1
|
Linzey M, DiSano K, Welsh N, Ford JC, Gilli F, Wishart H, Pachner A. High throughput method for detecting murine brain atrophy using a clinical 3T MRI. BMC Med Imaging 2023; 23:183. [PMID: 37957588 PMCID: PMC10641942 DOI: 10.1186/s12880-023-01124-0] [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: 07/08/2022] [Accepted: 10/10/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND There is a lack of understanding of the mechanisms by which the CNS is injured in multiple sclerosis (MS). Since Theiler's murine encephalomyelitis virus (TMEV) infection in SJL/J mice is an established model of progressive disability in MS, and CNS atrophy correlates with progressive disability in MS, we used in vivo MRI to quantify total ventricular volume in TMEV infection. We then sought to identify immunological and virological biomarkers that correlated with increased ventricular size. METHODS Mice, both infected and control, were followed for 6 months. Cerebral ventricular volumes were determined by MRI, and disability was assessed by Rotarod. A range of immunological and virological measures was obtained using standard techniques. RESULTS Disability was present in infected mice with enlarged ventricles, while infected mice without enlarged ventricles had Rotarod performance similar to sham mice. Ventricular enlargement was detected as soon as 1 month after infection. None of the immunological and virological measures correlated with the development of ventricular enlargement. CONCLUSIONS These results support TMEV infection with brain MRI monitoring as a useful model for exploring the biology of disability progression in MS, but they did not identify an immunological or virological correlate with ventricular enlargement.
Collapse
Affiliation(s)
- Michael Linzey
- Integrative Neuroscience at Dartmouth, Dartmouth College, Hanover, NH, US.
| | - Krista DiSano
- Department of Veterans Affairs Medical Center, White River Junction, Vermont, US
| | - Nora Welsh
- Integrative Neuroscience at Dartmouth, Dartmouth College, Hanover, NH, US
| | - James C Ford
- Department of Psychiatry at Dartmouth Hitchcock Medical Center, New Hampshire, US
| | - Francesca Gilli
- Integrative Neuroscience at Dartmouth, Dartmouth College, Hanover, NH, US
- Department of Veterans Affairs Medical Center, White River Junction, Vermont, US
- Department of Neurology at Dartmouth Hitchcock Medical Center, Lebanon New Hampshire, US
| | - Heather Wishart
- Department of Psychiatry at Dartmouth Hitchcock Medical Center, New Hampshire, US
| | - Andrew Pachner
- Department of Neurology at Dartmouth Hitchcock Medical Center, Lebanon New Hampshire, US
| |
Collapse
|
2
|
Pol S, Dhanraj R, Taher A, Crever M, Charbonneau T, Schweser F, Dwyer M, Zivadinov R. Effect of Siponimod on Brain and Spinal Cord Imaging Markers of Neurodegeneration in the Theiler's Murine Encephalomyelitis Virus Model of Demyelination. Int J Mol Sci 2023; 24:12990. [PMID: 37629171 PMCID: PMC10455446 DOI: 10.3390/ijms241612990] [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: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Siponimod (Sp) is a Sphingosine 1-phosphate (S1P) receptor modulator, and it suppresses S1P- mediated autoimmune lymphocyte transport and inflammation. Theiler's murine encephalomyelitis virus (TMEV) infection mouse model of multiple sclerosis (MS) exhibits inflammation-driven acute and chronic phases, spinal cord lesions, brain and spinal cord atrophy, and white matter injury. The objective of the study was to investigate whether Sp treatment could attenuate inflammation-induced pathology in the TMEV model by inhibiting microglial activation and preventing the atrophy of central nervous tissue associated with neurodegeneration. Clinical disability score (CDS), body weight (BW), and rotarod retention time measures were used to assess Sp's impact on neurodegeneration and disease progression in 4 study groups of 102 animals, including 44 Sp-treated (SpT), 44 vehicle-treated, 6 saline-injected, and 8 age-matched healthy controls (HC). Next, 58 (22 SpT, 22 vehicle, 6 saline injected, and 8 HC) out of the 102 animals were further evaluated to assess the effect of Sp on brain region-specific and spinal cord volume changes, as well as microglial activation. Sp increased CDS and decreased BW and rotarod retention time in TMEV mice, but did not significantly affect most brain region volumes, except for lateral ventricle volume. Sp suppressed ventricular enlargement, suggesting reduced TMEV-induced inflammation in LV. No significant differences in spine volume changes were observed between Sp- and vehicle-treated animals, but there were differences between HC and TMEV groups, indicating TMEV-induced inflammation contributed to increased spine volume. Spine histology revealed no significant microglial density differences between groups in gray matter, but HC animals had higher type 1 morphology and lower type 2 morphology percentages in gray and white matter regions. This suggests that Sp did not significantly affect microglial density but may have modulated neuroinflammation in the spinal cord. Sp may have some effects on neuroinflammation and ventricular enlargement. However, it did not demonstrate a significant impact on neurodegeneration, spinal volume, or lesion volume in the TMEV mouse model. Further investigation is required to fully understand Sp's effect on microglial activation and its relevance to the pathophysiology of MS. The differences between the current study and previous research using other MS models, such as EAE, highlight the differences in pathological processes in these two disease models.
Collapse
Affiliation(s)
- Suyog Pol
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
- Center for Biomedical Imaging, Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Ravendra Dhanraj
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
| | - Anissa Taher
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
| | - Mateo Crever
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
| | - Taylor Charbonneau
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
| | - Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
- Center for Biomedical Imaging, Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Michael Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
- Center for Biomedical Imaging, Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA; (S.P.); (R.D.); (A.T.); (M.C.); (T.C.); (F.S.); (M.D.)
- Center for Biomedical Imaging, Clinical Translational Science Institute, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| |
Collapse
|
3
|
Gilli F, Ceccarelli A. Magnetic resonance imaging approaches for studying mouse models of multiple sclerosis: A mini review. J Neurosci Res 2023. [DOI: 10.1002/jnr.25193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023]
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center Geisel School of Medicine at Dartmouth Lebanon New Hampshire USA
| | - Antonia Ceccarelli
- Department of Neurology EpiCURA Centre Hospitalier Ath Belgium
- Hearthrhythmanagement, UZB Brussels Belgium
| |
Collapse
|
4
|
Goddery EN, Fain CE, Lipovsky CG, Ayasoufi K, Yokanovich LT, Malo CS, Khadka RH, Tritz ZP, Jin F, Hansen MJ, Johnson AJ. Microglia and Perivascular Macrophages Act as Antigen Presenting Cells to Promote CD8 T Cell Infiltration of the Brain. Front Immunol 2021; 12:726421. [PMID: 34526998 PMCID: PMC8435747 DOI: 10.3389/fimmu.2021.726421] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 01/01/2023] Open
Abstract
CD8 T cell infiltration of the central nervous system (CNS) is necessary for host protection but contributes to neuropathology. Antigen presenting cells (APCs) situated at CNS borders are thought to mediate T cell entry into the parenchyma during neuroinflammation. The identity of the CNS-resident APC that presents antigen via major histocompatibility complex (MHC) class I to CD8 T cells is unknown. Herein, we characterize MHC class I expression in the naïve and virally infected brain and identify microglia and macrophages (CNS-myeloid cells) as APCs that upregulate H-2Kb and H-2Db upon infection. Conditional ablation of H-2Kb and H-2Db from CNS-myeloid cells allowed us to determine that antigen presentation via H-2Db, but not H-2Kb, was required for CNS immune infiltration during Theiler's murine encephalomyelitis virus (TMEV) infection and drives brain atrophy as a consequence of infection. These results demonstrate that CNS-myeloid cells are key APCs mediating CD8 T cell brain infiltration.
Collapse
Affiliation(s)
- Emma N. Goddery
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Cori E. Fain
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Chloe G. Lipovsky
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | | | - Lila T. Yokanovich
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Courtney S. Malo
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Roman H. Khadka
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Zachariah P. Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Zivadinov R, Schweser F, Dwyer MG, Pol S. Detection of Monocyte/Macrophage and Microglia Activation in the TMEV Model of Chronic Demyelination Using USPIO-Enhanced Ultrahigh-Field Imaging. J Neuroimaging 2020; 30:769-778. [PMID: 32866329 DOI: 10.1111/jon.12768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Blood-derived monocytes/macrophages can be labeled with ultrasmall superparamagnetic iron oxides (USPIO) at periphery and subsequently migrate into areas of inflammation in the brain. We investigated temporal pattern of migration of peripheral immune cells in Theiler's murine encephalomyelitis virus (TMEV) model of chronic demyelination by USPIO-enhanced imaging. METHODS Fifteen SJL mice (Envigo, Indianapolis, IN) were injected with TMEV (n = 12) or saline (n = 3) at 7 weeks of age. Brain MRI of 9.4 T was performed at 3 months postinfection (mpi) (the peak of inflammatory phase), at 4, 5, and 7 mpi (throughout neurodegenerative phase) using T2*-weighted gradient echo MRI, and performed 24 hours after USPIO injection. Contrast enhancing lesion (CEL) number and volume were measured and development of brain atrophy was assessed across serial time points. Clinical disability scale and rotarod score assessed disease progression. RESULTS CEL was detected in a total of eight (66.7%) TMEV-infected animals and none of the Controls. The CEL was present in four (33.3%) TMEV-infected animals at 3 mpi, two (16.7%) at 4 mpi, six (54.5%) at 5 mpi, and four (44.4%) at 7 mpi, respectively. In TMEV-infected animals, the CEL number and volume increased significantly from 3 to 7 mpi (P < .01 for both). The correlation between total CEL number and volume with clinical and MRI outcomes was trending (P < .05). On histopathology analysis, CEL showed increased density of Iba1 staining for microglia activity. CONCLUSIONS Serial USPIO imaging is a promising biomarker for investigating the effect of therapeutic interventions on monocytes/macrophages and microglia activation and neurodegeneration in TMEV-infected animals.
Collapse
Affiliation(s)
- Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, NY
| | - Ferdinand Schweser
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, NY
| | - Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
- Center for Biomedical Imaging at Clinical Translational Science Institute, University at Buffalo, State University of New York, NY
| | - Suyog Pol
- Buffalo Neuroimaging Analysis Center, Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
6
|
Imaging in mice and men: Pathophysiological insights into multiple sclerosis from conventional and advanced MRI techniques. Prog Neurobiol 2019; 182:101663. [PMID: 31374243 DOI: 10.1016/j.pneurobio.2019.101663] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/17/2019] [Accepted: 07/17/2019] [Indexed: 01/16/2023]
Abstract
Magnetic resonance imaging (MRI) is the most important tool for diagnosing multiple sclerosis (MS). However, MRI is still unable to precisely quantify the specific pathophysiological processes that underlie imaging findings in MS. Because autopsy and biopsy samples of MS patients are rare and biased towards a chronic burnt-out end or fulminant acute early stage, the only available methods to identify human disease pathology are to apply MRI techniques in combination with subsequent histopathological examination to small animal models of MS and to transfer these insights to MS patients. This review summarizes the existing combined imaging and histopathological studies performed in MS mouse models and humans with MS (in vivo and ex vivo), to promote a better understanding of the pathophysiology that underlies conventional MRI, diffusion tensor and magnetization transfer imaging findings in MS patients. Moreover, it provides a critical view on imaging capabilities and results in MS patients and mouse models and for future studies recommends how to combine those particular MR sequences and parameters whose underlying pathophysiological basis could be partly clarified. Further combined longitudinal in vivo imaging and histopathological studies on rationally selected, appropriate mouse models are required.
Collapse
|
7
|
Comparison of Reported Spinal Cord Lesions in Progressive Multiple Sclerosis with Theiler's Murine Encephalomyelitis Virus Induced Demyelinating Disease. Int J Mol Sci 2019; 20:ijms20040989. [PMID: 30823515 PMCID: PMC6413032 DOI: 10.3390/ijms20040989] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/10/2019] [Accepted: 02/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Spinal cord (SC) lesions in Theiler's murine encephalomyelitis virus induced demyelinating disease (TMEV-IDD) resemble important features of brain lesions in progressive multiple sclerosis (MS) including inflammation, demyelination, and axonal damage. The aim of the present study was a comparison of SC lesions in MS and TMEV-IDD focusing on spatial and temporal distribution of demyelination, inflammation, SC atrophy (SCA), and axonal degeneration/loss in major descending motor pathways. METHODS TMEV and mock-infected mice were investigated clinically once a week. SC tissue was collected at 42, 98, 147, and 196 days post infection, and investigated using hematoxylin and eosin (HE) staining, immunohistochemistry targeting myelin basic protein (demyelination), Mac3 (microglia/macrophages), phosphorylated neurofilaments (axonal damage) and transmission electron microscopy. RESULTS Demyelination prevailed in SC white matter in TMEV-IDD, contrasting a predominant gray matter involvement in MS. TMEV-infected mice revealed a significant loss of axons similar to MS. Ultrastructural analysis in TMEV-IDD revealed denuded axons, degenerative myelin changes, axonal degeneration, as well as remyelination. SCA is a consistent finding in the SC of MS patients and was also detected at a late time point in TMEV-IDD. CONCLUSION This comparative study further indicates the suitability of TMEV-IDD as animal model also for the investigation of progressive SC lesions in MS.
Collapse
|
8
|
Laso-García F, Ramos-Cejudo J, Carrillo-Salinas FJ, Otero-Ortega L, Feliú A, Gómez-de Frutos M, Mecha M, Díez-Tejedor E, Guaza C, Gutiérrez-Fernández M. Therapeutic potential of extracellular vesicles derived from human mesenchymal stem cells in a model of progressive multiple sclerosis. PLoS One 2018; 13:e0202590. [PMID: 30231069 PMCID: PMC6145506 DOI: 10.1371/journal.pone.0202590] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/05/2018] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication and as possible therapeutic agents in inflammation-mediated demyelinating diseases, including multiple sclerosis (MS). In the present study, we investigated whether intravenously administered EVs derived from mesenchymal stem cells (MSCs) from human adipose tissue might mediate recovery in Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease, a progressive model of MS. SJL/J mice were subjected to EV treatment once the disease was established. We found that intravenous EV administration improved motor deficits, reduced brain atrophy, increased cell proliferation in the subventricular zone and decreased inflammatory infiltrates in the spinal cord in mice infected with TMEV. EV treatment was also capable of modulating neuroinflammation, given glial fibrillary acidic protein and Iba-1 staining were reduced in the brain, whereas myelin protein expression was increased. Changes in the morphology of microglial cells in the spinal cord suggest that EVs also modulate the activation state of microglia. The clear reduction in plasma cytokine levels, mainly in the Th1 and Th17 phenotypes, in TMEV mice treated with EVs confirms the immunomodulatory ability of intravenous EVs. According to our results, EV administration attenuates motor deficits through immunomodulatory actions, diminishing brain atrophy and promoting remyelination. Further studies are necessary to establish EV delivery as a possible therapy for the neurodegenerative phase of MS.
Collapse
Affiliation(s)
- Fernando Laso-García
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Jaime Ramos-Cejudo
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | | | - Laura Otero-Ortega
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Ana Feliú
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - MariCarmen Gómez-de Frutos
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Miriam Mecha
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| | - Carmen Guaza
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Cajal Institute, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|
9
|
Ingwersen J, De Santi L, Wingerath B, Graf J, Koop B, Schneider R, Hecker C, Schröter F, Bayer M, Engelke AD, Dietrich M, Albrecht P, Hartung HP, Annunziata P, Aktas O, Prozorovski T. Nimodipine confers clinical improvement in two models of experimental autoimmune encephalomyelitis. J Neurochem 2018; 146:86-98. [PMID: 29473171 DOI: 10.1111/jnc.14324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis is characterised by inflammatory neurodegeneration, with axonal injury and neuronal cell death occurring in parallel to demyelination. Regarding the molecular mechanisms responsible for demyelination and axonopathy, energy failure, aberrant expression of ion channels and excitotoxicity have been suggested to lead to Ca2+ overload and subsequent activation of calcium-dependent damage pathways. Thus, the inhibition of Ca2+ influx by pharmacological modulation of Ca2+ channels may represent a novel neuroprotective strategy in the treatment of secondary axonopathy. We therefore investigated the effects of the L-type voltage-gated calcium channel blocker nimodipine in two different models of mouse experimental autoimmune encephalomyelitis (EAE), an established experimental paradigm for multiple sclerosis. We show that preventive application of nimodipine (10 mg/kg per day) starting on the day of induction had ameliorating effects on EAE in SJL/J mice immunised with encephalitic myelin peptide PLP139-151 , specifically in late-stage disease. Furthermore, supporting these data, administration of nimodipine to MOG35-55 -immunised C57BL/6 mice starting at the peak of pre-established disease, also led to a significant decrease in disease score, indicating a protective effect on secondary CNS damage. Histological analysis confirmed that nimodipine attenuated demyelination, axonal loss and pathological axonal β-amyloid precursor protein accumulation in the cerebellum and spinal cord in the chronic phase of disease. Of note, we observed no effects of nimodipine on the peripheral immune response in EAE mice with regard to distribution, antigen-specific proliferation or activation patterns of lymphocytes. Taken together, our data suggest a CNS-specific effect of L-type voltage-gated calcium channel blockade to inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Lorenzo De Santi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Britta Wingerath
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jonas Graf
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Barbara Koop
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Friederike Schröter
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Mary Bayer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anna Dorothee Engelke
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Pasquale Annunziata
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tim Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
10
|
Gilli F, Royce DB, DiSano KD, Pachner AR. Pegylated interferon beta in the treatment of the Theiler's murine encephalomyelitis virus mouse model of multiple sclerosis. J Neuroimmunol 2017; 313:34-40. [DOI: 10.1016/j.jneuroim.2017.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 01/19/2023]
|
11
|
Modica CM, Schweser F, Sudyn ML, Bertolino N, Preda M, Polak P, Siebert DM, Krawiecki JC, Sveinsson M, Hagemeier J, Dwyer MG, Pol S, Zivadinov R. Effect of teriflunomide on cortex-basal ganglia-thalamus (CxBGTh) circuit glutamatergic dysregulation in the Theiler's Murine Encephalomyelitis Virus mouse model of multiple sclerosis. PLoS One 2017; 12:e0182729. [PMID: 28796815 PMCID: PMC5552032 DOI: 10.1371/journal.pone.0182729] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Pathology of gray matter is associated with development of physical and cognitive disability in patients with multiple sclerosis. In particular, glutamatergic dysregulation in the cortex-basal ganglia-thalamus (CxBGTh) circuit could be associated with decline in these behaviors. OBJECTIVES To investigate the effect of an immunomodulatory therapy (teriflunomide, Aubagio®) on changes of the CxBGTh loop in the Theiler's Murine Encephalomyelitis Virus, (TMEV) mouse model of MS. METHODS Forty-eight (48) mice were infected with TMEV, treated with teriflunomide (24) or control vehicle (24) and followed for 39 weeks. Mice were examined with MRS and volumetric MRI scans (0, 8, 26, and 39 weeks) in the cortex, basal ganglia and thalamus, using a 9.4T scanner, and with behavioral tests (0, 4, 8, 12, 17, 26, and 39 weeks). Within conditions, MRI measures were compared between two time points by paired samples t-test and across multiple time points by repeated measures ANOVA (rmANOVA), and between conditions by independent samples t-test and rmANOVA, respectively. Data were considered as significant at the p<0.01 level and as a trend at p<0.05 level. RESULTS In the thalamus, the teriflunomide arm exhibited trends toward decreased glutamate levels at 8 and 26 weeks compared to the control arm (p = 0.039 and p = 0.026), while the control arm exhibited a trend toward increased glutamate between 0 to 8 weeks (p = 0.045). In the basal ganglia, the teriflunomide arm exhibited a trend toward decreased glutamate earlier than the control arm, from 0 to 8 weeks (p = 0.011), resulting in decreased glutamate compared to the control arm at 8 weeks (p = 0.016). CONCLUSIONS Teriflunomide may reduce possible excitotoxicity in the thalamus and basal ganglia by lowering glutamate levels.
Collapse
Affiliation(s)
- Claire M Modica
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Ferdinand Schweser
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Translational Imaging Center, Clinical and Translational Science Institute, University at Buffalo, Buffalo, New York, United States of America
| | - Michelle L Sudyn
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Nicola Bertolino
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Marilena Preda
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Translational Imaging Center, Clinical and Translational Science Institute, University at Buffalo, Buffalo, New York, United States of America
| | - Paul Polak
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Danielle M Siebert
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Exercise Science, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York, United States of America
| | - Jacqueline C Krawiecki
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Department of Geology, University at Buffalo, Buffalo, New York, United States of America
| | - Michele Sveinsson
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Jesper Hagemeier
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Michael G Dwyer
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Suyog Pol
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - Robert Zivadinov
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, United States of America.,Translational Imaging Center, Clinical and Translational Science Institute, University at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
12
|
Huseby Kelcher AM, Atanga PA, Gamez JD, Cumba Garcia LM, Teclaw SJ, Pavelko KD, Macura SI, Johnson AJ. Brain atrophy in picornavirus-infected FVB mice is dependent on the H-2D b class I molecule. FASEB J 2017; 31:2267-2275. [PMID: 28188174 DOI: 10.1096/fj.201601055r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/23/2017] [Indexed: 01/09/2023]
Abstract
Brain atrophy is a common feature of numerous neurologic diseases in which the role of neuroinflammation remains ill-defined. In this study, we evaluated the contribution of major histocompatibility complex class I molecules to brain atrophy in Theiler's murine encephalomyelitis virus (TMEV)-infected transgenic FVB mice that express the Db class I molecule. FVB/Db and wild-type FVB mice were evaluated for changes in neuroinflammation, virus clearance, neuropathology, and development of brain atrophy via T2-weighted MRI and subsequent 3-dimensional volumetric analysis. Significant brain atrophy and hippocampal neuronal loss were observed in TMEV-infected FVB/Db mice, but not in wild-type FVB mice. Brain atrophy was observed at 1 mo postinfection and persisted through the 4-mo observation period. Of importance, virus-infected FVB/Db mice elicited a strong CD8 T-cell response toward the immunodominant Db-restricted TMEV-derived peptide, VP2121-130, and cleared TMEV from the CNS. In addition, immunofluorescence revealed CD8 T cells near virus-infected neurons; therefore, we hypothesize that class I restricted CD8 T-cell responses promote development of brain atrophy. This model provides an opportunity to analyze the contribution of immune cells to brain atrophy in a system where persistent virus infection and demyelination are not factors in long-term neuropathology.-Huseby Kelcher, A. M., Atanga, P. A., Gamez, J. D., Cumba Garcia, L. M., Teclaw, S. J., Pavelko, K. D., Macura, S. I., Johnson. A. J. Brain atrophy in picornavirus-infected FVB mice is dependent on the H-2Db class I molecule.
Collapse
Affiliation(s)
- April M Huseby Kelcher
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Pascal A Atanga
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey D Gamez
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Luz M Cumba Garcia
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA.,Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Slobodan I Macura
- Nuclear Magnetic Resonance Core Facility, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA; .,Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
13
|
Willenbring RC, Jin F, Hinton DJ, Hansen M, Choi DS, Pavelko KD, Johnson AJ. Modulatory effects of perforin gene dosage on pathogen-associated blood-brain barrier (BBB) disruption. J Neuroinflammation 2016; 13:222. [PMID: 27576583 PMCID: PMC5006384 DOI: 10.1186/s12974-016-0673-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/17/2016] [Indexed: 11/12/2022] Open
Abstract
Background CD8 T cell-mediated blood-brain barrier (BBB) disruption is dependent on the effector molecule perforin. Human perforin has extensive single nucleotide variants (SNVs), the significance of which is not fully understood. These SNVs can result in reduced, but not ablated, perforin activity or expression. However, complete loss of perforin expression or activity results in the lethal disease familial hemophagocytic lymphohistiocytosis type 2 (FHL 2). In this study, we address the hypothesis that a single perforin allele can alter the severity of BBB disruption in vivo using a well-established model of CNS vascular permeability in C57Bl/6 mice. The results of this study provide insight into the significance of perforin SNVs in the human population. Methods We isolated the effect a single perforin allele has on CNS vascular permeability through the use of perforin-heterozygous (perforin+/−) C57BL/6 mice in the peptide-induced fatal syndrome (PIFS) model of immune-mediated BBB disruption. Seven days following Theiler’s murine encephalomyelitis virus (TMEV) CNS infection, neuroinflammation and TMEV viral control were assessed through flow cytometric analysis and quantitative real-time PCR of the viral genome, respectively. Following immune-mediated BBB disruption, gadolinium-enhanced T1-weighted MRI, with 3D volumetric analysis, and confocal microscopy were used to define CNS vascular permeability. Finally, the open field behavior test was used to assess locomotor activity of mice following immune-mediated BBB disruption. Results Perforin-null mice had negligible CNS vascular permeability. Perforin-WT mice have extensive CNS vascular permeability. Interestingly, perforin-heterozygous mice had an intermediate level of CNS vascular permeability as measured by both gadolinium-enhanced T1-weighted MRI and fibrinogen leakage in the brain parenchyma. Differences in BBB disruption were not a result of increased CNS immune infiltrate. Additionally, TMEV was controlled in a perforin dose-dependent manner. Furthermore, a single perforin allele is sufficient to induce locomotor deficit during immune-mediated BBB disruption. Conclusions Perforin modulates BBB disruption in a dose-dependent manner. This study demonstrates a potentially advantageous role for decreased perforin expression in reducing BBB disruption. This study also provides insight into the effect SNVs in a single perforin allele could have on functional deficit in neurological disease.
Collapse
Affiliation(s)
- Robin C Willenbring
- Mayo Graduate School, Mayo Clinic, Rochester, MN, USA.,Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - David J Hinton
- Mayo Graduate School, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Mike Hansen
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Doo-Sup Choi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, USA. .,Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Gilli F, Chen X, Pachner AR, Gimi B. High-Resolution Diffusion Tensor Spinal Cord MRI Measures as Biomarkers of Disability Progression in a Rodent Model of Progressive Multiple Sclerosis. PLoS One 2016; 11:e0160071. [PMID: 27467829 PMCID: PMC4965026 DOI: 10.1371/journal.pone.0160071] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/13/2016] [Indexed: 01/02/2023] Open
Abstract
Disease in the spinal cord is a major component of disability in multiple sclerosis, yet current techniques of imaging spinal cord injury are insensitive and nonspecific. This study seeks to remove this major impediment to research in multiple sclerosis and other spinal cord diseases by identifying reliable biomarkers of disability progression using diffusion tensor imaging (DTI), a magnetic resonance imaging technique, to evaluate the spinal cord in a model of multiple sclerosis, i.e. the Theiler’s Murine Encephalitis Virus-Induced Demyelinating Disease (TMEV-IDD). Mice with TMEV-IDD with varying levels of clinical disease were imaged using a 9.4T small animal MRI scanner. Axial diffusivity, radial diffusivity, and fractional anisotropy were calculated. Disability was assessed periodically using Rotarod assay and data were expressed as a neurological function index. Correlation was performed between DTI measurements and disability scores. TMEV-IDD mice displayed significant increased neurological deficits over time when compared with controls (p<0.0001). Concurrently, the values of fractional anisotropy and axial diffusivity were both decreased compared to controls (both p<0.0001), while radial diffusivity was increased (p<0.0001). Overall, fractional anisotropy changes were larger in white matter than in grey matter and differences were more pronounced in the ventral region. Lower disability scores were associated with decreased fractional anisotropy values measured in the ventral (r = 0.68; p<0.0001) and ventral-lateral (r = 0.70; p<0.0001) regions of the white matter. These data demonstrate that DTI measures of the spinal cord contribute to strengthening the association between neuroradiological markers and clinical disability, and support the use of DTI measures in spinal cord imaging in MS patients.
Collapse
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| | - Xi Chen
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Andrew R. Pachner
- Department of Neurology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Barjor Gimi
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
15
|
Paz Soldán MM, Raman MR, Gamez JD, Lohrey AK, Chen Y, Pirko I, Johnson AJ. Correlation of Brain Atrophy, Disability, and Spinal Cord Atrophy in a Murine Model of Multiple Sclerosis. J Neuroimaging 2015; 25:595-9. [PMID: 25893491 DOI: 10.1111/jon.12250] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 03/06/2015] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Disability progression in multiple sclerosis (MS) remains incompletely understood. Unlike lesional measures, central nervous system atrophy has a strong correlation with disability. Theiler's murine encephalomyelitis virus infection in SJL/J mice is an established model of progressive MS. We utilized in vivo MRI to quantify brain and spinal cord atrophy in this model and analyzed the temporal relationship between atrophy and disability. METHODS Infected and control mice were followed for 12 months. Disability was assessed periodically using rotarod assay. Volumetric MRI datasets were acquired at 7 Tesla. Ventricular volume and C4-5 spinal cord cross-sectional area measurements were performed using Analyze 10. RESULTS At 3 months, brain atrophy reached statistical significance (P = .005). In contrast, disability did not differ until 4 months post-infection (P = .0005). Cord atrophy reached significance by 9 months (P = 0.009). By 12 months, brain atrophy resulted in 111.8% increased ventricular volume (P = .00003), while spinal cord cross-sectional area was 25.6% reduced (P = .001) among cases. CONCLUSIONS Our results suggest that significant brain atrophy precedes and predicts the development of disability, while spinal cord atrophy occurs late and correlates with severe disability. The observed temporal relationship establishes a framework for mechanisms of disability progression and enables further investigations of their underlying substrate.
Collapse
Affiliation(s)
| | - Mekala R Raman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Anne K Lohrey
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio
| | - Yi Chen
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio
| | - Istvan Pirko
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
16
|
Perforin competent CD8 T cells are sufficient to cause immune-mediated blood-brain barrier disruption. PLoS One 2014; 9:e111401. [PMID: 25337791 PMCID: PMC4206459 DOI: 10.1371/journal.pone.0111401] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 10/02/2014] [Indexed: 12/02/2022] Open
Abstract
Numerous neurological disorders are characterized by central nervous system (CNS) vascular permeability. However, the underlying contribution of inflammatory-derived factors leading to pathology associated with blood-brain barrier (BBB) disruption remains poorly understood. In order to address this, we developed an inducible model of BBB disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide induced fatal syndrome (PIFS) model is initiated by virus-specific CD8 T cells and results in severe CNS vascular permeability and death in the C57BL/6 mouse strain. While perforin is required for BBB disruption, the cellular source of perforin has remained unidentified. In addition to CD8 T cells, various innate immune cells also express perforin and therefore could also contribute to BBB disruption. To investigate this, we isolated the CD8 T cell as the sole perforin-expressing cell type in the PIFS model through adoptive transfer techniques. We determined that C57BL/6 perforin−/− mice reconstituted with perforin competent CD8 T cells and induced to undergo PIFS exhibited: 1) heightened CNS vascular permeability, 2) increased astrocyte activation as measured by GFAP expression, and 3) loss of linear organization of BBB tight junction proteins claudin-5 and occludin in areas of CNS vascular permeability when compared to mock-treated controls. These results are consistent with the characteristics associated with PIFS in perforin competent mice. Therefore, CD8 T cells are sufficient as a sole perforin-expressing cell type to cause BBB disruption in the PIFS model.
Collapse
|
17
|
Martinez NE, Karlsson F, Sato F, Kawai E, Omura S, Minagar A, Grisham MB, Tsunoda I. Protective and detrimental roles for regulatory T cells in a viral model for multiple sclerosis. Brain Pathol 2014; 24:436-51. [PMID: 24417588 PMCID: PMC4097993 DOI: 10.1111/bpa.12119] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/07/2014] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) has been proposed to be an immune-mediated disease in the central nervous system (CNS) that can be triggered by virus infections. In Theiler's murine encephalomyelitis virus (TMEV) infection, during the first week (acute stage), mice develop polioencephalomyelitis. After 3 weeks (chronic stage), mice develop immune-mediated demyelination with virus persistence, which has been used as a viral model for MS. Regulatory T cells (Tregs) can suppress inflammation, and have been suggested to be protective in immune-mediated diseases, including MS. However, in virus-induced inflammatory demyelination, although Tregs can suppress inflammation, preventing immune-mediated pathology, Tregs may also suppress antiviral immune responses, leading to more active viral replication and/or persistence. To determine the role and potential translational usage of Tregs in MS, we treated TMEV-infected mice with ex vivo generated induced Tregs (iTregs) on day 0 (early) or during the chronic stage (therapeutic). Early treatment worsened clinical signs during acute disease. The exacerbation of acute disease was associated with increased virus titers and decreased immune cell recruitment in the CNS. Therapeutic iTreg treatment reduced inflammatory demyelination during chronic disease. Immunologically, iTreg treatment increased interleukin-10 production from B cells, CD4(+) T cells and dendritic cells, which may contribute to the decreased CNS inflammation.
Collapse
Affiliation(s)
- Nicholas E. Martinez
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLA
| | - Fridrik Karlsson
- Department of Molecular and Cellular PhysiologyLouisiana State University Health Sciences CenterShreveportLA
| | - Fumitaka Sato
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLA
| | - Eiichiro Kawai
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLA
| | - Seiichi Omura
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLA
| | - Alireza Minagar
- Department of NeurologyLouisiana State University Health Sciences CenterShreveportLA
| | - Matthew. B. Grisham
- Department of Immunology and Molecular MicrobiologyTexas Tech University Health Sciences CenterLubbockTX
| | - Ikuo Tsunoda
- Department of Microbiology and ImmunologyCenter for Molecular and Tumor VirologyLouisiana State University Health Sciences CenterShreveportLA
| |
Collapse
|
18
|
Alomair OI, Smith MT, Brereton IM, Galloway GJ, Kurniawan ND. Current developments in MRI for assessing rodent models of multiple sclerosis. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.14.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: MRI is a key radiological imaging technique that plays an important role in the diagnosis and characterization of heterogeneous multiple sclerosis (MS) lesions. Various MRI methodologies such as conventional T 1/T 2 contrast, contrast agent enhancement, diffusion-weighted imaging, magnetization transfer imaging and susceptibility weighted imaging have been developed to determine the severity of MS pathology, including demyelination/remyelination and brain connectivity impairment from axonal loss. The broad spectrum of MS pathology manifests in diverse patient MRI presentations and affects the accuracy of patient diagnosis. To study specific pathological aspects of the disease, rodent models such as experimental autoimmune encephalomyelitis, virus-induced and toxin-induced demyelination have been developed. This review aims to present key developments in MRI methodology for better characterization of rodent models of MS.
Collapse
Affiliation(s)
- Othman I Alomair
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
- College of Applied Medical Science, King Saud University, Riyadh, Saudi Arabia
| | - Maree T Smith
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Integrated Preclinical Drug Development, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian M Brereton
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Graham J Galloway
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| | - Nyoman D Kurniawan
- Centre for Advanced Imaging, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
19
|
Nathoo N, Yong VW, Dunn JF. Understanding disease processes in multiple sclerosis through magnetic resonance imaging studies in animal models. NEUROIMAGE-CLINICAL 2014; 4:743-56. [PMID: 24936425 PMCID: PMC4053634 DOI: 10.1016/j.nicl.2014.04.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 01/11/2023]
Abstract
There are exciting new advances in multiple sclerosis (MS) resulting in a growing understanding of both the complexity of the disorder and the relative involvement of grey matter, white matter and inflammation. Increasing need for preclinical imaging is anticipated, as animal models provide insights into the pathophysiology of the disease. Magnetic resonance (MR) is the key imaging tool used to diagnose and to monitor disease progression in MS, and thus will be a cornerstone for future research. Although gadolinium-enhancing and T2 lesions on MRI have been useful for detecting MS pathology, they are not correlative of disability. Therefore, new MRI methods are needed. Such methods require validation in animal models. The increasing necessity for MRI of animal models makes it critical and timely to understand what research has been conducted in this area and what potential there is for use of MRI in preclinical models of MS. Here, we provide a review of MRI and magnetic resonance spectroscopy (MRS) studies that have been carried out in animal models of MS that focus on pathology. We compare the MRI phenotypes of animals and patients and provide advice on how best to use animal MR studies to increase our understanding of the linkages between MR and pathology in patients. This review describes how MRI studies of animal models have been, and will continue to be, used in the ongoing effort to understand MS. MRI studies of pathology in various animal models of MS are reviewed. MRI phenotypes in animal models of MS and MS patients are compared. Animal MRI can increase understanding of links between MR and pathology in patients.
Collapse
Affiliation(s)
- Nabeela Nathoo
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
| | - V. Wee Yong
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F. Dunn
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Experimental Imaging Centre, University of Calgary, Calgary, Alberta, Canada
- Corresponding author at: Department of Radiology, University of Calgary, 3330 Hospital Drive, N.W., Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
20
|
Zhou L, Miranda-Saksena M, Saksena NK. Viruses and neurodegeneration. Virol J 2013; 10:172. [PMID: 23724961 PMCID: PMC3679988 DOI: 10.1186/1743-422x-10-172] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 05/20/2013] [Indexed: 11/10/2022] Open
Abstract
Neurodegenerative diseases (NDs) are chronic degenerative diseases of the central nervous system (CNS), which affect 37 million people worldwide. As the lifespan increases, the NDs are the fourth leading cause of death in the developed countries and becoming increasingly prevalent in developing countries. Despite considerable research, the underlying mechanisms remain poorly understood. Although the large majority of studies do not show support for the involvement of pathogenic aetiology in classical NDs, a number of emerging studies show support for possible association of viruses with classical neurodegenerative diseases in humans. Space does not permit for extensive details to be discussed here on non-viral-induced neurodegenerative diseases in humans, as they are well described in literature.Viruses induce alterations and degenerations of neurons both directly and indirectly. Their ability to attack the host immune system, regions of nervous tissue implies that they can interfere with the same pathways involved in classical NDs in humans. Supporting this, many similarities between classical NDs and virus-mediated neurodegeneration (non-classical) have been shown at the anatomic, sub-cellular, genomic and proteomic levels suggesting that viruses can explain neurodegenerative disorders mechanistically. The main objective of this review is to provide readers a detailed snapshot of similarities viral and non-viral neurodegenerative diseases share, so that mechanistic pathways of neurodegeneration in human NDs can be clearly understood. Viruses can guide us to unveil these pathways in human NDs. This will further stimulate the birth of new concepts in the biological research, which is needed for gaining deeper insights into the treatment of human NDs and delineate mechanisms underlying neurodegeneration.
Collapse
Affiliation(s)
- Li Zhou
- Retroviral Genetics Division, Center for Virus Research, Westmead Millennium Institute, Westmead Hospital, The University of Sydney, Westmead NSW 2145, Sydney Australia
| | | | | |
Collapse
|
21
|
Bagnato F, Hametner S, Welch EB. Visualizing iron in multiple sclerosis. Magn Reson Imaging 2013; 31:376-84. [PMID: 23347601 PMCID: PMC4776767 DOI: 10.1016/j.mri.2012.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 10/30/2012] [Accepted: 11/10/2012] [Indexed: 01/19/2023]
Abstract
Magnetic resonance imaging (MRI) protocols that are designed to be sensitive to iron typically take advantage of (1) iron effects on the relaxation of water protons and/or (2) iron-induced local magnetic field susceptibility changes. Increasing evidence sustains the notion that imaging iron in brain of patients with multiple sclerosis (MS) may add some specificity toward the identification of the disease pathology. The present review summarizes currently reported in vivo and post mortem MRI evidence of (1) iron detection in white matter and gray matter of MS brains, (2) pathological and physiological correlates of iron as disclosed by imaging and (3) relations between iron accumulation and disease progression as measured by clinical metrics.
Collapse
Affiliation(s)
- Francesca Bagnato
- Radiology Department, Vanderbilt University, Institute of Imaging Science, Nashville, TN 37232, USA. :
| | | | | |
Collapse
|
22
|
Mecha M, Carrillo-Salinas FJ, Mestre L, Feliú A, Guaza C. Viral models of multiple sclerosis: neurodegeneration and demyelination in mice infected with Theiler's virus. Prog Neurobiol 2013; 101-102:46-64. [PMID: 23201558 PMCID: PMC7117056 DOI: 10.1016/j.pneurobio.2012.11.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/30/2012] [Accepted: 11/12/2012] [Indexed: 11/02/2022]
Abstract
Multiple sclerosis (MS) is a complex inflammatory disease of unknown etiology that affects the central nervous system (CNS) white matter, and for which no effective cure exists. Indeed, whether the primary event in MS pathology affects myelin or axons of the CNS remains unclear. Animal models are necessary to identify the immunopathological mechanisms involved in MS and to develop novel therapeutic and reparative approaches. Specifically, viral models of chronic demyelination and axonal damage have been used to study the contribution of viruses in human MS, and they have led to important breakthroughs in our understanding of MS pathology. The Theiler's murine encephalomyelitis virus (TMEV) model is one of the most commonly used MS models, although other viral models are also used, including neurotropic strains of mouse hepatitis virus (MHV) that induce chronic inflammatory demyelination with similar histological features to those observed in MS. This review will discuss the immunopathological mechanisms involved in TMEV-induced demyelinating disease (TMEV-IDD). The TMEV model reproduces a chronic progressive disease due to the persistence of the virus for the entire lifespan in susceptible mice. The evolution and significance of the axonal damage and neuroinflammation, the importance of epitope spread from viral to myelin epitopes, the presence of abortive remyelination and the existence of a brain pathology in addition to the classical spinal cord demyelination, are some of the findings that will be discussed in the context of this TMEV-IDD model. Despite their limitations, viral models remain an important tool to study the etiology of MS, and to understand the clinical and pathological variability associated with this disease.
Collapse
Key Words
- ab, antibody
- ag, antigen
- apc, antigen presenting cell
- bbb, blood–brain barrier
- cns, central nervous system
- cox-2, cyclooxygenase-2
- ctl, cytotoxic t lymphocytes
- dpi, days post-infection
- da, daniels strain of theiler's virus
- eae, experimental autoimmune encephalomyelitis
- galc, galactocerebroside
- mbp, myelin basic protein
- mnc, mononuclear cells
- mhc, major histocompatibility complex
- mhv, mouse hepatitis virus
- mog, myelin oligodendrocyte glycoprotein
- ms, multiple sclerosis
- naa, n-acetylaspartate
- no, nitric oxide
- pcr, polymerase chain reaction
- plp, myelin proteolipid protein
- pprs, pattern recognition receptors
- sfv, semliki forest virus
- sv, sindbis virus
- tmev, theiler's murine encephalomyelitis virus
- tmev-idd, theiler's murine encephalomyelitis virus-induced demyelinating disease
- tregs, regulatory t cells
- theiler's virus
- multiple sclerosis
- demyelination
- axonal damage
- neuroinflammation
- spinal cord pathology
- brain pathology
Collapse
Affiliation(s)
| | | | | | | | - Carmen Guaza
- Neuroimmunology Group, Functional and System Neurobiology Department, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Avda Dr Arce 37, 28002 Madrid, Spain
| |
Collapse
|
23
|
Cognitive impairment and optic nerve axonal loss in patients with clinically isolated syndrome. Clin Neurol Neurosurg 2012. [PMID: 23182176 DOI: 10.1016/j.clineuro.2012.10.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To investigate cognitive impairment, to assess optical nerve axonal loss, and to determinate whether there is correlation between optical nerve axonal loss and cognition impairment in Clinically Isolated Syndrome (CIS). METHODS Fifteen CIS patients and 15 controls were submitted to Wechsler memory scale, Rey Auditory Verbal Learning, Rey Complex Figure, Paced Auditory Serial Addition, Digit Span, verbal fluency, stroop color, D2, and Digit Symbol tests. CIS patients were evaluated by optical coherence tomography (OCT) (23 eyes). RESULTS CIS patients had worse performance in Paced Auditory Serial Addition Test (PASAT) 2 seconds (P=0.009) and fluency tests (P=0.0038). Optical nerve axonal loss was found more frequently in eyes with previous optic neuritis (ON) (85.7%) than in those without previous ON (21.7%) (P=0.0146). There were no significant correlations between optical nerve axonal loss and cognitive findings. CONCLUSIONS CIS patients had worse cognitive performance than controls. OCT can detect axonal loss resulting from optical neuritis and subclinical axonal loss in eyes without previous optical neuritis. Optical nerve axonal loss was not correlated with cognition.
Collapse
|
24
|
Johnson HL, Chen Y, Jin F, Hanson LM, Gamez JD, Pirko I, Johnson AJ. CD8 T cell-initiated blood-brain barrier disruption is independent of neutrophil support. THE JOURNAL OF IMMUNOLOGY 2012; 189:1937-45. [PMID: 22772449 DOI: 10.4049/jimmunol.1200658] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Blood-brain barrier (BBB) disruption is a common feature of numerous neurologic disorders. A fundamental question in these diseases is the extent inflammatory immune cells contribute to CNS vascular permeability. We have previously shown that CD8 T cells play a critical role in initiating BBB disruption in the peptide-induced fatal syndrome model developed by our laboratory. However, myelomonocytic cells such as neutrophils have also been implicated in promoting CNS vascular permeability and functional deficit in murine models of neuroinflammatory disease. For this reason, we evaluated neutrophil depletion in a murine model of CD8 T cell-initiated BBB disruption by employing traditionally used anti-granulocyte receptor-1 mAb RB6-8C5 and Ly-6G-specific mAb 1A8. We report that CNS-infiltrating antiviral CD8 T cells express high levels of granulocyte receptor-1 protein and are depleted by treatment with RB6-8C5. Mice treated with RB6-8C5, but not 1A8, display: 1) intact BBB tight junction proteins; 2) reduced CNS vascular permeability visible by gadolinium-enhanced T1-weighted magnetic resonance imaging; and 3) preservation of motor function. These studies demonstrate that traditional methods of neutrophil depletion with RB6-8C5 are broadly immune ablating. Our data also provide evidence that CD8 T cells initiate disruption of BBB tight junction proteins and CNS vascular permeability in the absence of neutrophil support.
Collapse
Affiliation(s)
- Holly L Johnson
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Kummerfeld M, Seehusen F, Klein S, Ulrich R, Kreutzer R, Gerhauser I, Herder V, Baumgärtner W, Beineke A. Periventricular demyelination and axonal pathology is associated with subependymal virus spread in a murine model for multiple sclerosis. Intervirology 2012; 55:401-16. [PMID: 22538300 DOI: 10.1159/000336563] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/02/2012] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Theiler's murine encephalomyelitis virus (TMEV) infection of mice is a widely used animal model for demyelinating disorders, such as multiple sclerosis (MS). The aim of the present study was to identify topographical differences of TMEV spread and demyelination in the brain of experimentally infected susceptible SJL/J mice and resistant C57BL/6 mice. METHODS Demyelination was confirmed by Luxol fast blue and cresyl violet staining and axonal damage by neurofilament-specific and β-amyloid precursor protein-specific immunohistochemistry. Viral dissemination within the central nervous system (CNS) was quantified by immunohistochemistry and in situ hybridization. Further, the phenotype of infected cells was determined by confocal laser scanning microscopy. RESULTS An early transient infection of periventricular cells followed by demyelination and axonopathies around the fourth ventricle in SJL/J mice was noticed. Periventricular and brain stem demyelination was associated with a predominant infection of microglia/macrophages and oligodendrocytes. CONCLUSIONS Summarized, the demonstration of ependymal infection and subjacent spread into the brain parenchyma as well as regional virus clearance despite ongoing demyelination and axonal damage in other CNS compartments allows new insights into TME pathogenesis. This novel aspect of TMEV CNS interaction will enhance the understanding of region-specific susceptibilities to injury and regenerative capacities of the brain in this MS model.
Collapse
Affiliation(s)
- Maren Kummerfeld
- Department of Pathology, University of Veterinary Medicine Hanover, Hanover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Johnson HL, Chen Y, Suidan GL, McDole JR, Lohrey AK, Hanson LM, Jin F, Pirko I, Johnson AJ. A hematopoietic contribution to microhemorrhage formation during antiviral CD8 T cell-initiated blood-brain barrier disruption. J Neuroinflammation 2012; 9:60. [PMID: 22452799 PMCID: PMC3350446 DOI: 10.1186/1742-2094-9-60] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 03/27/2012] [Indexed: 01/09/2023] Open
Abstract
Background The extent to which susceptibility to brain hemorrhage is derived from blood-derived factors or stromal tissue remains largely unknown. We have developed an inducible model of CD8 T cell-initiated blood-brain barrier (BBB) disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide-induced fatal syndrome (PIFS) model results in severe central nervous system (CNS) vascular permeability and death in the C57BL/6 mouse strain, but not in the 129 SvIm mouse strain, despite the two strains' having indistinguishable CD8 T-cell responses. Therefore, we hypothesize that hematopoietic factors contribute to susceptibility to brain hemorrhage, CNS vascular permeability and death following induction of PIFS. Methods PIFS was induced by intravenous injection of VP2121-130 peptide at 7 days post-TMEV infection. We then investigated brain inflammation, astrocyte activation, vascular permeability, functional deficit and microhemorrhage formation using T2*-weighted magnetic resonance imaging (MRI) in C57BL/6 and 129 SvIm mice. To investigate the contribution of hematopoietic cells in this model, hemorrhage-resistant 129 SvIm mice were reconstituted with C57BL/6 or autologous 129 SvIm bone marrow. Gadolinium-enhanced, T1-weighted MRI was used to visualize the extent of CNS vascular permeability after bone marrow transfer. Results C57BL/6 and 129 SvIm mice had similar inflammation in the CNS during acute infection. After administration of VP2121-130 peptide, however, C57BL/6 mice had increased astrocyte activation, CNS vascular permeability, microhemorrhage formation and functional deficits compared to 129 SvIm mice. The 129 SvIm mice reconstituted with C57BL/6 but not autologous bone marrow had increased microhemorrhage formation as measured by T2*-weighted MRI, exhibited a profound increase in CNS vascular permeability as measured by three-dimensional volumetric analysis of gadolinium-enhanced, T1-weighted MRI, and became moribund in this model system. Conclusion C57BL/6 mice are highly susceptible to microhemorrhage formation, severe CNS vascular permeability and morbidity compared to the 129 SvIm mouse. This susceptibility is transferable with the bone marrow compartment, demonstrating that hematopoietic factors are responsible for the onset of brain microhemorrhage and vascular permeability in immune-mediated fatal BBB disruption.
Collapse
|
27
|
Meeusen JW, Klein CJ, Pirko I, Haselkorn KE, Kryzer TJ, Pittock SJ, Lachance DH, Dyck PJ, Lennon VA. Potassium channel complex autoimmunity induced by inhaled brain tissue aerosol. Ann Neurol 2012; 71:417-26. [PMID: 22451206 PMCID: PMC3315155 DOI: 10.1002/ana.22674] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To test the hypothesis that autoimmunity induced by inhalation of aerosolized brain tissue caused outbreaks of sensory-predominant polyradiculoneuropathy among swine abattoir employees in the Midwestern United States. METHODS Mice were exposed intranasally, 5 days per week, to liquefied brain tissue. Serum from exposed mice, patients, and unaffected abattoir employees were analyzed for clinically pertinent neural autoantibodies. RESULTS Patients, coworkers, and mice exposed to liquefied brain tissue had an autoantibody profile dominated by neural cation channel immunoglobulin Gs (IgGs). The most compelling link between patients and exposed mice was magnetic resonance imaging (MRI) evidence of grossly swollen spinal nerve roots. Autoantibody responses in patients and mice were dose-dependent and declined after antigen exposure ceased. Autoantibodies detected most frequently, and at high levels, bound to detergent-solubilized macromolecular complexes containing neuronal voltage-gated potassium channels ligated with a high affinity Kv1 channel antagonist, 125I-α-dendrotoxin. Exposed mice exhibited a behavioral phenotype consistent with potassium channel dysfunction recognized in drosophila with mutant ("shaker") channels: reduced sensitivity to isoflurane-induced anesthesia. Pathological and electrophysiological findings in patients supported peripheral nerve hyperexcitability over destructive axonal loss. The pain-predominant symptoms were consistent with sensory nerve hyperexcitability. INTERPRETATION Our observations establish that inhaled neural antigens readily induce neurological autoimmunity and identify voltage-gated potassium channel complexes as a major immunogen.
Collapse
Affiliation(s)
- Jeffrey W. Meeusen
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Christopher J. Klein
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Istvan Pirko
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Keegan E. Haselkorn
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Thomas J. Kryzer
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Sean J. Pittock
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Daniel H. Lachance
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - P. James Dyck
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| | - Vanda A. Lennon
- Department of Laboratory Medicine and Pathology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
- Department of Laboratory Neurology and College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905
| |
Collapse
|
28
|
CMV infection attenuates the disease course in a murine model of multiple sclerosis. PLoS One 2012; 7:e32767. [PMID: 22393447 PMCID: PMC3290597 DOI: 10.1371/journal.pone.0032767] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 01/30/2012] [Indexed: 11/19/2022] Open
Abstract
Recent evidence in multiple sclerosis (MS) suggests that active CMV infection may result in more benign clinical disease. The goal of this pilot study was to determine whether underlying murine CMV (MCMV) infection affects the course of the Theiler's murine encephalitis virus (TMEV) induced murine model of MS. A group of eight TMEV-infected mice were co-infected with MCMV at 2 weeks prior to TMEV infection while a second group of TMEV-infected mice received MCMV two weeks post TMEV. We also used 2 control groups, where at the above time points MCMV was replaced with PBS. Outcome measures included (1) monthly monitoring of disability via rotarod for 8 months; (2) in vivo MRI for brain atrophy studies and (3) FACS analysis of brain infiltrating lymphocytes at 8 months post TMEV infection. Co-infection with MCMV influenced the disease course in mice infected prior to TMEV infection. In this group, rotarod detectable motor performance was significantly improved starting 3 months post-infection and beyond (p≤0.024). In addition, their brain atrophy was close to 30% reduced at 8 months, but this was only present as a trend due to low power (p = 0.19). A significant reduction in the proportion of brain infiltrating CD3+ cells was detected in this group (p = 0.026), while the proportion of CD45+ Mac1+ cells significantly increased (p = 0.003). There was also a strong trend for a reduced proportion of CD4+ cells (p = 0.17) while CD8 and B220+ cell proportion did not change. These findings support an immunomodulatory effect of MCMV infection in this MS model. Future studies in this co-infection model will provide insight into mechanisms which modulate the development of demyelination and may be utilized for the development of novel therapeutic strategies.
Collapse
|
29
|
Pirko I, Chen Y, Lohrey AK, McDole J, Gamez JD, Allen KS, Pavelko KD, Lindquist DM, Dunn RS, Macura SI, Johnson AJ. Contrasting roles for CD4 vs. CD8 T-cells in a murine model of virally induced "T1 black hole" formation. PLoS One 2012; 7:e31459. [PMID: 22348089 PMCID: PMC3278445 DOI: 10.1371/journal.pone.0031459] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 01/08/2012] [Indexed: 12/31/2022] Open
Abstract
MRI is sensitive to tissue pathology in multiple sclerosis (MS); however, most lesional MRI findings have limited correlation with disability. Chronic T1 hypointense lesions or "T1 black holes" (T1BH), observed in a subset of MS patients and thought to represent axonal damage, show moderate to strong correlation with disability. The pathogenesis of T1BH remains unclear. We previously reported the first and as of yet only model of T1BH formation in the Theiler's murine encephalitis virus induced model of acute CNS neuroinflammation induced injury, where CD8 T-cells are critical mediators of axonal damage and related T1BH formation. The purpose of this study was to further analyze the role of CD8 and CD4 T-cells through adoptive transfer experiments and to determine if the relevant CD8 T-cells are classic epitope specific lymphocytes or different subsets. C57BL/6 mice were used as donors and RAG-1 deficient mice as hosts in our adoptive transfer experiments. In vivo 3-dimensional MRI images were acquired using a 7 Tesla small animal MRI system. For image analysis, we used semi-automated methods in Analyze 9.1; transfer efficiency was monitored using FACS of brain infiltrating lymphocytes. Using a peptide depletion method, we demonstrated that the majority of CD8 T-cells are classic epitope specific cytotoxic cells. CD8 T-cell transfer successfully restored the immune system's capability to mediate T1BH formation in animals that lack adaptive immune system, whereas CD4 T-cell transfer results in an attenuated phenotype with significantly less T1BH formation. These findings demonstrate contrasting roles for these cell types, with additional evidence for a direct pathogenic role of CD8 T-cells in our model of T1 black hole formation.
Collapse
Affiliation(s)
- Istvan Pirko
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Yi Chen
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Anne K. Lohrey
- Department of Neurology, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jeremiah McDole
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Jeffrey D. Gamez
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kathleen S. Allen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kevin D. Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Diana M. Lindquist
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - R. Scott Dunn
- Imaging Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Slobodan I. Macura
- Department of Biochemistry, NMR Core Facility, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
30
|
MacKenzie-Graham A, Rinek GA, Avedisian A, Gold SM, Frew AJ, Aguilar C, Lin DR, Umeda E, Voskuhl RR, Alger JR. Cortical atrophy in experimental autoimmune encephalomyelitis: in vivo imaging. Neuroimage 2011; 60:95-104. [PMID: 22182769 DOI: 10.1016/j.neuroimage.2011.11.099] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 11/13/2011] [Accepted: 11/30/2011] [Indexed: 01/16/2023] Open
Abstract
There are strong correlations between cortical atrophy observed by MRI and clinical disability and disease duration in multiple sclerosis (MS). The objective of this study was to evaluate the progression of cortical atrophy over time in vivo in experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model for MS. Volumetric changes in brains of EAE mice and matched healthy controls were quantified by collecting high-resolution T2-weighted magnetic resonance images in vivo and labeling anatomical structures on the images. In vivo scanning permitted us to evaluate brain structure volumes in individual animals over time and we observed that though brain atrophy progressed differently in each individual animal, all mice with EAE demonstrated significant atrophy in whole brain, cerebral cortex, and whole cerebellum compared to normal controls. Furthermore, we found a strong correlation between cerebellar atrophy and cumulative disease score in mice with EAE. Ex vivo MRI showed a significant decrease in brain and cerebellar volume and a trend that did not reach significance in cerebral cortex volume in mice with EAE compared to controls. Cross modality correlations revealed a significant association between neuronal loss on neuropathology and in vivo atrophy of the cerebral cortex by neuroimaging. These results demonstrate that longitudinal in vivo imaging is more sensitive to changes that occur in neurodegenerative disease models than cross-sectional ex vivo imaging. This is the first report of progressive cortical atrophy in vivo in a mouse model of MS.
Collapse
|
31
|
Williams R, Buchheit CL, Berman NEJ, LeVine SM. Pathogenic implications of iron accumulation in multiple sclerosis. J Neurochem 2011; 120:7-25. [PMID: 22004421 DOI: 10.1111/j.1471-4159.2011.07536.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Iron, an essential element used for a multitude of biochemical reactions, abnormally accumulates in the CNS of patients with multiple sclerosis (MS). The mechanisms of abnormal iron deposition in MS are not fully understood, nor do we know whether these deposits have adverse consequences, that is, contribute to pathogenesis. With some exceptions, excess levels of iron are represented concomitantly in multiple deep gray matter structures often with bilateral representation, whereas in white matter, pathological iron deposits are usually located at sites of inflammation that are associated with veins. These distinct spatial patterns suggest disparate mechanisms of iron accumulation between these regions. Iron has been postulated to promote disease activity in MS by various means: (i) iron can amplify the activated state of microglia resulting in the increased production of proinflammatory mediators; (ii) excess intracellular iron deposits could promote mitochondria dysfunction; and (iii) improperly managed iron could catalyze the production of damaging reactive oxygen species (ROS). The pathological consequences of abnormal iron deposits may be dependent on the affected brain region and/or accumulation process. Here, we review putative mechanisms of enhanced iron uptake in MS and address the likely roles of iron in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|
32
|
Kalinowska-Łyszczarz A, Pawlak MA, Michalak S, Paprzycki W, Losy J. Immune cell NT-3 expression is associated with brain atrophy in multiple sclerosis patients. J Neuroimmunol 2011; 240-241:109-13. [PMID: 22036954 DOI: 10.1016/j.jneuroim.2011.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/17/2011] [Accepted: 10/07/2011] [Indexed: 01/03/2023]
Abstract
While neurotrophins mediate cell survival and proliferation in the nervous system, they are also expressed within peripheral blood mononuclear cells (PBMCs) of the immunological system. In multiple sclerosis (MS) neurotrophins released from PBMCs might play a neuroprotective role, delaying neurodegeneration within central nervous system. We aimed for identifying the link between neurotrophins' PBMCs expression and brain atrophy markers in relapsing-remitting MS (RRMS) patients. We have found that neurotrophin-3 PBMCs concentration is strongly correlated with brain-parenchymal fraction and corpus callosum cross-sectional area, which are well-established brain atrophy measures. Thus, PBMC-derived neurotrophin-3 might exert a direct or indirect neuroprotective effect in MS.
Collapse
Affiliation(s)
- Alicja Kalinowska-Łyszczarz
- Department of Clinical Neuroimmunology, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland.
| | | | | | | | | |
Collapse
|
33
|
Multiple sclerosis as a neurodegenerative disease: pathology, mechanisms and therapeutic implications. Curr Opin Neurol 2011; 24:224-9. [PMID: 21455066 DOI: 10.1097/wco.0b013e328346056f] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) treatments targeting the inflammatory nature of the disease have become increasingly effective in recent years. However, our efforts at targeting the progressive disease phase have so far been largely unsuccessful. This has led to the hypothesis that disease mechanisms independent of an adaptive immune response contribute to disease progression and closely resemble neurodegeneration. RECENT FINDINGS Nonfocal, diffuse changes in the MS brain, especially axonal loss and mitochondrial dysfunction, prove better correlates of disability than total lesion load and have been associated with disease progression. Molecular changes in nondemyelinated MS tissue also suggest that alterations in the MS brain are widespread and consist of pro-inflammatory as well as anti-inflammatory responses. However, local lymphocytic inflammation and microglial activation are salient features of the chronic disease, and T-cell-mediated inflammation contributes to tissue damage. In addition, neuroaxonal cytoskeletal alterations have been associated with disease progression. SUMMARY Our knowledge of the molecular mechanisms leading to neuroaxonal damage and demise in MS is steadily increasing. Experimental therapies targeting neuroaxonal ionic imbalances and energy metabolism in part show promising results. A better understanding of the molecular mechanisms underlying chronic progression will substantially aid the development of new treatment strategies.
Collapse
|