical Research Centre (A.T., F.B., O.C.), UK.
| |
Collapse
|
16
|
Mahajan KR, Nakamura K, Cohen JA, Trapp BD, Ontaneda D. Intrinsic and Extrinsic Mechanisms of Thalamic Pathology in Multiple Sclerosis. Ann Neurol 2020; 88:81-92. [PMID: 32286701 PMCID: PMC8291218 DOI: 10.1002/ana.25743] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Thalamic atrophy is among the earliest brain changes detected in patients with multiple sclerosis (MS) and the degree of thalamic atrophy is a strong predictor of disability progression. The causes of thalamic atrophy are not fully understood. Here, we investigate the contributions of thalamic demyelinated lesions, thalamic neuronal loss, and cerebral white matter (WM) lesions to thalamic volume. METHODS We used postmortem in situ magnetic resonance imaging (MRI) scans of 95 subjects with MS to correlate thalamic lesion volumes with global MRI metrics. We histologically characterized thalamic demyelination patterns and compared neuronal loss and neuritic pathology in the thalami with the extremes of volume. RESULTS Grossly apparent thalamic discolorations in cm-thick brain slices were T2/fluid-attenuated inversion recovery (FLAIR) hyperintense, T1-hypointense, and appeared as perivascular demyelinated lesions with dystrophic neurons/axons. Subependymal demyelinated lesions with axonal loss and microglial/macrophage activation were also observed. The 12 subjects with the least thalamic volume had a 17.6% reduction of median neuronal density in the dorsomedial/ventrolateral and pulvinar nuclei compared with the 14 subjects with the greatest thalamic volume (p = 0.03). After correcting for age, disease duration, sex, and T2 lesion volume, the total (p = 0.20), ovoid (p = 0.31), or subependymal (p = 0.44) MRI thalamic lesion volumes correlated with thalamic volume. Thalamic volume correlated with cerebral T2 lesion volume (Spearman's rho = -0.65, p < 0.001; p < 0.0001 after correcting for age, disease duration, and sex). INTERPRETATION Our findings suggest the degeneration of efferent/afferent thalamic projections and/or a neurodegenerative process as greater contributors to thalamic atrophy than thalamic demyelinating lesions. ANN NEUROL 2020 ANN NEUROL 2020;88:81-92.
Collapse
Affiliation(s)
- Kedar R. Mahajan
- Mellen Center for MS Treatment and Research, Neurologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Neurologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jeffrey A. Cohen
- Mellen Center for MS Treatment and Research, Neurologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bruce D. Trapp
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Daniel Ontaneda
- Mellen Center for MS Treatment and Research, Neurologic Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
17
|
Wildner P, Zydorczak E, Oset M, Siger M, Wilczyński M, Stasiołek M, Matysiak M. The Role of Optical Coherence Tomography in Differential Diagnosis of Multiple Sclerosis and Autoimmune Connective Tissue Diseases with CNS Involvement. J Clin Med 2020; 9:jcm9051565. [PMID: 32455833 PMCID: PMC7290953 DOI: 10.3390/jcm9051565] [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: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 11/28/2022] Open
Abstract
The purpose of this study was to examine whether application of optical coherence tomography (OCT) measurements can provide a useful biomarker for distinguishing central nervous system (CNS) involvement in autoimmune connective tissue diseases (CTD) from multiple sclerosis (MS). An observational study included non-optic neuritis eyes of 121 individuals: 59 patients with MS, 30 patients with CNS involvement in CTD, and 32 healthy controls. OCT examination was performed in all subjects to measure retinal nerve fiber layer (RNFL) thickness, ganglion cell complex (GCC) thickness, ganglion cell layer-inner plexiform layer (GCIPL) thickness, and volume of the macula. There was a significant group effect with regard to superior optic disc RNFL, macular RNFL, GCC, and GCIPL thickness, and macular volume. Post-hoc analysis revealed that MS patients have significantly smaller macular volume and thinner superior optic disc RNFL, macular RNFL, GCC, and GCIPL compared to healthy controls. CTD patients have significantly smaller superior optic disc RNFL, GCIPL, and GCC thickness compared to healthy controls. However, no significant group differences were observed between the patient groups (MS vs. CTD) on any outcome. Although a prominent retinal thinning may be a useful biomarker in MS patients, in a general population of individuals with a confirmed CNS involvement the use of OCT is not specific enough to discriminate between MS and autoimmune CTD.
Collapse
Affiliation(s)
- Paula Wildner
- Department of Neurology, Medical University of Lodz, 90-414 Lodz, Poland; (P.W.); (M.O.); (M.S.); (M.M.)
| | - Ewa Zydorczak
- Department of Ophthalmology, Medical University of Lodz, 90-414 Lodz, Poland; (E.Z.); (M.W.)
| | - Magdalena Oset
- Department of Neurology, Medical University of Lodz, 90-414 Lodz, Poland; (P.W.); (M.O.); (M.S.); (M.M.)
| | - Małgorzata Siger
- Department of Neurology, Medical University of Lodz, 90-414 Lodz, Poland; (P.W.); (M.O.); (M.S.); (M.M.)
| | - Michał Wilczyński
- Department of Ophthalmology, Medical University of Lodz, 90-414 Lodz, Poland; (E.Z.); (M.W.)
| | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, 90-414 Lodz, Poland; (P.W.); (M.O.); (M.S.); (M.M.)
- Correspondence:
| | - Mariola Matysiak
- Department of Neurology, Medical University of Lodz, 90-414 Lodz, Poland; (P.W.); (M.O.); (M.S.); (M.M.)
| |
Collapse
|
18
|
Deep learning segmentation of orbital fat to calibrate conventional MRI for longitudinal studies. Neuroimage 2020; 208:116442. [DOI: 10.1016/j.neuroimage.2019.116442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/04/2019] [Accepted: 12/03/2019] [Indexed: 01/21/2023] Open
|
19
|
Wildner P, Stasiołek M, Matysiak M. Differential diagnosis of multiple sclerosis and other inflammatory CNS diseases. Mult Scler Relat Disord 2020; 37:101452. [DOI: 10.1016/j.msard.2019.101452] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/11/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022]
|
20
|
Makhija EP, Espinosa-Hoyos D, Jagielska A, Van Vliet KJ. Mechanical regulation of oligodendrocyte biology. Neurosci Lett 2019; 717:134673. [PMID: 31838017 DOI: 10.1016/j.neulet.2019.134673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 11/25/2019] [Accepted: 12/01/2019] [Indexed: 12/27/2022]
Abstract
Oligodendrocytes (OL) are a subset of glial cells in the central nervous system (CNS) comprising the brain and spinal cord. The CNS environment is defined by complex biochemical and biophysical cues during development and response to injury or disease. In the last decade, significant progress has been made in understanding some of the key biophysical factors in the CNS that modulate OL biology, including their key role in myelination of neurons. Taken together, those studies offer translational implications for remyelination therapies, pharmacological research, identification of novel drug targets, and improvements in methods to generate human oligodendrocyte progenitor cells (OPCs) and OLs from donor stem cells in vitro. This review summarizes current knowledge of how various physical and mechanical cues affect OL biology and its implications for disease, therapeutic approaches, and generation of human OPCs and OLs.
Collapse
Affiliation(s)
- Ekta P Makhija
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore
| | - Daniela Espinosa-Hoyos
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Anna Jagielska
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| | - Krystyn J Van Vliet
- BioSystems & Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, Singapore 138602; Critical Analytics for Manufacturing Personalized-Medicine (CAMP) Interdisciplinary Research Group, Singapore-MIT Alliance for Research & Technology (SMART) CREATE, 138602, Singapore; Department of Materials Science & Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| |
Collapse
|
21
|
Magnetic Resonance Imaging as a Biomarker in Rodent Peripheral Nerve Injury Models Reveals an Age-Related Impairment of Nerve Regeneration. Sci Rep 2019; 9:13508. [PMID: 31534149 PMCID: PMC6751200 DOI: 10.1038/s41598-019-49850-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/24/2019] [Indexed: 12/12/2022] Open
Abstract
Assessment of myelin integrity in peripheral nerve injuries and pathologies has largely been limited to post-mortem analysis owing to the difficulty in obtaining biopsies without affecting nerve function. This is further encumbered by the small size of the tissue and its location. Therefore, the development of robust, non-invasive methods is highly attractive. In this study, we used magnetic resonance imaging (MRI) techniques, including magnetization transfer ratio (MTR), to longitudinally and non-invasively characterize both the sciatic nerve crush and lysolecithin (LCP) demyelination models of peripheral nerve injury in rodents. Electrophysiological, gene expression and histological assessments complemented the extensive MRI analyses in young and aged animals. In the nerve crush model, MTR analysis indicated a slower recovery in regions distal to the site of injury in aged animals, as well as incomplete recovery at six weeks post-crush when analyzing across the entire nerve surface. Similar regional impairments were also found in the LCP demyelination model. This research underlines the power of MTR for the study of peripheral nerve injury in small tissues such as the sciatic nerve of rodents and contributes new knowledge to the effect of aging on recovery after injury. A particular advantage of the approach is the translational potential to human neuropathies.
Collapse
|
22
|
Cadavid D, Mellion M, Hupperts R, Edwards KR, Calabresi PA, Drulović J, Giovannoni G, Hartung HP, Arnold DL, Fisher E, Rudick R, Mi S, Chai Y, Li J, Zhang Y, Cheng W, Xu L, Zhu B, Green SM, Chang I, Deykin A, Sheikh SI. Safety and efficacy of opicinumab in patients with relapsing multiple sclerosis (SYNERGY): a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 2019; 18:845-856. [PMID: 31285147 DOI: 10.1016/s1474-4422(19)30137-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/26/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Opicinumab is a human monoclonal antibody against LINGO-1, an inhibitor of oligodendrocyte differentiation and axonal regeneration. Previous findings suggested that opicinumab treatment might enhance remyelination in patients with CNS demyelinating diseases. We aimed to assess the safety and efficacy of opicinumab in patients with relapsing multiple sclerosis. METHODS We did a randomised, double-blind, placebo-controlled, dose-ranging, phase 2 study (SYNERGY) at 72 sites in 12 countries. Participants (aged 18-58 years) with relapsing multiple sclerosis (relapsing-remitting multiple sclerosis and secondary progressive multiple sclerosis with relapses) were randomised in a 1:2:2:2:2 ratio by an interactive voice and web response system to opicinumab 3 mg/kg, 10 mg/kg, 30 mg/kg, or 100 mg/kg, or placebo. An identical volume of study drug was administered intravenously once every 4 weeks. All participants self-administered intramuscular interferon beta-1a as background anti-inflammatory treatment once a week. The primary endpoint was the percentage of participants achieving confirmed disability improvement over 72 weeks, which was a multicomponent endpoint measured by the Expanded Disability Status Scale, the Timed 25-Foot Walk, the Nine-Hole Peg Test, and the 3 s Paced Auditory Serial Addition Test. The primary endpoint was analysed under intention-to-treat principles. This study is registered at ClinicalTrials.gov, number NCT01864148. FINDINGS Between Aug 13, 2013, and July 31, 2014, 419 patients were enrolled and randomly assigned either placebo (n=93) or opicinumab 3 mg/kg (n=45), 10 mg/kg (n=95), 30 mg/kg (n=94; one patient did not receive the assigned treatment), or 100 mg/kg (n=92). The last patient visit was on March 29, 2016. Confirmed disability improvement over 72 weeks was seen in 45 (49%) of 91 patients assigned to placebo, 21 (47%) of 45 assigned to opicinumab 3 mg/kg, 59 (63%) of 94 assigned to opicinumab 10 mg/kg, 59 (65%) of 91 assigned to opicinumab 30 mg/kg, and 36 (40%) of 91 assigned to opicinumab 100 mg/kg. A linear dose-response in the probability of confirmed disability improvement was not seen (linear trend test p=0·89). Adverse events occurred in 79 (85%) patients assigned placebo and in 275 (85%) assigned any dose of opicinumab. The most common adverse events of any grade in patients assigned any dose of opicinumab included influenza-like illness (140 [43%] with any dose of opicinumab vs 37 [40%] with placebo), multiple sclerosis relapses (117 [36%] vs 30 [32%]), and headache (51 [16%] vs 23 [25%]). Serious adverse events reported as related to treatment were urinary tract infection in one (1%) participant in the the placebo group, suicidal ideation and intentional overdose in one (1%) participant in the 30 mg/kg opicinumab group, bipolar disorder in one (1%) participant in the 100 mg/kg opicinumab group, and hypersensitivity in four (4%) participants in the 100 mg/kg opicinumab group. One patient in the opicinumab 30 mg/kg group died during the study due to a traffic accident, which was not considered related to study treatment. INTERPRETATION Our findings did not show a significant dose-linear improvement in disability compared with placebo in patients with relapsing multiple sclerosis. Further studies are needed to investigate whether some subpopulations identified in the study might benefit from opicinumab treatment at an optimum dose. FUNDING Biogen.
Collapse
Affiliation(s)
| | | | | | - Keith R Edwards
- Multiple Sclerosis Center of Northeastern New York, Latham, NY, USA
| | | | - Jelena Drulović
- Clinic of Neurology, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Gavin Giovannoni
- Barts & The London School of Medicine & Dentistry, Queen Mary University, London, UK
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Douglas L Arnold
- Montreal Neurological Institute, Montreal, QC, Canada; NeuroRx Research, Montreal, QC, Canada
| | | | | | - Sha Mi
- Biogen, Cambridge, MA, USA
| | | | - Jie Li
- Biogen, Cambridge, MA, USA
| | | | | | - Lei Xu
- Biogen, Cambridge, MA, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Schmierer K, Miquel ME. Magnetic resonance imaging correlates of neuro-axonal pathology in the MS spinal cord. Brain Pathol 2019; 28:765-772. [PMID: 30375114 DOI: 10.1111/bpa.12648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 07/18/2018] [Indexed: 12/21/2022] Open
Abstract
In people with multiple sclerosis (MS), the spinal cord is the structure most commonly affected by clinically detectable pathology at presentation, and a key part of the central nervous system involved in chronic disease deterioration. Indices, such as the spinal cord cross-sectional area at the level C2 have been developed as tools to predict future disability, and-by inference-axonal loss. However, this and other histo-pathological correlates of spinal cord magnetic resonance imaging (MRI) changes in MS remain incompletely understood. In recent years, there has been a surge of interest in developing quantitative MRI tools to measure specific tissue features, including axonal density, myelin content, neurite density, and orientation, among others, with an emphasis on the spinal cord. Quantitative MRI techniques including T1 and T2 , magnetization transfer and a number of diffusion-derived indices have all been applied to MS spinal cord. Particularly diffusion-based MRI techniques combined with microscopic resolution achievable using high magnetic field scanners enable a new level of anatomical detail and quantification of indices that are clinically meaningful.
Collapse
Affiliation(s)
- Klaus Schmierer
- Queen Mary University of London, Barts and The London School of Medicine & Dentistry, Blizard Institute (Neuroscience), London, UK.,Barts Health NHS Trust, Clinical Board Medicine (Neuroscience), The Royal London Hospital, London, UK
| | - Marc E Miquel
- Barts Health NHS Trust, Clinical Physics, London, UK
| |
Collapse
|
24
|
Göçmen R. The Relevance of Neuroimaging Findings to Physical Disability in Multiple Sclerosis. ACTA ACUST UNITED AC 2019; 55:S31-S36. [PMID: 30692852 DOI: 10.29399/npa.23409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system and one of the leading causes of disability in young adults. While some patients with MS have a benign course in which they develop limited disability even after many years, other patients have a rapidly progressive course resulting in severe disability. However, the progression of the disease, particularly disability, is currently a predictable course with neuroimaging features to some extend. Magnetic resonance imaging (MRI) is not only the main diagnostic tool but also used to monitor response to therapies, thanks to its high sensitivity and ability to identify clinically silent lesions. This report presents a literature review which examines in detail the relationship between MRI findings and disability.
Collapse
Affiliation(s)
- Rahşan Göçmen
- Hacettepe University School of Medicine, Department of Radiology, Ankara, Turkey
| |
Collapse
|
25
|
O'Muircheartaigh J, Vavasour I, Ljungberg E, Li DKB, Rauscher A, Levesque V, Garren H, Clayton D, Tam R, Traboulsee A, Kolind S. Quantitative neuroimaging measures of myelin in the healthy brain and in multiple sclerosis. Hum Brain Mapp 2019; 40:2104-2116. [PMID: 30648315 PMCID: PMC6590140 DOI: 10.1002/hbm.24510] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 12/28/2018] [Accepted: 01/02/2019] [Indexed: 12/25/2022] Open
Abstract
Quantitative magnetic resonance imaging (MRI) techniques have been developed as imaging biomarkers, aiming to improve the specificity of MRI to underlying pathology compared to conventional weighted MRI. For assessing the integrity of white matter (WM), myelin, in particular, several techniques have been proposed and investigated individually. However, comparisons between these methods are lacking. In this study, we compared four established myelin‐sensitive MRI techniques in 56 patients with relapsing–remitting multiple sclerosis (MS) and 38 healthy controls. We used T2‐relaxation with combined GRadient And Spin Echoes (GRASE) to measure myelin water fraction (MWF‐G), multi‐component driven equilibrium single pulse observation of T1 and T2 (mcDESPOT) to measure MWF‐D, magnetization‐transfer imaging to measure magnetization‐transfer ratio (MTR), and T1 relaxation to measure quantitative T1 (qT1). Using voxelwise Spearman correlations, we tested the correspondence of methods throughout the brain. All four methods showed associations that varied across tissue types; the highest correlations were found between MWF‐D and qT1 (median ρ across tissue classes 0.8) and MWF‐G and MWF‐D (median ρ = 0.59). In eight WM tracts, all measures showed differences (p < 0.05) between MS normal‐appearing WM and healthy control WM, with qT1 showing the highest number of different regions (8), followed by MWF‐D and MTR (6), and MWF‐G (n = 4). Comparing the methods in terms of their statistical sensitivity to MS lesions in WM, MWF‐D demonstrated the best accuracy (p < 0.05, after multiple comparison correction). To aid future power analysis, we provide the average and standard deviation volumes of the four techniques, estimated from the healthy control sample.
Collapse
Affiliation(s)
- Jonathan O'Muircheartaigh
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, United Kingdom.,Centre for the Developing Brain, Department of Perinatal Imaging and Health, St. Thomas' Hospital, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Irene Vavasour
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emil Ljungberg
- Department of Neuroimaging, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, United Kingdom
| | - David K B Li
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada.,MS/MRI Research Group, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexander Rauscher
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | - Roger Tam
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada.,MS/MRI Research Group, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anthony Traboulsee
- MS/MRI Research Group, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.,Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shannon Kolind
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada.,MS/MRI Research Group, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Physics and Astronomy, University of British Columbia, Vancouver, British Columbia, Canada.,Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Nakamura Y, Gaetano L, Matsushita T, Anna A, Sprenger T, Radue EW, Wuerfel J, Bauer L, Amann M, Shinoda K, Isobe N, Yamasaki R, Saida T, Kappos L, Kira JI. A comparison of brain magnetic resonance imaging lesions in multiple sclerosis by race with reference to disability progression. J Neuroinflammation 2018; 15:255. [PMID: 30185189 PMCID: PMC6125988 DOI: 10.1186/s12974-018-1295-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 08/28/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We compared the magnetic resonance imaging (MRI) features between Japanese and Caucasian patients with multiple sclerosis (MS), and identified the relationships between MRI features and disability. METHODS From the baseline data of phase II fingolimod trials, 95 Japanese and 246 Caucasian relapsing-remitting MS patients were enrolled. The number, volume, and distribution of brain MRI lesions were evaluated using T2-weighted (T2W) images. Cross-sectional total normalized brain volume (NBV), normalized cortical gray matter volume, normalized deep gray matter volume (NDGMV), normalized white matter volume (NWMV), and normalized thalamic volume were measured. RESULTS Japanese patients had significantly lower Expanded Disability Status Scale (EDSS) scores than Caucasian patients (mean 2.0 vs. 2.3, p = 0.008), despite a similar disease duration. Japanese patients showed a trend towards fewer T2W-lesions (median 50 vs. 65, p = 0.08) and significantly lower frequencies of cerebellar and parietal lobe lesions (p = 0.02 for both) than Caucasian patients. There were no differences in T2W-lesion volume between races, whereas Japanese patients had a significantly larger T2W-lesion volume per lesion compared with Caucasian patients (median 140 mm3 vs. 85 mm3, p < 0.0001). T2W-lesion volumes were positively correlated with EDSS scores in Japanese patients (p < 0.0001). In both races, NBV, normalized cortical gray matter volume, NDGMV, and thalamic volume were negatively correlated with disease duration and EDSS scores (p < 0.01 for all). NWMV was negatively correlated with disease duration and EDSS scores only in Caucasian patients (p = 0.03 and p = 0.004, respectively). NBV, NDGMV, NWMV, and thalamic volume were consistently smaller in Japanese compared with Caucasian patients throughout the entire examined disease duration (p = 0.046, p = 0.01, p = 0.005, and p = 0.04, respectively). Japanese patients had a significantly faster reduction in NDGMV (p = 0.001), particularly for thalamic volume (p = 0.001), with disease duration compared with Caucasian patients. CONCLUSIONS Gray matter atrophy is a common denominator for disability in Japanese and Caucasian patients. Additional contributory factors for disability include T2W-lesion volume in Japanese patients and white matter atrophy in Caucasian patients. Less frequent parietal and cerebellar involvement with fewer T2W-lesions may underlie milder disability in Japanese patients.
Collapse
Affiliation(s)
- Yuri Nakamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Laura Gaetano
- Medical Image Analysis Center (MIAC AG), Marktgasse 8, 4051, Basel, Switzerland.,Neurology and Department of Biomedicine, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Altermatt Anna
- Medical Image Analysis Center (MIAC AG), Marktgasse 8, 4051, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Marktgasse 8, 4051, Basel, Switzerland
| | - Till Sprenger
- DKD Helios Klinik Wiesbaden, Aukammallee 33, 65191, Wiesbaden, Germany
| | - Ernst-Wilhelm Radue
- Biomedical Research and Education GmbH, Mittlere Strasse 91, 4031, Basel, Switzerland
| | - Jens Wuerfel
- Medical Image Analysis Center (MIAC AG), Marktgasse 8, 4051, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Marktgasse 8, 4051, Basel, Switzerland
| | - Lorena Bauer
- Medical Image Analysis Center (MIAC AG), Marktgasse 8, 4051, Basel, Switzerland.,Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Michael Amann
- Medical Image Analysis Center (MIAC AG), Marktgasse 8, 4051, Basel, Switzerland.,Neurology and Department of Biomedicine, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland.,Division of Diagnostic and Interventional Neuroradiology, Department of Radiology, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Koji Shinoda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Noriko Isobe
- Department of Neurological Therapeutics, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takahiko Saida
- Institute of Neurotherapeutics, 16-1 Nishinokyoukasugachou, Nakagyo-ku, Kyoto, 604-8453, Japan.,Department of Neurology, Kyoto Min-Iren-Central Hospital, 16-1 Nishinokyoukasugachou, Nakagyo-ku, Kyoto, 604-8453, Japan
| | - Ludwig Kappos
- Neurology and Department of Biomedicine, University Hospital Basel, Spitalstrasse 21, 4031, Basel, Switzerland
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
27
|
Xiang B, Wen J, Cross AH, Yablonskiy DA. Single scan quantitative gradient recalled echo MRI for evaluation of tissue damage in lesions and normal appearing gray and white matter in multiple sclerosis. J Magn Reson Imaging 2018; 49:487-498. [PMID: 30155934 DOI: 10.1002/jmri.26218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/22/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic disease affecting the human central nervous system (CNS) and leading to neurologic disability. Although conventional MRI techniques can readily detect focal white matter (WM) lesions, it remains challenging to quantify tissue damage in normal-appearing gray matter (GM) and WM. PURPOSE To demonstrate that a new MRI biomarker, R2t*, can provide quantitative analysis of tissue damage across the brain in MS patients in a single scan. STUDY TYPE Prospective. SUBJECTS Forty-four MS patients and 19 healthy controls (HC). FIELD STRENGTH/SEQUENCE 3T, quantitative gradient-recalled-echo (qGRE), Magnetization-prepared rapid gradient-echo, fluid-attenuated inversion recovery. ASSESSMENT Severity of tissue damage was assessed by reduced R2t*. Tissue atrophy was assessed by cortical thickness and cervical spinal cord cross-sectional area (CSA). Multiple Sclerosis Functional Composite was used for clinical assessment. RESULTS R2t* in cortical GM was more sensitive to MS damage than cortical atrophy. Using more than two standard deviations (SD) reduction versus age-matched HC as the cutoff, 48% of MS patients showed lower R2t*, versus only 9% with lower cortical thickness. Significant correlations between severities of tissue injury were identified among 1) upper cervical cord and several cortical regions, including motor cortex (P < 0.001), and 2) adjacent regions of GM and subcortical WM (P < 0.001). R2t*-defined tissue cellular damage in cortical GM was greater relative to adjacent WM. Reductions in cortical R2t* correlated with cognitive impairment (P < 0.01). Motor-related clinical signs correlated most with cervical cord CSA (P < 0.001). DATA CONCLUSION Reductions in R2t* within cortical GM was more sensitive to tissue damage than atrophy, potentially allowing a reduced sample size in clinical trials. R2t* together with structural morphometry suggested topographic patterns of regions showing correlated tissue damage throughout the brain and the cervical spinal cord of MS patients. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 3 J. Magn. Reson. Imaging 2019;49:487-498.
Collapse
Affiliation(s)
- Biao Xiang
- Department of Chemistry, Washington University, St. Louis, Missouri, USA
| | - Jie Wen
- Department of Radiology, Washington University, St. Louis, Missouri, USA
| | - Anne H Cross
- Department of Neurology, Washington University, St. Louis, Missouri, USA
| | | |
Collapse
|
28
|
Trapp BD, Vignos M, Dudman J, Chang A, Fisher E, Staugaitis SM, Battapady H, Mork S, Ontaneda D, Jones SE, Fox RJ, Chen J, Nakamura K, Rudick RA. Cortical neuronal densities and cerebral white matter demyelination in multiple sclerosis: a retrospective study. Lancet Neurol 2018; 17:870-884. [PMID: 30143361 DOI: 10.1016/s1474-4422(18)30245-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 05/18/2018] [Accepted: 06/11/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Demyelination of cerebral white matter is thought to drive neuronal degeneration and permanent neurological disability in individuals with multiple sclerosis. Findings from brain MRI studies, however, support the possibility that demyelination and neuronal degeneration can occur independently. We aimed to establish whether post-mortem brains from patients with multiple sclerosis show pathological evidence of cortical neuronal loss that is independent of cerebral white-matter demyelination. METHODS Brains and spinal cords were removed at autopsy from patients, who had died with multiple sclerosis, at the Cleveland Clinic in Cleveland, OH, USA. Visual examination of centimetre-thick slices of cerebral hemispheres was done to identify brains without areas of cerebral white-matter discoloration that were indicative of demyelinated lesions (referred to as myelocortical multiple sclerosis) and brains that had cerebral white-matter discolorations or demyelinated lesions (referred to as typical multiple sclerosis). These individuals with myelocortical multiple sclerosis were matched by age, sex, MRI protocol, multiple sclerosis disease subtype, disease duration, and Expanded Disability Status Scale, with individuals with typical multiple sclerosis. Demyelinated lesion area in tissue sections of cerebral white matter, spinal cord, and cerebral cortex from individuals classed as having myelocortical and typical multiple sclerosis were compared using myelin protein immunocytochemistry. Neuronal densities in cortical layers III, V, and VI from five cortical regions not directly connected to spinal cord (cingulate gyrus and inferior frontal cortex, superior temporal cortex, and superior insular cortex and inferior insular cortex) were also compared between the two groups and with aged-matched post-mortem brains from individuals without evidence of neurological disease. FINDINGS Brains and spinal cords were collected from 100 deceased patients between May, 1998, and November, 2012, and this retrospective study was done between Sept 6, 2011, and Feb 2, 2018. 12 individuals were identified as having myelocortical multiple sclerosis and were compared with 12 individuals identified as having typical multiple sclerosis. Demyelinated lesions were detected in spinal cord and cerebral cortex, but not in cerebral white matter, of people with myelocortical multiple sclerosis. Cortical demyelinated lesion area was similar between myelocortical and typical multiple sclerosis (median 4·45% [IQR 2·54-10·81] in myelocortical vs 9·74% [1·35-19·50] in typical multiple sclerosis; p=0·5512). Spinal cord demyelinated area was significantly greater in typical than in myelocortical multiple sclerosis (median 3·81% [IQR 1·72-7·42] in myelocortical vs 13·81% [6·51-29·01] in typical multiple sclerosis; p=0·0083). Despite the lack of cerebral white-matter demyelination in myelocortical multiple sclerosis, mean cortical neuronal densities were significantly decreased compared with control brains (349·8 neurons per mm2 [SD 51·9] in myelocortical multiple sclerosis vs 419·0 [43·6] in controls in layer III [p=0·0104]; 355·6 [46·5] vs 454·2 [48·3] in layer V [p=0·0006]; 366·6 [50·9] vs 458·3 [48·4] in layer VI [p=0·0049]). By contrast, mean cortical neuronal densities were decreased in typical multiple sclerosis brains compared with those from controls in layer V (392·5 [59·0] vs 454·2 [48·3]; p=0·0182) but not layers III and VI. INTERPRETATION We propose that myelocortical multiple sclerosis is a subtype of multiple sclerosis that is characterised by demyelination of spinal cord and cerebral cortex but not of cerebral white matter. Cortical neuronal loss is not accompanied by cerebral white-matter demyelination and can be an independent pathological event in myelocortical multiple sclerosis. Compared with control brains, cortical neuronal loss was greater in myelocortical multiple sclerosis cortex than in typical multiple sclerosis cortex. The molecular mechanisms of primary neuronal degeneration and axonal pathology in myelocortical multiple sclerosis should be investigated in future studies. FUNDING US National Institutes of Health and National Multiple Sclerosis Society.
Collapse
Affiliation(s)
- Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Megan Vignos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Jessica Dudman
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ansi Chang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elizabeth Fisher
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Susan M Staugaitis
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Pathology, Pathology and Laboratory, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Sverre Mork
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Gade Lab for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Daniel Ontaneda
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Robert J Fox
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jacqueline Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kunio Nakamura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Richard A Rudick
- Medicine Institute, Mellen Center for Treatment and Research in Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
29
|
Lemus HN, Warrington AE, Rodriguez M. Multiple Sclerosis: Mechanisms of Disease and Strategies for Myelin and Axonal Repair. Neurol Clin 2018; 36:1-11. [PMID: 29157392 PMCID: PMC7125639 DOI: 10.1016/j.ncl.2017.08.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Hernan Nicolas Lemus
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Arthur E Warrington
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| | - Moses Rodriguez
- Department of Neurology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| |
Collapse
|
30
|
Abstract
Since its technical development in the early 1980s, magnetic resonance imaging (MRI) has quickly been adopted as an essential tool in supporting the diagnosis, longitudinal monitoring, evaluation of therapeutic response, and scientific investigations in multiple sclerosis (MS). The clinical usage of MRI has increased in parallel with technical innovations in the technique itself; the widespread adoption of clinically routine MRI at 1.5T has allowed sensitive qualitative and quantitative assessments of macroscopic central nervous system (CNS) inflammatory demyelinating lesions and tissue atrophy. However, conventional MRI lesion measures lack specificity for the underlying MS pathology and only weakly correlate with clinical status. Higher field strength units and newer, advanced MRI techniques offer increased sensitivity and specificity in the detection of disease activity and disease severity. This review summarizes the current status and future prospects regarding the role of MRI in the characterization of MS-related brain and spinal cord involvement.
Collapse
Affiliation(s)
- Christopher C Hemond
- Laboratory for Neuroimaging Research, Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Departments of Neurology and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Rohit Bakshi
- Laboratory for Neuroimaging Research, Partners Multiple Sclerosis Center, Ann Romney Center for Neurologic Diseases, Departments of Neurology and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
31
|
Petrova N, Carassiti D, Altmann DR, Baker D, Schmierer K. Axonal loss in the multiple sclerosis spinal cord revisited. Brain Pathol 2018; 28:334-348. [PMID: 28401686 PMCID: PMC8028682 DOI: 10.1111/bpa.12516] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 04/05/2017] [Indexed: 01/06/2023] Open
Abstract
Preventing chronic disease deterioration is an unmet need in people with multiple sclerosis, where axonal loss is considered a key substrate of disability. Clinically, chronic multiple sclerosis often presents as progressive myelopathy. Spinal cord cross-sectional area (CSA) assessed using MRI predicts increasing disability and has, by inference, been proposed as an indirect index of axonal degeneration. However, the association between CSA and axonal loss, and their correlation with demyelination, have never been systematically investigated using human post mortem tissue. We extensively sampled spinal cords of seven women and six men with multiple sclerosis (mean disease duration= 29 years) and five healthy controls to quantify axonal density and its association with demyelination and CSA. 396 tissue blocks were embedded in paraffin and immuno-stained for myelin basic protein and phosphorylated neurofilaments. Measurements included total CSA, areas of (i) lateral cortico-spinal tracts, (ii) gray matter, (iii) white matter, (iv) demyelination, and the number of axons within the lateral cortico-spinal tracts. Linear mixed models were used to analyze relationships. In multiple sclerosis CSA reduction at cervical, thoracic and lumbar levels ranged between 19 and 24% with white (19-24%) and gray (17-21%) matter atrophy contributing equally across levels. Axonal density in multiple sclerosis was lower by 57-62% across all levels and affected all fibers regardless of diameter. Demyelination affected 24-48% of the gray matter, most extensively at the thoracic level, and 11-13% of the white matter, with no significant differences across levels. Disease duration was associated with reduced axonal density, however not with any area index. Significant association was detected between focal demyelination and decreased axonal density. In conclusion, over nearly 30 years multiple sclerosis reduces axonal density by 60% throughout the spinal cord. Spinal cord cross sectional area, reduced by about 20%, appears to be a poor predictor of axonal density.
Collapse
Affiliation(s)
- Natalia Petrova
- Blizard Institute (Neuroscience), Barts and the London School of Medicine & DentistryQueen Mary University of LondonLondonUK
| | - Daniele Carassiti
- Blizard Institute (Neuroscience), Barts and the London School of Medicine & DentistryQueen Mary University of LondonLondonUK
| | | | - David Baker
- Blizard Institute (Neuroscience), Barts and the London School of Medicine & DentistryQueen Mary University of LondonLondonUK
| | - Klaus Schmierer
- Blizard Institute (Neuroscience), Barts and the London School of Medicine & DentistryQueen Mary University of LondonLondonUK
- Neurosciences Clinical Academic Groupthe Royal London Hospital, Barts Health NHS TrustLondonUK
| |
Collapse
|
32
|
Schmierer K, McDowell A, Petrova N, Carassiti D, Thomas DL, Miquel ME. Quantifying multiple sclerosis pathology in post mortem spinal cord using MRI. Neuroimage 2018; 182:251-258. [PMID: 29373838 DOI: 10.1016/j.neuroimage.2018.01.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/04/2018] [Accepted: 01/21/2018] [Indexed: 11/26/2022] Open
Abstract
Multiple sclerosis (MS) is a common inflammatory, demyelinating and degenerative disease of the central nervous system. The majority of people with MS present with symptoms due to spinal cord damage, and in more advanced MS a clinical syndrome resembling that of progressive myelopathy is not uncommon. Significant efforts have been undertaken to predict MS-related disability based on short-term observations, for example, the spinal cord cross-sectional area measured using MRI. The histo-pathological correlates of spinal cord MRI changes in MS are incompletely understood, however a surge of interest in tissue microstructure has recently led to new approaches to improve the precision with which MRI indices relate to underlying tissue features, such as myelin content, neurite density and orientation, among others. Quantitative MRI techniques including T1 and T2, magnetisation transfer (MT) and a number of diffusion-derived indices have all been successfully applied to post mortem MS spinal cord. Combining advanced quantification of histological features with quantitative - particularly diffusion-based - MRI techniques provide a new platform for high-quality MR/pathology data generation. To more accurately quantify grey matter pathology in the MS spinal cord, a key driver of physical disability in advanced MS, remains an important challenge of microstructural imaging.
Collapse
Affiliation(s)
- K Schmierer
- Queen Mary University of London, Barts and The London School of Medicine & Dentistry, Blizard Institute (Neuroscience), London, UK; Barts Health NHS Trust, Clinical Board Medicine (Neuroscience), The Royal London Hospital, London, UK.
| | - A McDowell
- UCL Great Ormond Street Institute of Child Health, Developmental Imaging and Biophysics Section, London, UK
| | - N Petrova
- Queen Mary University of London, Barts and The London School of Medicine & Dentistry, Blizard Institute (Neuroscience), London, UK
| | - D Carassiti
- Queen Mary University of London, Barts and The London School of Medicine & Dentistry, Blizard Institute (Neuroscience), London, UK
| | - D L Thomas
- UCL Institute of Neurology, Leonard Wolfson Experimental Neurology Centre, Department of Brain Repair and Rehabilitation, Queen Square, London, UK
| | - M E Miquel
- Barts Health NHS Trust, Clinical Physics, London, UK
| |
Collapse
|
33
|
Espinosa-Hoyos D, Jagielska A, Homan KA, Du H, Busbee T, Anderson DG, Fang NX, Lewis JA, Van Vliet KJ. Engineered 3D-printed artificial axons. Sci Rep 2018; 8:478. [PMID: 29323240 PMCID: PMC5765144 DOI: 10.1038/s41598-017-18744-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/16/2017] [Indexed: 12/02/2022] Open
Abstract
Myelination is critical for transduction of neuronal signals, neuron survival and normal function of the nervous system. Myelin disorders account for many debilitating neurological diseases such as multiple sclerosis and leukodystrophies. The lack of experimental models and tools to observe and manipulate this process in vitro has constrained progress in understanding and promoting myelination, and ultimately developing effective remyelination therapies. To address this problem, we developed synthetic mimics of neuronal axons, representing key geometric, mechanical, and surface chemistry components of biological axons. These artificial axons exhibit low mechanical stiffness approaching that of a human axon, over unsupported spans that facilitate engagement and wrapping by glial cells, to enable study of myelination in environments reflecting mechanical cues that neurons present in vivo. Our 3D printing approach provides the capacity to vary independently the complex features of the artificial axons that can reflect specific states of development, disease, or injury. Here, we demonstrate that oligodendrocytes' production and wrapping of myelin depend on artificial axon stiffness, diameter, and ligand coating. This biofidelic platform provides direct visualization and quantification of myelin formation and myelinating cells' response to both physical cues and pharmacological agents.
Collapse
Affiliation(s)
- Daniela Espinosa-Hoyos
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Biosystems & Micromechanics Interdisciplinary Research Group (BioSyM), Singapore-MIT Alliance in Research & Technology (SMART), Singapore, Singapore
| | - Anna Jagielska
- Biosystems & Micromechanics Interdisciplinary Research Group (BioSyM), Singapore-MIT Alliance in Research & Technology (SMART), Singapore, Singapore
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kimberly A Homan
- Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA
- School of Engineering and Applied Sciences, Harvard University, Harvard, MA, 02138, USA
| | - Huifeng Du
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Travis Busbee
- Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA
- School of Engineering and Applied Sciences, Harvard University, Harvard, MA, 02138, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, 02139, USA
| | - Nicholas X Fang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA
- School of Engineering and Applied Sciences, Harvard University, Harvard, MA, 02138, USA
| | - Krystyn J Van Vliet
- Biosystems & Micromechanics Interdisciplinary Research Group (BioSyM), Singapore-MIT Alliance in Research & Technology (SMART), Singapore, Singapore.
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
34
|
Mahajan KR, Ontaneda D. The Role of Advanced Magnetic Resonance Imaging Techniques in Multiple Sclerosis Clinical Trials. Neurotherapeutics 2017; 14:905-923. [PMID: 28770481 PMCID: PMC5722766 DOI: 10.1007/s13311-017-0561-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Magnetic resonance imaging has been crucial in the development of anti-inflammatory disease-modifying treatments. The current landscape of multiple sclerosis clinical trials is currently expanding to include testing not only of anti-inflammatory agents, but also neuroprotective, remyelinating, neuromodulating, and restorative therapies. This is especially true of therapies targeting progressive forms of the disease where neurodegeneration is a prominent feature. Imaging techniques of the brain and spinal cord have rapidly evolved in the last decade to permit in vivo characterization of tissue microstructural changes, connectivity, metabolic changes, neuronal loss, glial activity, and demyelination. Advanced magnetic resonance imaging techniques hold significant promise for accelerating the development of different treatment modalities targeting a variety of pathways in MS.
Collapse
Affiliation(s)
- Kedar R Mahajan
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, 9500 Euclid Avenue, U-10, Cleveland, OH, 44195, USA
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis Treatment and Research, Cleveland Clinic, 9500 Euclid Avenue, U-10, Cleveland, OH, 44195, USA.
| |
Collapse
|
35
|
Jagielska A, Lowe AL, Makhija E, Wroblewska L, Guck J, Franklin RJM, Shivashankar GV, Van Vliet KJ. Mechanical Strain Promotes Oligodendrocyte Differentiation by Global Changes of Gene Expression. Front Cell Neurosci 2017; 11:93. [PMID: 28473753 PMCID: PMC5397481 DOI: 10.3389/fncel.2017.00093] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/20/2017] [Indexed: 11/13/2022] Open
Abstract
Differentiation of oligodendrocyte progenitor cells (OPC) to oligodendrocytes and subsequent axon myelination are critical steps in vertebrate central nervous system (CNS) development and regeneration. Growing evidence supports the significance of mechanical factors in oligodendrocyte biology. Here, we explore the effect of mechanical strains within physiological range on OPC proliferation and differentiation, and strain-associated changes in chromatin structure, epigenetics, and gene expression. Sustained tensile strain of 10-15% inhibited OPC proliferation and promoted differentiation into oligodendrocytes. This response to strain required specific interactions of OPCs with extracellular matrix ligands. Applied strain induced changes in nuclear shape, chromatin organization, and resulted in enhanced histone deacetylation, consistent with increased oligodendrocyte differentiation. This response was concurrent with increased mRNA levels of the epigenetic modifier histone deacetylase Hdac11. Inhibition of HDAC proteins eliminated the strain-mediated increase of OPC differentiation, demonstrating a role of HDACs in mechanotransduction of strain to chromatin. RNA sequencing revealed global changes in gene expression associated with strain. Specifically, expression of multiple genes associated with oligodendrocyte differentiation and axon-oligodendrocyte interactions was increased, including cell surface ligands (Ncam, ephrins), cyto- and nucleo-skeleton genes (Fyn, actinins, myosin, nesprin, Sun1), transcription factors (Sox10, Zfp191, Nkx2.2), and myelin genes (Cnp, Plp, Mag). These findings show how mechanical strain can be transmitted to the nucleus to promote oligodendrocyte differentiation, and identify the global landscape of signaling pathways involved in mechanotransduction. These data provide a source of potential new therapeutic avenues to enhance OPC differentiation in vivo.
Collapse
Affiliation(s)
- Anna Jagielska
- Department of Materials Science and Engineering, Massachusetts Institute of TechnologyCambridge, MA, USA
| | - Alexis L Lowe
- Department of Neuroscience, Wellesley CollegeWellesley, MA, USA
| | - Ekta Makhija
- Mechanobiology Institute, National University of SingaporeSingapore, Singapore
| | - Liliana Wroblewska
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridge, MA, USA
| | - Jochen Guck
- Biotechnology Center, Technische Universität DresdenDresden, Germany
| | - Robin J M Franklin
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of CambridgeCambridge, UK
| | - G V Shivashankar
- Mechanobiology Institute, National University of SingaporeSingapore, Singapore
| | - Krystyn J Van Vliet
- Department of Materials Science and Engineering, Massachusetts Institute of TechnologyCambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of TechnologyCambridge, MA, USA.,BioSystems and Micromechanics Inter-Disciplinary Research Group, Singapore-MIT Alliance for Research and TechnologySingapore, Singapore
| |
Collapse
|
36
|
Lee H, Narayanan S, Brown RA, Chen JT, Atkins HL, Freedman MS, Arnold DL. Brain atrophy after bone marrow transplantation for treatment of multiple sclerosis. Mult Scler 2016; 23:420-431. [DOI: 10.1177/1352458516650992] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: A cohort of patients with poor-prognosis multiple sclerosis (MS) underwent chemotherapy-based immune ablation followed by immune reconstitution with an autologous hematopoietic stem cell transplant (IA/aHSCT). This eliminated new focal inflammatory activity, but resulted in early acceleration of brain atrophy. Objective: We modeled the time course of whole-brain volume in 19 patients to identify the baseline predictors of atrophy and to estimate the average rate of atrophy after IA/aHSCT. Methods: Percentage whole-brain volume changes were calculated between the baseline and follow-up magnetic resonance imaging (MRI; mean duration: 5 years). A mixed-effects model was applied using two predictors: total busulfan dose and baseline volume of T1-weighted white-matter lesions. Results: Treatment was followed by accelerated whole-brain volume loss averaging 3.3%. Both the busulfan dose and the baseline lesion volume were significant predictors. The atrophy slowed progressively over approximately 2.5 years. There was no evidence that resolution of edema contributed to volume loss. The mean rate of long-term atrophy was −0.23% per year, consistent with the rate expected from normal aging. Conclusion: Following IA/aHSCT, MS patients showed accelerated whole-brain atrophy that was likely associated with treatment-related toxicity and degeneration of “committed” tissues. Atrophy eventually slowed to that expected from normal aging, suggesting that stopping inflammatory activity in MS can reduce secondary degeneration and atrophy.
Collapse
Affiliation(s)
- Hyunwoo Lee
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Sridar Narayanan
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Robert A Brown
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jacqueline T Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Harold L Atkins
- Ottawa Hospital Blood and Marrow Transplant Program, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Mark S Freedman
- University of Ottawa and the Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Douglas L Arnold
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
37
|
Huang SY, Tobyne SM, Nummenmaa A, Witzel T, Wald LL, McNab JA, Klawiter EC. Characterization of Axonal Disease in Patients with Multiple Sclerosis Using High-Gradient-Diffusion MR Imaging. Radiology 2016; 280:244-51. [PMID: 26859256 DOI: 10.1148/radiol.2016151582] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To evaluate the ability of high-gradient-diffusion magnetic resonance (MR) imaging by using gradient strengths of up to 300 mT/m to depict axonal disease in lesions and normal-appearing white matter (NAWM) in patients with multiple sclerosis (MS) and to compare high-gradient-diffusion MR findings in these patients with those in healthy control subjects. Materials and Methods In this HIPAA-compliant institutional review board-approved prospective study in which all subjects provided written informed consent, six patients with relapsing-remitting MS and six healthy control subjects underwent diffusion-weighted imaging with a range of diffusion weightings performed with a 3-T human MR imager by using gradient strengths of up to 300 mT/m. A model of intra-axonal, extra-axonal, and free water diffusion was fitted to obtain estimates of axon diameter and density. Differences in axon diameter and density between lesions and NAWM in patients with MS were assessed by using the nonparametric Wilcoxon matched-pairs signed rank test, and differences between NAWM in subjects with MS and white matter in healthy control subjects were assessed by using the Mann-Whitney U test. Results MS lesions showed increased mean axon diameter (10.3 vs 7.9 μm in the genu, 10.4 vs 9.3 μm in the body, and 10.6 vs 8.2 μm in the splenium; P < .05) and decreased axon density ([0.48 vs 1.1] × 10(10)/m(2) in the genu, [0.40 vs 0.70] × 10(10)/m(2) in the body, and [0.35 vs 1.1] × 10(10)/m(2) in the splenium; P < .05) compared with adjacent NAWM. No significant difference in mean axon diameter or axon density was detected between NAWM in subjects with MS and white matter in healthy control subjects. Conclusion High-gradient-diffusion MR imaging using gradient strengths of up to 300 mT/m can be used to characterize axonal disease in patients with MS, with results that agree with known trends from neuropathologic data showing increased axon diameter and decreased axon density in MS lesions when compared with NAWM. (©) RSNA, 2016 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Susie Y Huang
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Sean M Tobyne
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Aapo Nummenmaa
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Thomas Witzel
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Lawrence L Wald
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Jennifer A McNab
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| | - Eric C Klawiter
- From the Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th St, Charlestown, MA 02129 (S.Y.H., A.N., T.W., L.L.W.); Department of Neurology, Massachusetts General Hospital, Boston, Mass (S.M.T., E.C.K.); and Richard M. Lucas Center for Imaging, Department of Radiology, Stanford University, Stanford, Calif (J.A.M.)
| |
Collapse
|
38
|
Stone LA, Cutter GR, Fisher E, Richert N, McCartin J, Ohayon J, Bash C, McFarland H. Relapse May Serve as a Mediator Variable in Longitudinal Outcomes in Multiple Sclerosis. J Neuroimaging 2015; 26:296-302. [PMID: 26686343 DOI: 10.1111/jon.12321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 11/02/2015] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND/PURPOSE Contrast-enhancing lesions (CEL) on magnetic resonance imaging (MRI) are believed to represent inflammatory disease activity in multiple sclerosis (MS), but their relationship to subsequent long-term disability and progression is unclear, particularly at longer time periods such as 8-10 years. METHODS Between 1989 and 1994, 111 MS patients were seen at the National Institutes of Health for clinical evaluations and 3 monthly contrast-enhanced MRI scans. Of these, 94 patients were re-evaluated a mean of 8 years later (range 6.1-10.5 years) with a single MRI scan and clinical evaluation. CEL number and volume were determined at baseline and follow-up. The number of relapses was ascertained over the follow-up period and annualized relapse rates were calculated. Other MRI parameters, such as T2 hyperintensity volume, T1 volume, and brain parenchymal fraction, were also calculated. RESULTS While there was no direct correlation between CEL number or volume at baseline and disability status at follow-up, CEL measures at baseline did correlate with number of relapses observed in the subsequent years, and the number of relapses in turn correlated with subsequent disability as well as transition to progressive MS. CONCLUSION While number and volume of CEL at baseline do not directly correlate with disability in the longer term in MS, our data suggest that 1 route to disability involves relapses as a mediator variable in the causal sequence of MS progression from CEL to disability. Further studies using relapse as a mediator variable in a larger data set may be warranted.
Collapse
Affiliation(s)
- Lael Anne Stone
- Mellen Center for Treatment and Research, Cleveland Clinic Foundation, Cleveland, OH
| | | | | | | | - Jennifer McCartin
- Neuroimmunology Branch of the National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Joan Ohayon
- Neuroimmunology Branch of the National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Craig Bash
- Department of Neuroradiology, Uniformed Services School of Medicine, Bethesda, MD
| | | |
Collapse
|
39
|
Goodin DS, Reder AT, Bermel RA, Cutter GR, Fox RJ, John GR, Lublin FD, Lucchinetti CF, Miller AE, Pelletier D, Racke MK, Trapp BD, Vartanian T, Waubant E. Relapses in multiple sclerosis: Relationship to disability. Mult Scler Relat Disord 2015; 6:10-20. [PMID: 27063617 DOI: 10.1016/j.msard.2015.09.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 08/21/2015] [Accepted: 09/02/2015] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a recurrent inflammatory disease of the central nervous system, which ultimately causes substantial disability in many patients. A key clinical feature of this disease is the occurrence of relapses, consisting of episodes of neurological dysfunction followed by periods of remission. This review considers in detail the importance of the occurrence of relapses to the ultimate course of MS and the impact of relap setreatment (both acutely and prophylactically) on the long-term outcome for individuals. The ultimate goal of therapy in MS is the reduction of long-term disability. Clinical trials in MS, however, typically only extend for a very short time period compared to the time it takes for disability to evolve. Consequently, short-term outcome measures that are associated with, and predict, future disability need to be identified. In this regard, not only are relapses a characteristic feature of MS, they have also been proven to be associated with the occurrence of long-term disability. Moreover, treatments that reduce the number and severity of these attacks improve the long-term prognosis.
Collapse
Affiliation(s)
- Douglas S Goodin
- Multiple Sclerosis Center, University of California, San Francisco Medical Center, San Francisco, CA, United States; Department of Neurology, University of California, San Francisco School of Medicine, San Francisco, CA, United States.
| | - Anthony T Reder
- Department of Neurology, The University of Chicago, Chicago, IL, United States
| | - Robert A Bermel
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, United States
| | - Gary R Cutter
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert J Fox
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Gareth R John
- Multiple Sclerosis Research Laboratory, Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Friedman Brain Institute, New York, NY, United States; Department of Neurology, Mount Sinai School of Medicine, New York, NY, United States
| | - Fred D Lublin
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Aaron E Miller
- Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Daniel Pelletier
- Neuro-Immunology Division and Yale Multiple Sclerosis Center, Advanced Imaging in Multiple Sclerosis (AIMS) Laboratory, Yale University School of Medicine, New Haven, CT, United States
| | - Michael K Racke
- Department of Neurology, Wexner Medical Center at The Ohio State University, Columbus, OH, United States
| | - Bruce D Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Timothy Vartanian
- Judith Jaffe Multiple Sclerosis Center, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medical College, United States
| | - Emmanuelle Waubant
- UCSF Regional Pediatric MS Center, Race to Erase MS, San Francisco, CA, United States
| |
Collapse
|
40
|
Multicontrast MRI Quantification of Focal Inflammation and Degeneration in Multiple Sclerosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:569123. [PMID: 26295042 PMCID: PMC4532805 DOI: 10.1155/2015/569123] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/07/2014] [Accepted: 11/07/2014] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Local microstructural pathology in multiple sclerosis patients might influence their clinical performance. This study applied multicontrast MRI to quantify inflammation and neurodegeneration in MS lesions. We explored the impact of MRI-based lesion pathology in cognition and disability. METHODS 36 relapsing-remitting MS subjects and 18 healthy controls underwent neurological, cognitive, behavioural examinations and 3 T MRI including (i) fluid attenuated inversion recovery, double inversion recovery, and magnetization-prepared gradient echo for lesion count; (ii) T1, T2, and T2(*) relaxometry and magnetisation transfer imaging for lesion tissue characterization. Lesions were classified according to the extent of inflammation/neurodegeneration. A generalized linear model assessed the contribution of lesion groups to clinical performances. RESULTS Four lesion groups were identified and characterized by (1) absence of significant alterations, (2) prevalent inflammation, (3) concomitant inflammation and microdegeneration, and (4) prevalent tissue loss. Groups 1, 3, 4 correlated with general disability (Adj-R (2) = 0.6; P = 0.0005), executive function (Adj-R (2) = 0.5; P = 0.004), verbal memory (Adj-R (2) = 0.4; P = 0.02), and attention (Adj-R (2) = 0.5; P = 0.002). CONCLUSION Multicontrast MRI provides a new approach to infer in vivo histopathology of plaques. Our results support evidence that neurodegeneration is the major determinant of patients' disability and cognitive dysfunction.
Collapse
|
41
|
Tovar-Moll F, Evangelou IE, Chiu AW, Auh S, Chen C, Ehrmantraut M, Ohayon JM, Richert N, Bagnato F. Diffuse and focal corticospinal tract disease and its impact on patient disability in multiple sclerosis. J Neuroimaging 2014; 25:200-206. [PMID: 25318661 DOI: 10.1111/jon.12171] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 06/15/2014] [Accepted: 07/13/2014] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE We investigated the impact of focal and diffuse corticospinal tracts damage on sensory-motor disability in multiple sclerosis (MS) patients. METHODS Twenty-five MS patients underwent 3.0 Tesla (3T) magnetic resonance imaging with diffusion tensor imaging (DTI). The Expanded Disability Status Scale (EDSS) and the Timed 25-Foot Walk test (T25FW) quantified patient physical disability. Fractional anisotropy (FA) and mean diffusivity (MD) of the corticospinal tracts, whole brain and corticospinal tracts lesion volume were also computed. Spearman rank correlation analyses measured the associations between DTI-derived metrics and other measures of disease. Partial correlation analyses between DTI and disability measures were performed and corrected for lesion volumes as appropriate. RESULTS Significant associations were seen between FA of the corticospinal tracts and EDSS (r = -.500, P = .0011), motor-EDSS (r = -.519, P = .008), and T25WF (r = -.637, P = .001) scores and MD of the corticospinal tracts and motor-EDSS (r = .469, P = .018) and T25WF (r = .428, P = .033) scores. When correcting for lesion volumes, only the association between FA of the corticospinal tracts and EDSS (r ≤ -.516, p ≤ .01) or motor-EDSS score (r ≤ -.516, p ≤ .01) persisted. CONCLUSIONS DTI at 3T shows that the impact of diffuse corticospinal tracts disease on sensory-motor disability is greatly mediated by focal lesions in MS.
Collapse
Affiliation(s)
- Fernanda Tovar-Moll
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD.,D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.,Institute of Biomedical Sciences and National Center of Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Iordanis E Evangelou
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Annie W Chiu
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Sungyoung Auh
- Office of the Clinical Director, NINDS, NIH, Bethesda, MD
| | - Christina Chen
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Mary Ehrmantraut
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Joan M Ohayon
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Nancy Richert
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| | - Francesca Bagnato
- Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD
| |
Collapse
|
42
|
Postmortem magnetic resonance imaging to guide the pathologic cut: individualized, 3-dimensionally printed cutting boxes for fixed brains. J Neuropathol Exp Neurol 2014; 73:780-8. [PMID: 25007244 DOI: 10.1097/nen.0000000000000096] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Interfacing magnetic resonance imaging (MRI) with pathology is critically important for understanding the pathologic basis of MRI signal changes in vivo and for clinicopathologic correlations. Postmortem MRI is an intermediate step in this process; unfortunately, however, relating the data to standard pathologic sections, which are relatively thick and often nonparallel, is both time-consuming and insufficiently accurate. The aim of this project was to develop technology to integrate postmortem, high-resolution, whole-brain MRI into the planning and execution of pathologic analysis through precise localization of the target and coordinates of cut. Compared with standard pathologic sectioning, the use of an individualized, 3-dimensionally printed cutting box-designed based on postmortem MRI of formalin-fixed whole brains-improved the speed, quality, and accuracy of radiologic-pathologic correlations and, specifically, the histopathologic localization of imaging findings. The technology described herein is easily implemented, applicable to any brain disorder, and potentially extendable to other organs. From the point of view of the pathologist, this technique can improve localization of small or subtle abnormalities, whereas from the point of view of the radiologist, it has the potential to improve understanding of MRI signal changes observed in diseases.
Collapse
|
43
|
Ciccarelli O, Barkhof F, Bodini B, Stefano ND, Golay X, Nicolay K, Pelletier D, Pouwels PJW, Smith SA, Wheeler-Kingshott CAM, Stankoff B, Yousry T, Miller DH. Pathogenesis of multiple sclerosis: insights from molecular and metabolic imaging. Lancet Neurol 2014; 13:807-22. [DOI: 10.1016/s1474-4422(14)70101-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
44
|
Beall EB, Lowe MJ. SimPACE: generating simulated motion corrupted BOLD data with synthetic-navigated acquisition for the development and evaluation of SLOMOCO: a new, highly effective slicewise motion correction. Neuroimage 2014; 101:21-34. [PMID: 24969568 DOI: 10.1016/j.neuroimage.2014.06.038] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 06/10/2014] [Accepted: 06/15/2014] [Indexed: 10/25/2022] Open
Abstract
Head motion in functional MRI and resting-state MRI is a major problem. Existing methods do not robustly reflect the true level of motion artifact for in vivo fMRI data. The primary issue is that current methods assume that motion is synchronized to the volume acquisition and thus ignore intra-volume motion. This manuscript covers three sections in the use of gold-standard motion-corrupted data to pursue an intra-volume motion correction. First, we present a way to get motion corrupted data with accurately known motion at the slice acquisition level. This technique simulates important data acquisition-related motion artifacts while acquiring real BOLD MRI data. It is based on a novel motion-injection pulse sequence that introduces known motion independently for every slice: Simulated Prospective Acquisition CorrEction (SimPACE). Secondly, with data acquired using SimPACE, we evaluate several motion correction and characterization techniques, including several commonly used BOLD signal- and motion parameter-based metrics. Finally, we introduce and evaluate a novel, slice-based motion correction technique. Our novel method, SLice-Oriented MOtion COrrection (SLOMOCO) performs better than the volumetric methods and, moreover, accurately detects the motion of independent slices, in this case equivalent to the known injected motion. We demonstrate that SLOMOCO can model and correct for nearly all effects of motion in BOLD data. Also, none of the commonly used motion metrics was observed to robustly identify motion corrupted events, especially in the most realistic scenario of sudden head movement. For some popular metrics, performance was poor even when using the ideal known slice motion instead of volumetric parameters. This has negative implications for methods relying on these metrics, such as recently proposed motion correction methods such as data censoring and global signal regression.
Collapse
Affiliation(s)
- Erik B Beall
- Imaging Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA.
| | - Mark J Lowe
- Imaging Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH 44195, USA
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW The predominant clinical disease course of multiple sclerosis starts with reversible episodes of neurological disability, which transforms into progressive neurological decline. This review provides insight into the pathological differences during relapsing and progressive phases of multiple sclerosis. RECENT FINDINGS The clinical course of multiple sclerosis is variable, and the disease can be classified into relapsing and progressive phases. Pathological studies have been successful in distinguishing between these two forms of the disease and correlate with the clinical findings in terms of cellular responses, the inflammatory environment, and the location of lesions. SUMMARY Available therapies for multiple sclerosis patients, while effective during the relapsing phase, have little benefit for progressive multiple sclerosis patients. Development of therapies to benefit progressive multiple sclerosis patients will require a better understanding of the pathogenesis of progressive multiple sclerosis. This review discusses and compares the pathological findings in relapsing and progressive multiple sclerosis patients.
Collapse
Affiliation(s)
- Ranjan Dutta
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | |
Collapse
|
46
|
Aung WY, Mar S, Benzinger TL. Diffusion tensor MRI as a biomarker in axonal and myelin damage. ACTA ACUST UNITED AC 2013; 5:427-440. [PMID: 24795779 DOI: 10.2217/iim.13.49] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Diffusion tensor imaging has been used extensively as a research tool to understand the structural changes associated with white matter pathology. Using water diffusion as the basis to construct anatomic details, diffusion tensor imaging offers the potential to identify structural and functional adaptations before gross anatomical changes, such as lesions and tumors, become apparent on conventional MRI. Over the past 10 years, further parameters, such as axial and radial diffusivity, have been developed to characterize white matter changes specific to axons and myelin. In this paper, the potential application and outstanding issues on the use of diffusion tensor imaging directional diffusivity as a biomarker in axonal and myelin damage in neurological disorders will be reviewed.
Collapse
Affiliation(s)
- Wint Yan Aung
- Department of Radiology, Washington University, School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA
| | - Soe Mar
- Department of Pediatric & Developmental Neurology, Washington University School of Medicine, St Louis, MO, USA
| | - Tammie Ls Benzinger
- Department of Radiology, Washington University, School of Medicine, 510 South Kingshighway Boulevard, St Louis, MO 63110, USA ; Department of Neurological Surgery, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
47
|
Zhang Y, Moore GRW, Laule C, Bjarnason TA, Kozlowski P, Traboulsee A, Li DKB. Pathological correlates of magnetic resonance imaging texture heterogeneity in multiple sclerosis. Ann Neurol 2013; 74:91-9. [PMID: 23939554 DOI: 10.1002/ana.23867] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 01/18/2013] [Accepted: 02/01/2013] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To analyze the texture of T2-weighted magnetic resonance imaging (MRI) of postmortem multiple sclerosis (MS) brain, and to determine whether and how MRI texture correlates with tissue pathology. METHODS Ten brain samples from 3 subjects with MS were examined. Areas of complete, partial, or no loss of Luxol fast blue (myelin) and Bielschowsky (axons) staining were marked on histological images, and matched on corresponding MRI as lesions, diffusely abnormal white matter (DAWM), and normal-appearing white matter (NAWM). The number of CD45(+) cells (inflammation) was also counted. MRI texture was computed using polar Stockwell transform and compared to histology. RESULTS Thirty-four lesions, 17 DAWM regions, and 36 NAWM regions were identified. After mixed effects modeling, MRI texture heterogeneity was greater in lesions than in DAWM (p < 0.001) and NAWM (p < 0.001), and was greater in DAWM than in NAWM (p < 0.001); the number of CD45+ cells was greater in both lesions (p < 0.001) and DAWM (p = 0.005) than in NAWM. In MRI, a gradient of texture heterogeneity was detected in lesions, with gradual tapering toward perilesional NAWM. Moreover, besides univariate correlation with histological markers, texture heterogeneity correlated independently with normalized myelin density (p < 0.01) when random effects were considered. Within sample, MRI texture correlated with myelin and axonal density in 7 of 10 samples (p < 0.01). INTERPRETATION Texture analysis performed on routine clinical magnetic resonance images may be a potential measure of tissue integrity. Tissues with more severe myelin and axonal pathology are associated with greater texture heterogeneity.
Collapse
Affiliation(s)
- Yunyan Zhang
- Department of Radiology, University of Calgary, Calgary, Alberta; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta
| | | | | | | | | | | | | |
Collapse
|
48
|
Luo J, Yablonskiy DA, Hildebolt CF, Lancia S, Cross AH. Gradient echo magnetic resonance imaging correlates with clinical measures and allows visualization of veins within multiple sclerosis lesions. Mult Scler 2013; 20:349-55. [PMID: 23836876 DOI: 10.1177/1352458513495935] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Conventional magnetic resonance imaging (MRI) methods do not quantify the severity of multiple sclerosis (MS) white matter lesions or measure pathology within normal-appearing white matter (NAWM). OBJECTIVE Gradient Echo Plural Contrast Imaging (GEPCI), a fast MRI technique producing inherently co-registered images for qualitative and quantitative assessment of MS, was used to 1) correlate with disability; 2) distinguish clinical MS subtypes; 3) determine prevalence of veins co-localized within lesions in WM. METHODS Thirty subjects representing relapsing-remitting MS (RRMS), secondary progressive MS (SPMS) and primary progressive MS (PPMS) subtypes were scanned with clinical and GEPCI protocols. Standard measures of physical disability and cognition were correlated with magnetic resonance metrics. Lesions with central veins were counted for RRMS subjects. RESULTS Tissue damage load (TDL-GEPCI) and lesion load (LL-GEPCI) derived with GEPCI correlated better with MS functional composite (MSFC) measures and most other neurologic measures than lesion load derived with FLAIR (LL-FLAIR). GEPCI correctly classified clinical subtypes in 70% subjects. A central vein could be identified in 76% of WM lesions in RRMS subjects on GEPCI T2*-SWI images. CONCLUSION GEPCI lesion metrics correlated better with neurologic disability than lesion load derived using FLAIR imaging, and showed promise in classifying clinical subtypes of MS. These improvements are likely attributable to the ability of GEPCI to quantify tissue damage.
Collapse
Affiliation(s)
- Jie Luo
- Department of Radiology, Washington University, St. Louis MO, USA
| | | | | | | | | |
Collapse
|
49
|
Fiber pathway pathology, synapse loss and decline of cortical function in schizophrenia. PLoS One 2013; 8:e60518. [PMID: 23593232 PMCID: PMC3620229 DOI: 10.1371/journal.pone.0060518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/28/2013] [Indexed: 11/19/2022] Open
Abstract
A quantitative cortical model is developed, based on both computational and simulation approaches, which relates measured changes in cortical activity of gray matter with changes in the integrity of longitudinal fiber pathways. The model consists of modules of up to 5,000 neurons each, 80% excitatory and 20% inhibitory, with these having different degrees of synaptic connectiveness both within a module as well as between modules. It is shown that if the inter-modular synaptic connections are reduced to zero while maintaining the intra-modular synaptic connections constant, then activity in the modules is reduced by about 50%. This agrees with experimental observations in which cortical electrical activity in a region of interest, measured using the rate of oxidative glucose metabolism (CMRglc(ox)), is reduced by about 50% when the cortical region is isolated, either by surgical means or by transient cold block. There is also a 50% decrease in measured cortical activity following inactivation of the nucleus of Meynert and the intra-laminar nuclei of the thalamus, which arise either following appropriate lesions or in sleep. This occurs in the model if the inter-modular synaptic connections require input from these nuclei in order to function. In schizophrenia there is a 24% decrease in functional anisotropy of longitudinal fasciculi accompanied by a 7% decrease in cortical activity (CMRglc(ox)).The cortical model predicts this, namely for a 24% decrease in the functioning of the inter-modular connections, either through the complete loss of 24% of axons subserving the connections or due to such a decrease in the efficacy of all the inter-modular connections, there will be about a 7% decrease in the activity of the modules. This work suggests that deterioration of longitudinal fasciculi in schizophrenia explains the loss of activity in the gray matter.
Collapse
|
50
|
Chen JTH, Easley K, Schneider C, Nakamura K, Kidd GJ, Chang A, Staugaitis SM, Fox RJ, Fisher E, Arnold DL, Trapp BD. Clinically feasible MTR is sensitive to cortical demyelination in MS. Neurology 2012; 80:246-52. [PMID: 23269598 DOI: 10.1212/wnl.0b013e31827deb99] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE Presently there is no clinically feasible imaging modality that can effectively detect cortical demyelination in patients with multiple sclerosis (MS). The objective of this study is to determine if clinically feasible magnetization transfer ratio (MTR) imaging is sensitive to cortical demyelination in MS. METHODS MRI were acquired in situ on 7 recently deceased patients with MS using clinically feasible sequences at 3 T, including relatively high-resolution T1-weighted and proton density-weighted images with/without a magnetization transfer pulse for calculation of MTR. The brains were rapidly removed and placed in fixative. Multiple cortical regions from each brain were immunostained for myelin proteolipid protein and classified as mostly myelinated (MM(ctx)), mostly demyelinated (MD(ctx)), or intermediately demyelinated (ID(ctx)). MRIs were registered with the cortical sections so that the cortex corresponding to each cortical section could be identified, along with adjacent subcortical white matter (WM). Mean cortical MTR normalized to mean WM MTR was calculated for each cortical region. Linear mixed-effects models were used to test if mean normalized cortical MTR was significantly lower in demyelinated cortex. RESULTS We found that mean normalized cortical MTR was significantly lower in cortical tissue with any demyelination (ID(ctx) or MD(ctx)) compared to MM(ctx) (demyelinated cortex: least-squares mean [LSM] = 0.797, SE = 0.007; MM(ctx): LSM = 0.837, SE = 0.006; p = 0.01, n = 89). CONCLUSIONS This result demonstrates that clinically feasible MTR imaging is sensitive to cortical demyelination and suggests that MTR will be a useful tool to help detect MS cortical lesions in living patients with MS.
Collapse
|