1
|
Anti-TNFR1 targeting in humanized mice ameliorates disease in a model of multiple sclerosis. Sci Rep 2018; 8:13628. [PMID: 30206422 PMCID: PMC6133964 DOI: 10.1038/s41598-018-31957-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/30/2018] [Indexed: 01/11/2023] Open
Abstract
Tumour necrosis factor (TNF) signalling is mediated via two receptors, TNF-receptor 1 (TNFR1) and TNF-receptor 2 (TNFR2), which work antithetically to balance CNS immune responses involved in autoimmune diseases such as multiple sclerosis. To determine the therapeutic potential of selectively inhibiting TNFR1 in mice with experimental autoimmune encephalomyelitis, we used chimeric human/mouse TNFR1 knock-in mice allowing the evaluation of antagonistic anti-human TNFR1 antibody efficacy. Treatment of mice after onset of disease with ATROSAB resulted in a robust amelioration of disease severity, correlating with reduced central nervous system immune cell infiltration. Long-term efficacy of treatment was achieved by treatment with the parental mouse anti-human TNFR1 antibody, H398, and extended by subsequent re-treatment of mice following relapse. Our data support the hypothesis that anti-TNFR1 therapy restricts immune cell infiltration across the blood-brain barrier through the down-regulation of TNF-induced adhesion molecules, rather than altering immune cell composition or activity. Collectively, we demonstrate the potential for anti-human TNFR1 therapies to effectively modulate immune responses in autoimmune disease.
Collapse
|
2
|
Effects of Fullerenols on Mouse Brain Microvascular Endothelial Cells. Int J Mol Sci 2017; 18:ijms18081783. [PMID: 28817067 PMCID: PMC5578172 DOI: 10.3390/ijms18081783] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/26/2022] Open
Abstract
Fullerenols, water-soluble C60-fullerene derivatives, have been shown to exert neuroprotective effects in vitro and in vivo, most likely due to their capability to scavenge free radicals. However, little is known about the effects of fullerenols on the blood–brain barrier (BBB), especially on cerebral endothelial cells under inflammatory conditions. Here, we investigated whether the treatment of primary mouse brain microvascular endothelial cells with fullerenols impacts basal and inflammatory blood–brain barrier (BBB) properties in vitro. While fullerenols (1, 10, and 100 µg/mL) did not change transendothelial electrical resistance under basal and inflammatory conditions, 100 µg/mL of fullerenol significantly reduced erk1/2 activation and resulted in an activation of NFκB in an inflammatory milieu. Our findings suggest that fullerenols might counteract oxidative stress via the erk1/2 and NFκB pathways, and thus are able to protect microvascular endothelial cells under inflammatory conditions.
Collapse
|
3
|
Lim JL, van der Pol SMA, Di Dio F, van Het Hof B, Kooij G, de Vries HE, van Horssen J. Protective effects of monomethyl fumarate at the inflamed blood-brain barrier. Microvasc Res 2015; 105:61-9. [PMID: 26679389 DOI: 10.1016/j.mvr.2015.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Reactive oxygen species play a key role in the pathogenesis of multiple sclerosis as they induce blood-brain barrier disruption and enhance transendothelial leukocyte migration. Thus, therapeutic compounds with antioxidant and anti-inflammatory potential could have clinical value in multiple sclerosis. The aim of the current study was to elucidate the therapeutic effects of monomethyl fumarate on inflammatory-mediated changes in blood-brain barrier function and gain insight into the underlying mechanism. METHODS The effects of monomethyl fumarate on monocyte transendothelial migration across and adhesion to inflamed human brain endothelial cells (hCMEC/D3) were quantified using standardized in vitro migration and adhesion assays. Flow cytometry analysis and qPCR were used to measure the concomitant effects of monomethyl fumarate treatment on protein expression of cell adhesion molecules. Furthermore, the effects of monomethyl fumarate on the expression and nuclear localization of proteins involved in the activation of antioxidant and inflammatory pathways in human brain endothelial cells were elucidated using nuclear fractionation and Western blotting. Statistical analysis was performed using one-way ANOVA followed by the Bonferroni post-hoc test. RESULTS Our results show that monomethyl fumarate induced nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 and concomitant production of the antioxidant enzymes heme oxygenase-1 and NADPH:quinone oxidoreductase-1 in brain endothelial cells. Importantly, monomethyl fumarate treatment markedly decreased monocyte transendothelial migration across and adhesion to inflamed human brain endothelial cells. Treatment of brain endothelial cells with monomethyl fumarate resulted in a striking reduction of vascular cell adhesion molecule expression. Surprisingly, monomethyl fumarate did not affect nuclear translocation of nuclear factor-кB suggesting that monomethyl fumarate potentially affects activity of nuclear factor-ĸB downstream of nuclear translocation. CONCLUSIONS Taken together, we show that monomethyl fumarate, the primary metabolite of dimethyl fumarate, which is currently used in the clinics for the treatment of relapsing-remitting multiple sclerosis, demonstrates beneficial therapeutic effects at the inflamed blood-brain barrier.
Collapse
Affiliation(s)
- Jamie L Lim
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Susanne M A van der Pol
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Flaminia Di Dio
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Bert van Het Hof
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
4
|
Opp MR, George A, Ringgold KM, Hansen KM, Bullock KM, Banks WA. Sleep fragmentation and sepsis differentially impact blood-brain barrier integrity and transport of tumor necrosis factor-α in aging. Brain Behav Immun 2015; 50. [PMID: 26218294 PMCID: PMC4831867 DOI: 10.1016/j.bbi.2015.07.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The factors by which aging predisposes to critical illness are varied, complex, and not well understood. Sepsis is considered a quintessential disease of old age because the incidence and mortality of severe sepsis increases in old and the oldest old individuals. Aging is associated with dramatic changes in sleep quality and quantity and sleep increasingly becomes fragmented with age. In healthy adults, sleep disruption induces inflammation. Multiple aspects of aging and of sleep dysregulation interact via neuroimmune mechanisms. Tumor necrosis factor-α (TNF), a cytokine involved in sleep regulation and neuroimmune processes, exerts some of its effects on the CNS by crossing the blood-brain barrier (BBB). In this study we examined the impact of sepsis, sleep fragmentation, and aging on BBB disruption and TNF transport into brain. We used the cecal ligation and puncture (CLP) model of sepsis in young and aged mice that were either undisturbed or had their sleep disrupted. There was a dichotomous effect of sepsis and sleep disruption with age: sepsis disrupted the BBB and increased TNF transport in young mice but not in aged mice, whereas sleep fragmentation disrupted the BBB and increased TNF transport in aged mice, but not in young mice. Combining sleep fragmentation and CLP did not produce a greater effect on either of these BBB parameters than did either of these manipulations alone. These results suggest that the mechanisms by which sleep fragmentation and sepsis alter BBB functions are fundamentally different from one another and that a major change in the organism's responses to those insults occurs with aging.
Collapse
Affiliation(s)
- Mark R. Opp
- Department of Anesthesiology & Pain Medicine, University of Washington School of Medicine, Seattle, WA 98104, United States
| | - Amrita George
- Department of Anesthesiology & Pain Medicine, University of Washington School of Medicine, Seattle, WA 98104, United States
| | - Kristyn M. Ringgold
- Department of Anesthesiology & Pain Medicine, University of Washington School of Medicine, Seattle, WA 98104, United States
| | - Kim M. Hansen
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, United States,Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, United States
| | - Kristin M. Bullock
- Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, United States
| | - William A. Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, United States,Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, United States,Corresponding author at: WAB, Rm 810A, Bldg 1, VAPSHCS, 1660 S. Columbian Way, Seattle, WA 98108, United States. (W.A. Banks)
| |
Collapse
|
5
|
The role of lymphotoxin signaling in the development of autoimmune pancreatitis and associated secondary extra-pancreatic pathologies. Cytokine Growth Factor Rev 2014; 25:125-37. [DOI: 10.1016/j.cytogfr.2014.01.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 12/23/2013] [Accepted: 01/02/2014] [Indexed: 12/24/2022]
|
6
|
Olsson T. Role of cytokines in multiple sclerosis and experimental autoimmune encephalomyelitis. Eur J Neurol 2013; 1:7-19. [PMID: 24283424 DOI: 10.1111/j.1468-1331.1994.tb00045.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- T Olsson
- Division of Neurology, Department of Clinical Neuroscience and Family Medicine, Karolinska Institute, Huddinge Hospital, S-141 86 Huddinge, Sweden
| |
Collapse
|
7
|
Evans MC, Couch Y, Sibson N, Turner MR. Inflammation and neurovascular changes in amyotrophic lateral sclerosis. Mol Cell Neurosci 2013; 53:34-41. [PMID: 23110760 DOI: 10.1016/j.mcn.2012.10.008] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 10/17/2012] [Accepted: 10/19/2012] [Indexed: 11/16/2022] Open
Abstract
Neuroinflammation in now established as an important factor in the pathogenesis of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). At various time points, astrocytes and microglia are markedly activated, either producing neuroprotective or pro-inflammatory molecules, which can decrease or increase the rate of primary motor neuron degeneration respectively. Recent research has shown that this neuroinflammatory component is affected by the peripheral immune system; T lymphocytes in particular are able to cross into the brain and spinal cord parenchyma, where they interact with resident microglia, either inducing them to adopt an M1 (cytotoxic) or M2 (protective) phenotype, depending on the stage of disease. Clearly understanding the changes that occur to allow the interaction between peripheral and central immune responses will be essential in any attempt to manipulate the disease process via neuroinflammatory mechanisms. However, our understanding of the endothelial changes, which facilitate the infiltration of peripheral immune cells into the brain and spinal cord, is still in its infancy. There are suggestions, though, of up-regulation of cellular adhesion molecules, which are able to arrest circulating leukocytes and facilitate diapedesis into the brain parenchyma. In addition, tight junction proteins appear to be down-regulated, leading to an increase in vascular permeability, an effect that is amplified by vascular damage late in the disease process. This review summarises our current knowledge regarding neuroinflammation, peripheral immune involvement, and endothelial changes in ALS. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
Affiliation(s)
- M C Evans
- Nuffield Department of Clinical Neurosciences, Oxford University, UK
| | | | | | | |
Collapse
|
8
|
Abstract
Abstract
Collapse
|
9
|
Zhu J, Mix E, Issazadeh S, Link H. Dynamics of mRNA expression of interferon-γ, interleukin 4 and transforming growth factor β1 in sciatic nerves and lymphoid organs in experimental allergic neuritis. Eur J Neurol 2011; 3:232-40. [DOI: 10.1111/j.1468-1331.1996.tb00428.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Domingues HS, Mues M, Lassmann H, Wekerle H, Krishnamoorthy G. Functional and pathogenic differences of Th1 and Th17 cells in experimental autoimmune encephalomyelitis. PLoS One 2010; 5:e15531. [PMID: 21209700 PMCID: PMC3000428 DOI: 10.1371/journal.pone.0015531] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 10/08/2010] [Indexed: 01/19/2023] Open
Abstract
Background There is consensus that experimental autoimmune encephalomyelitis (EAE) can be mediated by myelin specific T cells of Th1 as well as of Th17 phenotype, but the contribution of either subset to the pathogenic process has remained controversial. In this report, we compare functional differences and pathogenic potential of “monoclonal” T cell lines that recognize myelin oligodendrocyte glycoprotein (MOG) with the same transgenic TCR but are distinguished by an IFN-γ producing Th1-like and IL-17 producing Th17-like cytokine signature. Methods and Findings CD4+ T cell lines were derived from the transgenic mouse strain 2D2, which expresses a TCR recognizing MOG peptide 35–55 in the context of I-Ab. Adoptive transfer of Th1 cells into lymphopenic (Rag2−/−) recipients, predominantly induced “classic” paralytic EAE, whereas Th17 cells mediated “atypical” ataxic EAE in approximately 50% of the recipient animals. Combination of Th1 and Th17 cells potentiated the encephalitogenicity inducing classical EAE exclusively. Th1 and Th17 mediated EAE lesions differed in their composition but not in their localization within the CNS. While Th1 lesions contained IFN-γ, but no IL-17 producing T cells, the T cells in Th17 lesions showed plasticity, substantially converting to IFN-γ producing Th1-like cells. Th1 and Th17 cells differed drastically by their lytic potential. Th1 but not Th17 cells lysed autoantigen presenting astrocytes and fibroblasts in vitro in a contact-dependent manner. In contrast, Th17 cells acquired cytotoxic potential only after antigenic stimulation and conversion to IFN-γ producing Th1 phenotype. Conclusions Our data demonstrate that both Th1 and Th17 lineages possess the ability to induce CNS autoimmunity but can function with complementary as well as differential pathogenic mechanisms. We propose that Th17-like cells producing IL-17 are required for the generation of atypical EAE whereas IFN-γ producing Th1 cells induce classical EAE.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Astrocytes/immunology
- Astrocytes/metabolism
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- Cell Differentiation/immunology
- Cell Proliferation
- Cells, Cultured
- Coculture Techniques
- Cytotoxicity, Immunologic/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin Proteins
- Myelin-Associated Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein
- Spleen/immunology
- Spleen/metabolism
- Spleen/pathology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th1 Cells/transplantation
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/transplantation
Collapse
Affiliation(s)
- Helena S. Domingues
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
- PhD Program in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Marsilius Mues
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Hartmut Wekerle
- Department of Neuroimmunology, Max Planck Institute of Neurobiology, Martinsried, Germany
| | | |
Collapse
|
11
|
von Wedel-Parlow M, Schrot S, Lemmen J, Treeratanapiboon L, Wegener J, Galla HJ. Neutrophils cross the BBB primarily on transcellular pathways: an in vitro study. Brain Res 2010; 1367:62-76. [PMID: 20875807 DOI: 10.1016/j.brainres.2010.09.076] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Accepted: 09/21/2010] [Indexed: 01/13/2023]
Abstract
The cerebral microcapillary endothelium forms a highly important barrier between the blood and the interstitial fluid of the brain (blood-brain barrier) that controls the passage of molecules and cells in and out of the CNS. Several CNS diseases include leukocyte extravasation through the endothelium via two mechanistically distinct routes, the paracellular and the transcellular pathway. We established a new in vitro model of the inflamed blood-brain barrier consisting of primary cultured porcine brain capillary endothelial cells which express a tight endothelial barrier even under inflammatory conditions. By means of this specialized blood-brain barrier model we extensively studied the transmigration of neutrophils. Electron and scanning force microscopy as well as immunofluorescence imaging captured the penetrating neutrophil on the endothelial cellular body in between the junctions clearly suggesting a transcellular migration pathway. Electric cell-substrate impedance sensing and transendothelial electrical resistance measurements in combination with expression analysis of tight junction proteins demonstrate that the neutrophil-endothelial interaction does not disrupt the barrier. In conclusion, this study, based on an in vitro model of the blood-brain barrier under inflammatory conditions, evidently implicates that neutrophils preferentially migrate across the BBB via the transcellular route without impairing endothelial barrier function whereas paracellular transmigration plays only a minor role if the barrier is strongly expressed.
Collapse
Affiliation(s)
- Magdalena von Wedel-Parlow
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 2, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
12
|
Sayed BA, Christy AL, Walker ME, Brown MA. Meningeal mast cells affect early T cell central nervous system infiltration and blood-brain barrier integrity through TNF: a role for neutrophil recruitment? THE JOURNAL OF IMMUNOLOGY 2010; 184:6891-900. [PMID: 20488789 DOI: 10.4049/jimmunol.1000126] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mast cells contribute to the pathogenesis of experimental autoimmune encephalomyelitis, a rodent model of the human demyelinating disease multiple sclerosis. Yet their site and mode of action is unknown. In both diseases, myelin-specific T cells are initially activated in peripheral lymphoid organs. However, for disease to occur, these cells must enter the immunologically privileged CNS through a breach in the relatively impermeable blood-brain barrier. In this study, we demonstrate that a dense population of resident mast cells in the meninges, structures surrounding the brain and spinal cord, regulate basal CNS barrier function, facilitating initial T cell CNS entry. Through the expression of TNF, mast cells recruit an early wave of neutrophils to the CNS. We propose that neutrophils in turn promote the blood-brain barrier breach and together with T cells lead to further inflammatory cell influx and myelin damage. These findings provide specific targets for intervention in multiple sclerosis as well as other immune-mediated CNS diseases.
Collapse
Affiliation(s)
- Blayne A Sayed
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
13
|
Autoimmune Processes in the Central Nervous System. HANDBOOK OF NEUROCHEMISTRY AND MOLECULAR NEUROBIOLOGY 2008. [PMCID: PMC7121640 DOI: 10.1007/978-0-387-30398-7_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In this chapter we discuss the factors that contribute to the unique immunological environment of the central nervous system and the mechanisms that may account for the development of autoimmunity within the CNS, including infectious agents as inducers of autoimmune disease. Consideration is given to a variety of human neurological diseases of autoimmune or presumed autoimmune etiology: autism, neuromyelitis optica, neuromyotonia, schizophrenia, lethargic encephalitis and stiff‐man syndrome. Also, we discuss autoimmunity as a possible mediator of CNS repair and examples of the protective effects of bacterial and helminth infections on CNS disease. Multiple sclerosis and models of multiple sclerosis are discussed with special attention given to the Theiler's virus‐induced demyelination model.
Collapse
|
14
|
A promising therapeutic approach for multiple sclerosis: recombinant T-cell receptor ligands modulate experimental autoimmune encephalomyelitis by reducing interleukin-17 production and inhibiting migration of encephalitogenic cells into the CNS. J Neurosci 2007; 27:12531-9. [PMID: 18003831 DOI: 10.1523/jneurosci.3599-07.2007] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recombinant T-cell receptor ligands (RTLs) can prevent and reverse clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) in an antigen-specific manner and are currently in clinical trials for treatment of subjects with multiple sclerosis (MS). To evaluate regulatory mechanisms, we designed and tested RTL551, containing the alpha1 and beta1 domains of the I-A(b) class II molecule covalently linked to the encephalitogenic MOG-35-55 peptide in C57BL/6 mice. Treatment of active or passive EAE with RTL551 after disease onset significantly reduced clinical signs and spinal cord lesions. Moreover, RTL551 treatment strongly and selectively reduced secretion of interleukin-17 and tumor necrosis factor alpha by transferred green fluorescent protein-positive (GFP+) MOG-35-55-reactive T-cells and almost completely abrogated existent GFP+ cellular infiltrates in affected spinal cord sections. Reduced inflammation in spinal cords of RTL551-treated mice was accompanied by a highly significant downregulation of chemokines and their receptors and inhibition of VCAM-1 (vascular cell adhesion molecule-1) and ICAM-1 (intercellular adhesion molecule-1) expression by endothelial cells. Thus, RTL therapy cannot only inhibit systemic production of encephalitogenic cytokines by the targeted myelin oligodendrocyte glycoprotein-reactive T-cells but also impedes downstream local recruitment and retention of inflammatory cells in the CNS. These findings indicate that targeted immunotherapy of antigen-specific T-cells can result in a reversal of CNS lesion formation and lend strong support to the application of the RTL approach for therapy in MS.
Collapse
|
15
|
Wheeler RD, Zehntner SP, Kelly LM, Bourbonnière L, Owens T. Elevated interferon gamma expression in the central nervous system of tumour necrosis factor receptor 1-deficient mice with experimental autoimmune encephalomyelitis. Immunology 2006; 118:527-38. [PMID: 16780563 PMCID: PMC1782311 DOI: 10.1111/j.1365-2567.2006.02395.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Inflammation in the central nervous system (CNS) can be studied in experimental autoimmune encephalomyelitis (EAE). The proinflammatory cytokines interferon-gamma (IFN-gamma) and tumour necrosis factor (TNF) are implicated in EAE pathogenesis. Signals through the type 1 TNF receptor (TNFR1) are required for severe EAE to develop, whereas deficiency in IFN-gamma or its receptor result in more severe EAE. We investigated IFN-gamma expression in TNFR1-deficient (TNFR1-/-) mice. We describe here that there were more IFN-gamma-secreting T cells present in the CNS of TNFR1-/- mice during EAE compared to wild-type (WT) mice, despite that clinical symptoms were mild, with delayed onset. There was greater expression of IL-12/23p40 by antigen-presenting cells in these mice, and in vitro, TNFR1-/- antigen-presenting cells induced greater secretion of IFN-gamma but not interleukin (IL)-17 when cultured with primed T cells than did WT antigen presenting cells. TNFR1-/- mice with EAE had significantly higher expression of CXCL10 mRNA (but not CCL5 mRNA) in the CNS compared to WT mice with EAE. These data demonstrate that IFN-gamma expression is enhanced in the CNS of TNFR1-/- mice with EAE and suggest that IFN-gamma levels do not necessarily correlate with EAE severity.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/microbiology
- Gene Expression Regulation
- Interferon-gamma/analysis
- Interferon-gamma/genetics
- Macrophages/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Tumor Necrosis Factor, Type I/analysis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Spinal Cord/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Rachel D Wheeler
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Simone P Zehntner
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Lisa M Kelly
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Lyne Bourbonnière
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
| | - Trevor Owens
- Neuroimmunology Unit, Montreal Neurological InstituteMontreal, Quebec, Canada
- Medical Biotechnology Centre, University of Southern DenmarkOdense C, Denmark
| |
Collapse
|
16
|
Gimenez MA, Sim J, Archambault AS, Klein RS, Russell JH. A tumor necrosis factor receptor 1-dependent conversation between central nervous system-specific T cells and the central nervous system is required for inflammatory infiltration of the spinal cord. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1200-9. [PMID: 16565495 PMCID: PMC1606568 DOI: 10.2353/ajpath.2006.050332] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the role of tumor necrosis factor receptor 1 (TNFR1) in inflammation initiated by the adoptive transfer of central nervous system (CNS)-specific Th1 cells in experimental autoimmune encephalomyelitis, a murine model of multiple sclerosis. This adoptive transfer paradigm eliminates the confounding effects of bacterial adjuvants in the analysis of inflammation. We found that although T cells could reach the meninges and perivascular space in the absence of TNFR1, recruitment of other inflammatory cells from the blood was dramatically reduced. The reduction in the recruitment of CD11b(hi) cells correlated with a dramatic reduction in the production of the chemokines CCL2 (MCP-1) and CXLC2 (MIP-2) in TNFR1-deficient hosts. Bone marrow chimera experiments demonstrated that TNF can be effectively supplied by either the hematopoietic system or the CNS, but the essential TNFR1-responsive cells reside in the CNS. Previous work has demonstrated that microglia produce CCL2, and here we demonstrate that astrocytes and endothelial cells produced CXCL2 in the early stages of inflammation. Therefore, productive inflammation results from a conversation, or mutually responding signals, between the initiating T cells and cells in the parenchyma of the spinal cord.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Astrocytes/metabolism
- Bone Marrow Cells/metabolism
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- CD11b Antigen/metabolism
- Chemokine CCL2/biosynthesis
- Chemokine CXCL2
- Chemokines/biosynthesis
- Chemokines, CXC/biosynthesis
- Chimera
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Endothelial Cells/metabolism
- Mice
- Mice, Congenic
- Mice, Inbred C57BL
- Myelitis/immunology
- Myelitis/metabolism
- Myelitis/pathology
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes/immunology
- Th1 Cells/immunology
- Tumor Necrosis Factor Decoy Receptors
Collapse
Affiliation(s)
- Mary Ann Gimenez
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
17
|
Coisne C, Faveeuw C, Delplace Y, Dehouck L, Miller F, Cecchelli R, Dehouck B. Differential expression of selectins by mouse brain capillary endothelial cells in vitro in response to distinct inflammatory stimuli. Neurosci Lett 2005; 392:216-20. [PMID: 16214291 DOI: 10.1016/j.neulet.2005.09.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2005] [Revised: 08/19/2005] [Accepted: 09/09/2005] [Indexed: 12/01/2022]
Abstract
Increased lymphocyte trafficking across blood-brain barrier (BBB) is a prominent and early event in inflammatory and immune-mediated CNS diseases. The adhesion molecules that control the entry of leukocytes into the brain have not been fully elucidated. Although the role of ICAM-1 and VCAM-1 has been well documented, the expression and role of selectins is still a matter of controversy. In a mouse syngenic in vitro BBB model, highly relevant for examining immunological events, mouse brain capillary endothelial cells (MBCECs) do not express selectins. Treatment of MBCECs with LPS, induced E- and P-selectin expression, whereas TNF-alpha or IFN-gamma treatments did not. Finally, P-selectin but not E-selectin expression was induced in IL-1beta treated MBCECs. Thus, our study suggests that diverse inflammatory stimuli could differentially regulate selectin expression at the BBB.
Collapse
Affiliation(s)
- Caroline Coisne
- EA 2465-Université d'Artois, Faculté des Sciences Jean Perrin, 62307 Lens, France
| | | | | | | | | | | | | |
Collapse
|
18
|
Russell JH. Interaction between the immune and central nervous systems. Immunol Res 2005; 32:225-9. [PMID: 16106074 DOI: 10.1385/ir:32:1-3:225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Much of the understanding of tolerance has focused on the requirements for antigen-specific lymphocyte activation and function. However, there is increasing evidence for anatomic regulation of effector access to self antigens. Recently, a number of studies have provided evidence for tissue-specific "addressins" in chemokine/chemokine receptor pairs. The central nervous system (CNS) provides special anatomic barriers to the movement of cells from the vascular compartment to the parenchyma. Herein I raise the possibility that antigen, perhaps through specialized antigen-presenting cells, may play a role in regulating access of activated lymphocytes into the CNS parenchyma. The results suggest that a reexamination of the widely held dogma that all activated lymphocytes have access to the CNS parenchyma is necessary to understand the relationship between the immune and central nervous systems.
Collapse
Affiliation(s)
- John H Russell
- Program in Immunology, Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Kipar A, May H, Menger S, Weber M, Leukert W, Reinacher M. Morphologic features and development of granulomatous vasculitis in feline infectious peritonitis. Vet Pathol 2005; 42:321-30. [PMID: 15872378 DOI: 10.1354/vp.42-3-321] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Feline infectious peritonitis (FIP) is a fatal, coronavirus (CoV)-induced systemic disease in cats, characterized by granulomas in organs and granulomatous vasculitis. This study describes the morphologic features of granulomatous vasculitis in FIP as well as its development in the course of monocyte-associated feline CoV (FCoV) viremia in five naturally infected Domestic Shorthair cats with FIP. Monocyte-associated FCoV viremia was demonstrated by immunohistology, RNA in situ hybridization, and electron micropscopy. Granulomatous phlebitis at different stages of development was observed. Vasculitic processes ranged from attachment and emigration of FCoV-infected monocytes to vascular/perivascular granulomatous infiltrates with destruction of the vascular basal lamina. Monocytes as well as perivascular macrophages were activated because they were strongly positive for CD18 and expressed cytokines (tumor necrosis factor-alpha and interleukin-1beta) and matrix metalloproteinase-9. In addition, general activation of endothelial cells, represented by major histocompatibility complex II upregulation, was observed in all cases. These results confirm FIP as a monocyte-triggered systemic disease and demonstrate the central role of activated monocytes in FIP vasculitis.
Collapse
Affiliation(s)
- A Kipar
- Department of Veterinary Pathology, Faculty of Veterinary Science, University of Liverpool, Crown Street, Liverpool L69 7ZJ, UK.
| | | | | | | | | | | |
Collapse
|
20
|
Archambault AS, Sim J, Gimenez MAT, Russell JH. Defining antigen-dependent stages of T cell migration from the blood to the central nervous system parenchyma. Eur J Immunol 2005; 35:1076-85. [PMID: 15761850 DOI: 10.1002/eji.200425864] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In experimental autoimmune encephalomyelitis (EAE), intravenous transfer of activated CD4(+) myelin-specific T cells is sufficient to induce disease. Transferred T cells access the CNS parenchyma by trafficking across the blood brain barrier (BBB) vascular endothelium into the perivascular space, and then across the glial limitans that is made up of astrocytes and microglia. Flow cytometry analysis of cells isolated from CNS tissue does not distinguish between T cell populations at the various stages of migration. In this study, we have used GK1.5 (anti-CD4) treatment along with immunohistochemistry to distinguish between populations of T cells that are associated with the vasculature, T cells that have migrated into the perivascular space, and T cells in the parenchyma. We have also re-evaluated antigen specificity requirements of T cells as they are recruited to the CNS parenchyma. Activated myelin-specific T cells are restricted to the CNS vasculature for at least 24 h post transfer. MHC class II expression on the recipient is required for cells to traffic across the CNS vascular endothelium. Further, Con A-stimulated or non-CNS-specific (ovalbumin-specific) T cells fail to migrate into the perivascular space, and only enter the CNS parenchyma when co-transferred with myelin-specific T cells. Our results indicate that Th1 populations cannot accumulate in the perivascular (subarachnoid, Virchow-Robbins) space without a CNS antigen-specific signal.
Collapse
Affiliation(s)
- Angela S Archambault
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
21
|
Walker JE, Giri SN, Margolin SB. A double-blind, randomized, controlled study of oral pirfenidone for treatment of secondary progressive multiple sclerosis. Mult Scler 2005; 11:149-58. [PMID: 15794387 DOI: 10.1191/1352458505ms1134oa] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Currently, there are no approved treatments for secondary progressive multiple sclerosis (MS) that stabilize or reverse the neurological disabilities associated with this disease. Oral pirfenidone was found to stabilize and overcome the disabilities in two published independent open-label studies in secondary progressive MS. This led us to study pirfenidone in a phase II double-blind, randomized and controlled, clinical trial in patients with advanced secondary progressive MS for 12 months. Forty-three patients met the eligibility criteria approved by the IRB and accepted by the FDA. Of these patients, 18 were randomly assigned to placebo and 25 patients to oral pirfenidone groups. All eligible patients were included in the statistical analysis of the data according to intention-to-treat principles. Some patients on oral pirfenidone manifested mild drug-related adverse effects, but it was well tolerated overall. By one month, pirfenidone significantly (P < 0.05) improved the Scripps Neurological Rating Scale (SNRS) scores, and scores remained significantly improved for 3, 6 and 12 months when compared to the baseline SNRS scores. In contrast, the SNRS scores of patients on oral placebo were not significantly improved at 1, 3, 6 or 12 months of the study, when compared with baseline scores. Oral pirfenidone significantly (P <0.04) reduced the incidence of relapses (27.8% on placebo versus 8.0% on pirfenidone). Furthermore, oral pirfenidone treatment was associated with a marked improvement in bladder dysfunction (40.0% on pirfenidone versus 16.7% on placebo). Expanded Disability Status Scale scores and MRI lesion count were not significantly different in the placebo and pirfenidone groups. These findings indicate a significant effect of pirfenidone on clinical disability and bladder function for secondary progressive MS patients. A major multicentre, double-blind, randomized, controlled trial is justified.
Collapse
|
22
|
Gimenez MAT, Sim JE, Russell JH. TNFR1-dependent VCAM-1 expression by astrocytes exposes the CNS to destructive inflammation. J Neuroimmunol 2004; 151:116-25. [PMID: 15145610 DOI: 10.1016/j.jneuroim.2004.02.012] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Revised: 02/25/2004] [Accepted: 02/25/2004] [Indexed: 11/26/2022]
Abstract
VCAM-1 is an adhesion molecule that is important to leukocyte movement across the blood-brain barrier and is involved in the formation of destructive CNS inflammatory lesions in experimental autoimmune encephalomyelitis (EAE) and multiple sclerosis (MS). We examined VCAM-1 expression in the CNS of animals with passively induced EAE and found abundant expression not only on the CNS endothelium but also on astrocytes. We show that tumor necrosis factor receptor-1 (TNFR1) signaling is required for VCAM-1 expression by astrocytes, not the vascular endothelium. In addition, we demonstrate that VCAM-1 expression by astrocytes is crucial for T cell entry into the CNS parenchyma and is required for manifestation of neurological disease.
Collapse
Affiliation(s)
- Mary Ann T Gimenez
- Department of Molecular Biology and Pharmacology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
23
|
Bao F, Chen Y, Dekaban GA, Weaver LC. Early anti-inflammatory treatment reduces lipid peroxidation and protein nitration after spinal cord injury in rats. J Neurochem 2004; 88:1335-44. [PMID: 15009633 DOI: 10.1046/j.1471-4159.2003.02240.x] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We investigated mechanisms by which a monoclonal antibody (mAb) against the CD11d subunit of the leukocyte integrin CD11d/CD18 improves neurological recovery after spinal cord injury (SCI) in the rat. The effects of an anti-CD11d mAb treatment were assessed on ED-1 expression (estimating macrophage infiltration), myeloperoxidase activity (MPO, approximating neutrophil infiltration), lipid peroxidation, inducible nitric oxide synthase (iNOS) and nitrotyrosine (indicating protein nitration) expression in the spinal cord lesion after severe clip-compression injury. Protein expression was evaluated by western blotting and immunocytochemistry. Lipid peroxidation was assessed by thiobarbituric acid reactive substances (TBARS) production. After anti-CD11d mAb treatment, decreased ED-1 expression at 6-72 h after SCI indicated reduced macrophage infiltration. MPO activity (units/g tissue) was reduced significantly from 114 +/- 11 to 75 +/- 8 (- 34%) at 6 h and from 38 +/- 2 to 22 +/- 4 (- 42%) at 72 h. After SCI, anti-CD11d mAb treatment significantly reduced TBARS from 501 +/- 61 to 296 +/- 17 nm (- 41%) at 6 h and to approximately uninjured values (87 nm) at 72 h. The mAb treatment also attenuated the expression of iNOS and formation of nitrotyrosine at 6-72 h after SCI. These data indicate that anti-CD11d mAb treatment blocks intraspinal neutrophil and macrophage infiltration, reducing the intraspinal concentrations of reactive oxygen and nitrogen species. These effects likely underlie improved tissue preservation and neurological function resulting from the mAb treatment.
Collapse
Affiliation(s)
- Feng Bao
- Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute, London, Ontario, Canada
| | | | | | | |
Collapse
|
24
|
Krizanac-Bengez L, Kapural M, Parkinson F, Cucullo L, Hossain M, Mayberg MR, Janigro D. Effects of transient loss of shear stress on blood-brain barrier endothelium: role of nitric oxide and IL-6. Brain Res 2003; 977:239-46. [PMID: 12834884 DOI: 10.1016/s0006-8993(03)02689-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Loss of blood-brain barrier (BBB) function may contribute to post-ischemic cerebral injury by yet unknown mechanisms. Ischemia is associated with anoxia, aglycemia and loss of flow (i.e. shearing forces). We tested the hypothesis that loss of shear stress alone does not acutely affect BBB function due to a protective cascade of mechanisms involving cytokines and nitric oxide (NO). To determine the relative contribution of shear stress on BBB integrity we used a dynamic in vitro BBB model based on co-culture of rat brain microvascular endothelial cells (RBMEC) and astrocytes. Trans-endothelial electrical resistance (TEER), IL-6 release and NO levels were measured from the lumenal and ablumenal compartments throughout the experiment. Flow-exposed RBMEC were challenged with 1 h of normoxic-normoglycemic flow cessation (NNFC) followed by reperfusion for 2 to 24 h. NNFC caused a progressive drop in nitric oxide production during flow cessation followed by a time-dependent increase in ablumenal IL-6 associated with a prolonged NO increase during reperfusion. The nitric oxide synthetase (NOS) inhibitor L-NAME (10 microM) abrogated all effects of NNFC, including changes in NO and cytokine production. BBB permeability did not increase during or after NNFC/reperfusion, but was increased by treatment with L-NAME or when the effects of IL-6 were blocked. Flow adapted RBMEC and astrocytes respond to NNFC/reperfusion by overproduction of IL-6, possibly secondary to increased production of NO during the reperfusion. Maintenance of BBB function during and following NNFC appears to depend on intact NO signaling and IL-6 release.
Collapse
Affiliation(s)
- Ljiljana Krizanac-Bengez
- Cerebrovascular Research Center, Department of Neurological Surgery, Cleveland Clinic Foundation NB20, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Colmenares M, Kima PE, Samoff E, Soong L, McMahon-Pratt D. Perforin and gamma interferon are critical CD8+ T-cell-mediated responses in vaccine-induced immunity against Leishmania amazonensis infection. Infect Immun 2003; 71:3172-82. [PMID: 12761096 PMCID: PMC155724 DOI: 10.1128/iai.71.6.3172-3182.2003] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous studies have demonstrated that protection against New World leishmaniasis caused by Leishmania amazonensis can be elicited by immunization with the developmentally regulated Leishmania amastigote antigen, P-8. In this study, several independent experimental approaches were employed to investigate the protective immunological mechanisms involved. T-cell subset depletion experiments clearly indicate that elicitation of CD8(+) (as well as CD4(+)) effector responses is required for protection. Further, mice lacking beta(2)-microglobulin (and hence deficient in major histocompatibility complex class I antigen presentation) were not able to control a challenge infection after vaccination, indicating an essential protective role for CD8(+) T effector responses. Analysis of the events ongoing at the cutaneous site of infection indicated a changing cellular dynamic involved in protection. Early postinfection in protectively vaccinated mice, a predominance of CD8(+) T cells, secreting gamma interferon (IFN-gamma) and expressing perforin, was observed at the site of infection; subsequently, activated CD4(+) T cells producing IFN-gamma were primarily found. As protection correlated with the ratio of total IFN-gamma-producing cells (CD4(+) and CD8(+) T cells) to macrophages found at the site of infection, a role for IFN-gamma was evident; in addition, vaccination of IFN-gamma-deficient mice failed to provide protection. To further assess the effector mechanisms that mediate protection, mice deficient in perforin synthesis were examined. Perforin-deficient mice vaccinated with the P-8 antigen were unable to control infection. Thus, the elicitation of CD8(+) T cell effector mechanisms (perforin, IFN-gamma) are clearly required in the protective immune response against L. amazonensis infection in vaccinated mice.
Collapse
Affiliation(s)
- María Colmenares
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut 06520-8034, USA
| | | | | | | | | |
Collapse
|
26
|
Weaver LC, Marsh DR, Gris D, Meakin SO, Dekaban GA. Central mechanisms for autonomic dysreflexia after spinal cord injury. PROGRESS IN BRAIN RESEARCH 2002; 137:83-95. [PMID: 12440361 DOI: 10.1016/s0079-6123(02)37009-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Lynne C Weaver
- Spinal Cord Injury Laboratory, BioTherapeutics Research Group, John P. Robarts Research Institute, 100 Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada.
| | | | | | | | | |
Collapse
|
27
|
Floris S, Ruuls SR, Wierinckx A, van der Pol SMA, Döpp E, van der Meide PH, Dijkstra CD, De Vries HE. Interferon-beta directly influences monocyte infiltration into the central nervous system. J Neuroimmunol 2002; 127:69-79. [PMID: 12044977 DOI: 10.1016/s0165-5728(02)00098-x] [Citation(s) in RCA: 136] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Interferon-beta (IFN-beta) has beneficial effects on the clinical symptoms of multiple sclerosis (MS) patients, but its exact mechanism of action is yet unknown. We here suggest that IFN-beta directly modulates inflammatory events at the level of cerebral endothelium. IFN-beta treatment resulted in a marked reduction of perivascular infiltrates in acute experimental allergic encephalomyelitis (EAE), the rat model for MS, which was coupled to a major decrease in the expression of the adhesion molecules ICAM-1 and VCAM-1 on brain capillaries. In vitro, IFN-beta reduced the mRNA levels and protein expression of adhesion molecules of brain endothelial cell cultures and diminished monocyte transendothelial migration. Monocyte adhesion and subsequent migration was found to be predominantly regulated by VCAM-1. These data indicate that IFN-beta exerts direct antiinflammatory effects on brain endothelial cells thereby contributing to reduced lesion formation as observed in MS patients.
Collapse
Affiliation(s)
- Sarah Floris
- Department of Molecular Cell Biology, VU Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hellings N, Gelin G, Medaer R, Bruckers L, Palmers Y, Raus J, Stinissen P. Longitudinal study of antimyelin T-cell reactivity in relapsing-remitting multiple sclerosis: association with clinical and MRI activity. J Neuroimmunol 2002; 126:143-60. [PMID: 12020966 DOI: 10.1016/s0165-5728(02)00052-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In multiple sclerosis (MS), T-cells are considered to be critical in coordinating an immunopathological cascade that results in myelin damage. We investigated whether clinical disease activity or brain inflammatory activity as measured by magnetic resonance imaging (MRI) was associated with changes in autoreactive T-cell reactivities in MS patients. To this end, a longitudinal study was performed in which T-cell-related immune parameters and clinical parameters (including MRI) were monitored in seven relapsing-remitting (RR) MS patients and two healthy controls with bimonthly intervals over a period of 18 months. The serial evaluation of antimyelin (MBP, PLP, MOG) T-cell responses revealed highly dynamic shifts and fluctuations from one pattern to another in a patient-dependent manner. In some of the patients, changes in T-cell-related immune variables were found to concur with MRI activity and generally preceded clinical relapses. These alterations include: increased number of myelin-reactive IFN-gamma secreting T-cells, detection of clonally expanded myelin-reactive T-cells, elevated proinflammatory and decreased antiinflammatory cytokine production, upregulation of ICAM-1 membrane expression and highly increased serum levels of soluble VCAM-1. However, not all exacerbations and MRI changes were associated with changes in antimyelin reactivity. Some of the observed immune alterations were also detected in the healthy controls, indicating that additional regulatory mechanisms-which may be defective in MS-play a role in the downregulation of potentially pathological T-cell responses. In conclusion, this study provides further support for an important role of myelin-reactive T-cells in the pathogenesis of MS. In addition, the observed dynamic changes in the antimyelin T-cell reactivity pattern may be a major obstacle for the development of antigen-specific immunotherapies.
Collapse
Affiliation(s)
- Niels Hellings
- Biomedisch Onderzoeksintituut (BIOMED), Limburgs Universitair Centrum, Diepenbeek, Belgium
| | | | | | | | | | | | | |
Collapse
|
29
|
Middel P, Lippert U, Hummel KM, Bertsch HP, Artuc M, Schweyer S, Radzun HJ. Expression of lymphotoxin-alpha by keratinocytes: a further mediator for the lichenoid reaction. Pathobiology 2001; 68:291-300. [PMID: 11493763 DOI: 10.1159/000055940] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Lichen planus (LP) represents a disease in which autoimmune mechanisms mediated by Th1 T cells are involved. Lymphotoxin-alpha (LT-alpha) represents a Th1 cytokine with proinflammatory activities in LP, as has recently been demonstrated for interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). METHODS Expression of LT-alpha mRNA was investigated by RT-PCR and nonradioactive in situ hybridization. Double staining methods were applied to characterize the phenotype of cells expressing LT-alpha. Cell stimulation experiments were performed on the transformed squamous cell line HaCaT. RESULTS In contrast to normal skin, LT-alpha-specific RT-PCR products were found in all cases of LP. Cells in the inflammatory infiltrate expressing LT-alpha were identified as mainly T cells and mast cells, as shown by in situ hybridization. Furthermore, predominant LT-alpha mRNA expression could be observed in lesional keratinocytes adjacent to the band-like inflammatory infiltrate. In cell stimulation experiments, it could be shown that IFN-gamma induces LT-alpha and TNF-alpha mRNA in the human squamous cell line HaCaT, concomitant with upregulation of MHC class II and intercellular adhesion molecule-1, which could also be observed on lesional keratinocytes in LP. CONCLUSIONS In LP, LT-alpha mRNA is predominantly expressed by lesional keratinocytes and to a lesser extent by inflammatory cells. Induction of LT-alpha in keratinocytes is closely related to the expression of TNF-alpha and MHC class II. The loci of TNF-alpha and LT-alpha map to MHC class III on chromosome 6, which is closely linked to the MHC class II gene locus. Our results suggest that stimulation of keratinocytes with IFN-gamma results in the upregulation of proinflammatory cytokines such as LT-alpha and TNF-alpha as well as MHC class II, which map to the same gene region of immunoregulatory genes on chromosome 6 and may be involved in the induction and maintenance of the disease.
Collapse
Affiliation(s)
- P Middel
- Department of Pathology, Georg August University of Göttingen, Robert-Koch-Strasse 40, D-37073 Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
Cua DJ, Hutchins B, LaFace DM, Stohlman SA, Coffman RL. Central nervous system expression of IL-10 inhibits autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:602-8. [PMID: 11123343 DOI: 10.4049/jimmunol.166.1.602] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis, an inflammatory, demyelinating disease of the CNS currently lacks an effective therapy. We show here that CNS inflammation and clinical disease in experimental autoimmune encephalomyelitis, an experimental model of multiple sclerosis, could be prevented completely by a replication-defective adenovirus vector expressing the anti-inflammatory cytokine IL-10 (replication-deficient adenovirus expressing human IL-10), but only upon inoculation into the CNS where local infection and high IL-10 levels were achieved. High circulating levels of IL-10 produced by i. v. infection with replication-deficient adenovirus expressing human IL-10 was ineffective, although the immunological pathways for disease are initiated in the periphery in this disease model. In addition to this protective activity, intracranial injection of replication-deficient adenovirus expressing human IL-10 to mice with active disease blocked progression and accelerated disease remission. In a relapsing-remitting disease model, IL-10 gene transfer during remission prevented subsequent relapses. These data help explain the varying outcomes previously reported for systemic delivery of IL-10 in experimental autoimmune encephalomyelitis and show that, for optimum therapeutic activity, IL-10 must either access the CNS from the peripheral circulation or be delivered directly to it by strategies including the gene transfer described here.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Animals
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Gene Transfer Techniques
- Genetic Vectors/administration & dosage
- Genetic Vectors/immunology
- Injections, Intravenous
- Injections, Intraventricular
- Injections, Subcutaneous
- Interleukin-10/administration & dosage
- Interleukin-10/biosynthesis
- Interleukin-10/genetics
- Interleukin-10/physiology
- Mice
- Mice, Inbred BALB C
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Organ Specificity/genetics
- Organ Specificity/immunology
- Secondary Prevention
- Spinal Cord/immunology
- Spinal Cord/metabolism
Collapse
Affiliation(s)
- D J Cua
- DNAX Research Institute of Molecular and Cellular Biology, Inc., Palo Alto, CA 94304, USA
| | | | | | | | | |
Collapse
|
31
|
Subramanian S, Bourdette DN, Corless C, Vandenbark AA, Offner H, Jones RE. T lymphocytes promote the development of bone marrow-derived APC in the central nervous system. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:370-6. [PMID: 11123314 DOI: 10.4049/jimmunol.166.1.370] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Certain cells within the CNS, microglial cells and perivascular macrophages, develop from hemopoietic myelomonocytic lineage progenitors in the bone marrow (BM). Such BM-derived cells function as CNS APC during the development of T cell-mediated paralytic inflammation in diseases such as experimental autoimmune encephalomyelitis and multiple sclerosis. We used a novel, interspecies, rat-into-mouse T cell and/or BM cell-transfer method to examine the development and function of BM-derived APC in the CNS. Activated rat T cells, specific for either myelin or nonmyelin Ag, entered the SCID mouse CNS within 3-5 days of cell transfer and caused an accelerated recruitment of BM-derived APC into the CNS. Rat APC in the mouse CNS developed from transferred rat BM within an 8-day period and were entirely sufficient for induction of CNS inflammation and paralysis mediated by myelin-specific rat T cells. The results demonstrate that T cells modulate the development of BM-derived CNS APC in an Ag-independent fashion. This previously unrecognized regulatory pathway, governing the presence of functional APC in the CNS, may be relevant to pathogenesis in experimental autoimmune encephalomyelitis, multiple sclerosis, and/or other CNS diseases involving myelomonocytic lineage cells.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen-Presenting Cells/cytology
- Antigen-Presenting Cells/transplantation
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Bone Marrow Transplantation
- Cell Cycle/genetics
- Cell Cycle/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Female
- Genetic Predisposition to Disease
- Immunophenotyping
- Mice
- Mice, SCID
- Rats
- Rats, Inbred Lew
- Severity of Illness Index
- Spinal Cord/cytology
- Spinal Cord/immunology
- Spinal Cord/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transplantation, Heterologous
Collapse
Affiliation(s)
- S Subramanian
- Veterans' Affairs Medical Center, Portland, OR, 97201, USA
| | | | | | | | | | | |
Collapse
|
32
|
Mabon PJ, Weaver LC, Dekaban GA. Inhibition of monocyte/macrophage migration to a spinal cord injury site by an antibody to the integrin alphaD: a potential new anti-inflammatory treatment. Exp Neurol 2000; 166:52-64. [PMID: 11031083 DOI: 10.1006/exnr.2000.7488] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The inflammatory response that ensues during the initial 48 to 72 h after spinal cord injury causes considerable secondary damage to neurons and glia. Infiltration of proinflammatory-activated neutrophils and monocytes/macrophages into the cord contributes to spinal cord injury-associated secondary damage. beta2 integrins play an essential role in leukocyte trafficking and activation and arbitrate cell-cell interactions during inflammation. The beta2 integrin, alphaDbeta2, is expressed on monocytes/macrophages and neutrophils and binds to vascular adhesion molecule-1 (VCAM-1). The increased expression of VCAM-1 during central nervous system (CNS) inflammation likely contributes to leukocyte extravasation into the CNS. Accordingly, blocking the interaction between alphaDbeta2 and VCAM-1 may attenuate the inflammatory response at the SCI site. We investigated whether the administration of monoclonal antibodies (mAbs) specific for the rat alphaD subunit would reduce the inflammatory response after a spinal cord transection injury in rats. At a 1 mg/kg dose two of three anti-alphaD mAbs caused a significant ( approximately 65%) reduction in the number of macrophages at the injury site and one anti-alphaD mAb led to a approximately 43% reduction in the number of neutrophils at the SCI site. Thus, our results support the concept that the alphaDbeta2 integrins play an important role in the trafficking of leukocytes to a site of central nervous system inflammation. This study also offers preliminary evidence that anti-alphaD mAbs can reduce the extravasation of macrophages and, to a lesser extent, neutrophils, to the SCI site.
Collapse
Affiliation(s)
- P J Mabon
- The Neurodegeneration Research Group, The John P. Robarts Research Institute, London, Ontario, N6A 5K8, Canada
| | | | | |
Collapse
|
33
|
da Costa P, Yasuda CL, Scagliusi SM, Diaz-Bardales BM, Maciel E, Damasceno BP, Blotta MH, Tilbery CP, Santos LM. Pattern of cytokine secretion by peripheral blood cells of patients with multiple sclerosis in Brazil. Mult Scler 2000; 6:293-9. [PMID: 11064437 DOI: 10.1177/135245850000600501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Autoimmune T cells play a key role as regulators and effectors of organ-specific autoimmune disease. In multiple sclerosis (MS), activated T cells specific for myelin components produce a plethora of inflammatory cytokines and mediators that contribute to myelin damage. The production of proinflammatory and regulatory cytokines by peripheral blood cells from patients with active and stable MS and healthy controls were examined. The results show that TNF alpha production was somewhat elevated in active MS with no significant increase in the level IFN gamma, whereas in the chronic phase the anti-inflammatory cytokines IL-10 and TGF beta increased, accompanied by a reduction in IFN gamma when stimulated by myelin basic protein. Multiple Sclerosis (2000) 6 293 - 299
Collapse
Affiliation(s)
- P da Costa
- Department of Microbiology and Immunology, Institute of Biology, UNICAMP, Campinas, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liefner M, Siebert H, Sachse T, Michel U, Kollias G, Brück W. The role of TNF-alpha during Wallerian degeneration. J Neuroimmunol 2000; 108:147-52. [PMID: 10900348 DOI: 10.1016/s0165-5728(00)00262-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The role of TNF-alpha in the course of Wallerian degeneration of the sciatic nerve was studied in control and TNF-alpha deficient mice. In control animals, the characteristic phenomena of Wallerian degeneration such as axon and myelin degeneration as well as macrophage recruitment with subsequent myelin removal were observed. In TNF-alpha deficient mice, in contrast, macrophage recruitment into the degenerating nerves was impaired resulting in a delayed myelin removal. However, the myelin phagocytic capacity of macrophages was not affected as it could be demonstrated by a similar myelin load of control and TNF-alpha deficient macrophages. These data indicate that the main function of TNF-alpha during Wallerian degeneration is the induction of macrophage recruitment from the periphery without affecting myelin damage or phagocytosis.
Collapse
Affiliation(s)
- M Liefner
- Department of Neuropathology, Georg-August-Universität, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
35
|
Smith JP, Morris-Downes M, Brennan FR, Wallace GJ, Amor S. A role for alpha4-integrin in the pathology following Semliki Forest virus infection. J Neuroimmunol 2000; 106:60-8. [PMID: 10814783 DOI: 10.1016/s0165-5728(99)00235-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Migration of cells into the central nervous system (CNS) is a pivotal step in the pathogenesis of immune-mediated diseases such as multiple sclerosis (MS), experimental allergic encephalomyelitis (EAE) and virus-induced demyelinating diseases. Such migration is dependent on expression of adhesion molecules. The expression of adhesion molecules in the CNS was studied in Biozzi ABH mice infected with Semliki Forest virus (SFV) A7(74) - an important demyelinating model of MS. Expression of LFA-1alpha/CD11a, LFA-1beta/CD18 and ICAM-1/CD56 were rapidly elevated and remained high whereas MAC-1, CD44 and VCAM-1/CD106 were less widely expressed. The alpha4-integrin VLA-4/CD49d was more specifically associated with CNS lesions. To identify the importance of VLA-4, CD44, ICAM-1 and MAC-1 in the pathogenesis of SFV infection, monoclonal antibodies that block these adhesion molecules were administered in vivo during infection. Anti-VLA-4 treatment dramatically reduced the cellular infiltrates and demyelination within the CNS but did not affect the clearance of virus while antibodies to CD44, ICAM and MAC-1 antibody treatment had no effect. This study demonstrates that SFV infection induces the expression of adhesion molecules within the CNS and that VLA-4 plays an important role in the development of inflammation and demyelination in the CNS following SFV infection.
Collapse
Affiliation(s)
- J P Smith
- Immunology Department, Rayne Institute, United Medical and Dental School of Guy's and St. Thomas' Hospital, London, UK
| | | | | | | | | |
Collapse
|
36
|
Theil DJ, Tsunoda I, Libbey JE, Derfuss TJ, Fujinami RS. Alterations in cytokine but not chemokine mRNA expression during three distinct Theiler's virus infections. J Neuroimmunol 2000; 104:22-30. [PMID: 10683511 DOI: 10.1016/s0165-5728(99)00251-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
DA, GDVII and H101 are neurovirulent strains of Theiler's murine encephalomyelitis virus that cause very different neuropathology and CNS disease when inoculated into SJL/J mice. DA virus causes a chronic demyelinating disease, GDVII virus causes an acute fatal polioencephalomyelitis, and H101 virus causes an acute pachymeningitis with hydrocephalus. Performing RNase protection assays, we detected the same pattern of chemokine (RANTES, MCP-1, IP-10, MIP-1beta, MIP-1alpha and MIP-2) mRNA expression in brain and spinal cord during all three infections. In contrast, IFN-beta and IL-6 mRNA were highly expressed only in GDVII virus infection, whereas high levels of LT-alpha mRNA were only found during DA virus infection. Our study demonstrates that proinflammatory cytokines are involved in the neuropathogenesis of CNS disease and modulate the acute and chronic process underlying different pathologic features of disease.
Collapse
Affiliation(s)
- D J Theil
- Department of Neurology, University of Utah School of Medicine, 30 N 1900 East, RM 3R330, Salt Lake City, UT 84132, USA
| | | | | | | | | |
Collapse
|
37
|
Brod SA, Khan M, Nelson LD, Decuir B, Malone M, Henninger E. Adoptive transfer from interferon-alpha-fed mice is associated with inhibition of active experimental autoimmune encephalomyelitis by decreasing recipient tumor necrosis factor-alpha secretion. J Immunother 2000; 23:235-45. [PMID: 10746550 DOI: 10.1097/00002371-200003000-00008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ingested type I interferon (IFN) suppresses clinical relapse in murine chronic experimental autoimmune encephalomyelitis (EAE), inhibits clinical attacks more effectively than subcutaneous doses, and decreases the adoptive transfer of EAE. To determine whether splenocytes from IFN-fed donors were "suppressor-like" populations, donor SJL/J mice were immunized and fed with mock IFN-alpha or with IFN-alpha every other day for at least 4 weeks after initial clinical attack. Recipients of adoptively transferred CD8+ T cells from mock IFN-alpha-fed donors showed no clinical improvement of clinical disease compared with actively immunized controls. In contrast, recipients of adoptively transferred CD8+ T cells from IFN-alpha-fed donors showed decreased clinical disease compared with recipients of mock IFN-alpha-fed CD8+ T cells. To evaluate the mechanism of protection by donor CD8+ T cells and to determine if ingested IFN-alpha activates natural immunomodulatory cell populations, the authors used the acute EAE model and naïve-fed donor animals as sources of T cells and CD8+ T cells. Con A-activated spleen T cells from naïve nonimmunized mock IFN-alpha-fed donors inhibited actively induced disease and showed decreased recipient TNF-alpha secretion compared with recipients of T cells from mock IFN-fed mice. Donor activated spleen CD8+ T cells from naïve nonimmunized IFN-alpha-fed animals suppressed actively induced EAE in recipients and showed decreased IFN-gamma and TNF-alpha proinflammatory secretion. Decreased recipient TNF-alpha secretion correlates best with the disease protection from IFN-fed T and CD8+ T cells.
Collapse
Affiliation(s)
- S A Brod
- Department of Neurology and the Multiple Sclerosis Research Group, University of Texas Health Science Center at Houston, 77225, USA
| | | | | | | | | | | |
Collapse
|
38
|
Croxford JL, Triantaphyllopoulos KA, Neve RM, Feldmann M, Chernajovsky Y, Baker D. Gene therapy for chronic relapsing experimental allergic encephalomyelitis using cells expressing a novel soluble p75 dimeric TNF receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:2776-81. [PMID: 10679120 DOI: 10.4049/jimmunol.164.5.2776] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a murine relapsing experimental allergic encephalomyelitis (EAE) model, gene therapy to block TNF was investigated with the use of a retroviral dimeric p75 TNF receptor (dTNFR) construct. To effectively produce these TNF inhibitors in vivo, a conditionally immortalized syngeneic fibroblast line was established, using a temperature-sensitive SV40 large T Ag-expressing retrovirus. These cells were subsequently infected with a retrovirus expressing soluble dTNFR. CNS-injected cells could be detected 3 mo after transplantation and were shown to produce the transgene product by immunocytochemistry and ELISA of tissue fluids. These levels of dTNFR protein were biologically active and could significantly ameliorate both acute and relapsing EAE. This cell-based gene-vector approach is ideal for delivering proteins to the CNS and has particular relevance to the control of inflammatory CNS disease.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antigens, CD/administration & dosage
- Antigens, CD/biosynthesis
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Brain Tissue Transplantation/immunology
- Cell Line, Transformed
- Chronic Disease
- Dimerization
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Fibroblasts/immunology
- Fibroblasts/metabolism
- Genetic Therapy/methods
- Genetic Vectors/chemical synthesis
- Genetic Vectors/immunology
- Humans
- Injections, Intraventricular
- Kidney/cytology
- Mice
- Receptors, Tumor Necrosis Factor/administration & dosage
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type II
- Recurrence
- Solubility
Collapse
Affiliation(s)
- J L Croxford
- Neuroinflammation Group, Institute of Neurology, and Department of Clinical Science, Institute of Ophthalmology, University College London, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Juedes AE, Hjelmström P, Bergman CM, Neild AL, Ruddle NH. Kinetics and cellular origin of cytokines in the central nervous system: insight into mechanisms of myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:419-26. [PMID: 10605038 DOI: 10.4049/jimmunol.164.1.419] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental autoimmune encephalomyelitis induced by myelin oligodendrocyte glycoprotein (MOG) in C57BL/6 (H-2b) mice is characterized by early (day 12) acute paralysis, followed by a sustained chronic clinical course that gradually stabilizes. Extensive inflammation and demyelination coincide with clinical signs of disease. To identify the mechanisms of these processes, individual proinflammatory and anti-inflammatory cytokines and chemokines were studied. Sensitive single-cell assays were utilized to determine the cellular origin and kinetics of cytokine production in the CNS. Immunization with MOG35-55 peptide resulted in priming of both Th1 (lymphotoxin, IFN-gamma, and TNF-alpha) and Th2 (IL-4) cells in the spleen. However, only Th1 cells were apparent in the CNS. CD4 T cells that produced IFN-gamma or TNF-alpha were present in the CNS by day 7 after immunization with MOG35-55, peaked at day 20, and then waned. TNF-alpha was also produced in the CNS by Mac-1+ cells. On days 7 and 10 after immunization, the TNF-alpha-producing Mac1+ cells were predominantly microglia. By day 14, a switch occurred in that the Mac1+ TNF-alpha-producing cells had the phenotype of infiltrating macrophages. RANTES, IFN-inducible protein 10 (IP-10), and monocyte chemotactic protein 1 chemokine mRNA were detected in the CNS by day 8 after immunization. The early presence of monocyte chemotactic protein 1 (MCP-1) in the CNS provides a mechanism for the recruitment of macrophages. These data implicate TNF-alpha production by a continuum of T cells, microglia, and macrophages at various times during the course of disease. The importance of Th1 cytokines is highlighted, with little evidence for a role of Th2 cytokines.
Collapse
Affiliation(s)
- A E Juedes
- Department of Epidemiology and Public Health, Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
40
|
Brennan FR, O'Neill JK, Allen SJ, Butter C, Nuki G, Baker D. CD44 is involved in selective leucocyte extravasation during inflammatory central nervous system disease. Immunology 1999; 98:427-35. [PMID: 10583604 PMCID: PMC2326932 DOI: 10.1046/j.1365-2567.1999.00894.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clinical signs of experimental autoimmune encephalomyelitis (EAE) are associated with the selective recruitment of CD4+ memory (CD45RBlow CD44high) T cells into the central nervous system (CNS). However, we have found that many of these recently recruited memory cells are CD44low, suggesting that the CD44 antigen may be involved in, and transiently lost during, the extravasation process. Indeed, administration of a CD44-specific antibody (IM7.8.1) induced leucocyte CD44 shedding and both prevented the development and ameliorated the severity of established EAE by inhibiting mononuclear cell infiltration into the CNS. Trafficking of cells into lymph nodes, however, a property mainly of naïve cells, was essentially unaffected. In contrast, treatment with antibody to very late activation antigen-4 (VLA-4) prevented homing to both the CNS and to lymph nodes. This study contests previous reports that dismissed a role for CD44 in inflammation of the CNS and, coupled with observations in murine dermatitis and arthritis, suggests that CD44 is involved in the homing of primed lymphocytes to sites of inflammation. CD44 should therefore be considered a target for immunotherapy of T-cell-mediated inflammatory diseases, such as multiple sclerosis.
Collapse
Affiliation(s)
- F R Brennan
- Rheumatic Diseases Unit, Western General Hospital, Edinburgh, UK
| | | | | | | | | | | |
Collapse
|
41
|
Bourrié B, Bribes E, Esclangon M, Garcia L, Marchand J, Thomas C, Maffrand JP, Casellas P. The neuroprotective agent SR 57746A abrogates experimental autoimmune encephalomyelitis and impairs associated blood-brain barrier disruption: implications for multiple sclerosis treatment. Proc Natl Acad Sci U S A 1999; 96:12855-9. [PMID: 10536012 PMCID: PMC23131 DOI: 10.1073/pnas.96.22.12855] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a T cell autoimmune disorder that is a widely used animal model for multiple sclerosis (MS) and, as in MS, clinical signs of EAE are associated with blood-brain barrier (BBB) disruption. SR 57746A, a nonpeptide drug without classical immunosuppressive properties, efficiently protected the BBB and impaired intrathecal IgG synthesis (two conventional markers of MS exacerbation) and consequently suppressed EAE clinical signs. This compound inhibited EAE-induced spinal cord mononuclear cell invasion and normalized tumor necrosis factor alpha and IFN-gamma mRNA expression within the spinal cord. These data suggested that pharmacological intervention aimed at inhibiting proinflammatory cytokine expression within the central nervous system provided protection against BBB disruption, the first clinical sign of EAE and probably the key point of acute MS attacks. This finding could lead to the development of a new class of compounds for oral therapy of MS, as a supplement to immunosuppressive agents.
Collapse
Affiliation(s)
- B Bourrié
- Department of Immunopharmacology, Sanofi Recherche, 371 rue du Pr. J. Blayac, 34184 Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee SJ, Benveniste EN. Adhesion molecule expression and regulation on cells of the central nervous system. J Neuroimmunol 1999; 98:77-88. [PMID: 10430040 DOI: 10.1016/s0165-5728(99)00084-3] [Citation(s) in RCA: 222] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Cellular adhesion molecules were initially defined as cell surface structures mediating cell-cell and cell-extracellular matrix (ECM) interactions. Adhesion molecules involved in immune responses have been classified into three families according to their structure: selectins, immunoglobulin (Ig) superfamily, and integrins. It has been well documented that adhesion molecules of these family members (E-selectin, ICAM-1, and VCAM-1) are expressed on brain microvessel endothelial cells in active lesions of multiple sclerosis (MS) brain. In addition, accumulating data show that glial cells can express some of these adhesion molecules upon activation: astrocytes can express ICAM-1, VCAM-1, and E-selectin, and microglia express ICAM-1 and VCAM-1. In vitro studies show that these adhesion molecules are actively regulated by several cytokines which have relevance to MS or experimental autoimmune encephalomyelitis (EAE). In addition, soluble forms of adhesion molecules have been found in the serum and cerebrospinal fluid (CSF) of MS patients, and may be useful diagnostically. Experimental therapy of EAE using antibodies against several adhesion molecules clearly shows that adhesion molecules are critical for the pathogenesis of EAE. Thus far, the function of adhesion molecule expression on brain endothelial and glial cells has not been clearly elucidated. Studies on the possible role of adhesion molecules on brain endothelial and glial cells will be helpful in understanding their involvement in immune responses in the central nervous system (CNS).
Collapse
Affiliation(s)
- S J Lee
- Department of Cell Biology, The University of Alabama at Birmingham, 35294-0005, USA
| | | |
Collapse
|
43
|
Affiliation(s)
- Y Shimizu
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis 55455, USA
| | | | | |
Collapse
|
44
|
Abstract
Microglia are a major ghal component of the central nervous system (CNS) and are extremely sessile. Only a subtype, the perivascular microglia, are regularly replaced from the bone marrow in adult animals. Microglia respond to virtually any, even minor pathological events in the CNS. In most pathological settings microglia are aided by infiltrating hematogenous macrophages. Upon activation microglia and macrophages share most phenotypical markers and can exert similar effector functions. After transection of a CNS fibre tract microglia are insufficiently activated and hematogenous macrophages do not significantly enter the degenerating nerve stump. Thereby myelin debris that contains neurite outgrowth inhibiting activity persists for long time. This is in sharp contrast to the peripheral nervous system in which hematogenous macrophages are rapidly recruited in response to axotomy and clear myelin debris allowing regrowth of axons from the proximal stump. However, CNS lesion paradigms with breakdown of the blood-brain barrier such as cerebral ischemia, brain abscesses and stab wounds elicit prompt microglial activation, macrophage recruitment and debris clearance. There is increasing evidence that microglia play an active part in degenerative CNS diseases. In Alzheimer's disease activated microglia appear to be involved in plaque formation. In experimental globoid cell dystrophy T-cell independent induction of major histocompatibility complex class II molecules on microglia accelerates demyelination. In autoimmune diseases microglia probably have dual functions. Microglia present antigen to infiltrating T cells and exert effector functions thereby locally augmenting immune responses. On the other hand, microglia have the capacity to downregulate T cell responses. In the human acquired immunodeficiency syndrome (AIDS) virus infected macrophages probably introduce the virus to the CNS and in concert with microglia are involved in the pathophysiology of the AIDS dementia complex.
Collapse
Affiliation(s)
- G Stoll
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany.
| | | |
Collapse
|
45
|
Nataf S, Davoust N, Barnum SR. Kinetics of anaphylatoxin C5a receptor expression during experimental allergic encephalomyelitis. J Neuroimmunol 1998; 91:147-55. [PMID: 9846831 DOI: 10.1016/s0165-5728(98)00169-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In this study, we investigated the expression of the C5aR in spinal cords of Lewis rats with experimental allergic encephalomyelitis (EAE). Using in situ hybridization (ISH) we analyzed the kinetics of C5aR at different time points of EAE (preclinical stage, clinical peak, remission phase). We observed that C5aR mRNA was readily detected in the CNS of EAE rats at all the stages of the disease. Using a combination of ISH and immunohistochemistry, we formally demonstrated that C5aR is strongly expressed on microglial cells and hypertrophic astrocytes during EAE. The potential involvement of C5a receptor in EAE physiopathology is discussed.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/immunology
- Female
- Gene Expression/immunology
- Kinetics
- Macrophages/chemistry
- Macrophages/immunology
- Microglia/chemistry
- Microglia/immunology
- Monocytes/chemistry
- Monocytes/immunology
- Multiple Sclerosis/immunology
- RNA, Messenger/analysis
- Rats
- Rats, Inbred Lew
- Receptor, Anaphylatoxin C5a
- Receptors, Complement/genetics
- Receptors, Complement/immunology
- Spinal Cord/blood supply
- Spinal Cord/cytology
- Spinal Cord/immunology
Collapse
Affiliation(s)
- S Nataf
- Department of Microbiology, University of Alabama at Birmingham, 35294, USA
| | | | | |
Collapse
|
46
|
Hjelmström P, Juedes AE, Ruddle NH. Cytokines and antibodies in myelin oligodendrocyte glycoprotein-induced experimental allergic encephalomyelitis. RESEARCH IN IMMUNOLOGY 1998; 149:794-804; discussion 847-8, 855-60. [PMID: 9923635 DOI: 10.1016/s0923-2494(99)80007-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- P Hjelmström
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
47
|
Muñoz-Fernández MA, Fresno M. The role of tumour necrosis factor, interleukin 6, interferon-gamma and inducible nitric oxide synthase in the development and pathology of the nervous system. Prog Neurobiol 1998; 56:307-40. [PMID: 9770242 DOI: 10.1016/s0301-0082(98)00045-8] [Citation(s) in RCA: 286] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Proinflammatory cytokines, tumour necrosis factor (TNF)-alpha, interferon (IFN)-gamma and interleukin (IL)-6, have multiple effects in the central nervous system (CNS) not strictly cytotoxic being involved in controlling neuronal and glial activation, proliferation, differentiation and survival, thus influencing neuronal and glial plasticity, degeneration as well as development and regeneration of the nervous system. Moreover, they can contribute to CNS disorders, including multiple sclerosis. Alzheimer's disease and human immunodeficiency virus-associated dementia complex. Recent results with deficient mice in the expression of those cytokines indicate that they are in general more sensible to insults resulting in neural damage. Some of the actions induced by TNF-alpha, and IFN-gamma, including both beneficial and detrimental, are mediated by inducible nitric oxide synthase (iNOS)-derived nitric oxide (NO) production. NO produced by iNOS may be beneficial by promoting the differentiation and survival of neurons. IL-6 does not induce iNOS, explaining why this cytokine is less often involved in this dual role protection pathology. Some of the proinflammatory as well as the neurotrophic effects of those cytokines also involve upregulation of cell adhesion molecules (CAM). Those apparently conflicting results may be reconciled considering that proinflammatory cytokines are involved in promoting the disease, mostly by inducing expression of CAM leading to alteration of the blood-brain barrier integrity, whereas they have a protective role once disease is established due to its immunosuppressive or neurotrophic role. Understanding the dichotomy pathogenesis/neuroprotection of those cytokines may provide a rationale for better therapeutic strategies.
Collapse
Affiliation(s)
- M A Muñoz-Fernández
- Division of Immunology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
48
|
Eugster HP, Frei K, Kopf M, Lassmann H, Fontana A. IL-6-deficient mice resist myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. Eur J Immunol 1998; 28:2178-87. [PMID: 9692887 DOI: 10.1002/(sici)1521-4141(199807)28:07<2178::aid-immu2178>3.0.co;2-d] [Citation(s) in RCA: 248] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is induced by immunization with myelin components including myelin oligodendrocyte glycoprotein (MOG). Myelin-specific Th1 cells enter the central nervous system (CNS) via binding of very late antigen 4 (VLA-4) to the endothelial vascular cell adhesion molecule 1 (VCAM-1). In the present study, mice with a homologous disruption of the gene encoding IL-6 are found to be resistant to MOG-induced EAE as evidenced by absence of clinical symptoms, minimal infiltration of CD3+ T cells and monocytes into the CNS and lack of demyelination. The failure to induce EAE in IL-6-/- mice is not due to the absence of priming, since lymphocytes of immunized IL-6-/- mice proliferate in response to MOG and produce pro-inflammatory cytokines including IL-2 and IFN-gamma. However, in MOG-immunized IL-6-/- mice, serum anti-MOG antibody titers were found to be drastically reduced. This observation is unlikely to be responsible for resistance to EAE, because B cell-deficient (microMT) mice proved to be fully susceptible to the disease. A striking difference between MOG-immunized wild-type (wt) and IL-6-/- mice was the expression of endothelial VCAM-1 and ICAM-1, which were dramatically up-regulated in the CNS in wt but not in IL-6-/- mice. Taking into account recent studies on the role of VCAM-1 in the entry of Th1 cells into the CNS, the absence of VCAM-1 on endothelial cells in IL-6-/- mice may explain their resistance to EAE.
Collapse
Affiliation(s)
- H P Eugster
- Department of Internal Medicine, University Hospital Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
49
|
Rajan AJ, Klein JDS, Brosnan CF. The Effect of γδ T Cell Depletion on Cytokine Gene Expression in Experimental Allergic Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.12.5955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis, we showed previously that depletion of γδ T cells using the mAb GL3 immediately before disease onset, or during the chronic phase, significantly ameliorated clinical severity. We now report on the effect of γδ T cell depletion on expression of five cytokine genes, IL-1, IL-6, TNF, lymphotoxin, and IFN-γ in spinal cords of mice during the pre-onset, onset, height, and recovery phases of EAE, and on expression of type II nitric oxide synthase. In control animals, the mRNAs for IL-1 and IL-6 rose dramatically at disease onset and peaked before disease height, whereas the mRNAs for TNF, lymphotoxin, and IFN-γ rose more slowly and peaked with peak of disease. In GL3-treated animals, a dramatic reduction in all five cytokines was noted at disease onset, but only IFN-γ remained significantly reduced at a time point equivalent to height of disease in control animals. ELISA data confirmed the reduced levels of IL-1 and IL-6 at disease onset in GL3-treated animals, and pathologic analysis demonstrated a marked reduction in meningeal infiltrates at the same time point. Studies of type II NOS also demonstrated a significant reduction in both mRNA and protein expression at the height of disease in GL3-treated animals. These results suggest that γδ T cells contribute to the pathogenesis of EAE by regulating the influx of inflammatory cells into the spinal cord and by augmenting the proinflammatory cytokine profile of the inflammatory infiltrates.
Collapse
Affiliation(s)
- Alice J. Rajan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | - Celia F. Brosnan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
50
|
Selmaj K, Walczak A, Mycko M, Berkowicz T, Kohno T, Raine CS. Suppression of experimental autoimmune encephalomyelitis with a TNF binding protein (TNFbp) correlates with down-regulation of VCAM-1/VLA-4. Eur J Immunol 1998; 28:2035-44. [PMID: 9645385 DOI: 10.1002/(sici)1521-4141(199806)28:06<2035::aid-immu2035>3.0.co;2-a] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The effect of a novel TNF binding protein (TNFbp), a polyethylene glycol-linked form of the type I soluble receptor of TNF, on the expression of adhesion molecules has been investigated with a passive transfer model of experimental autoimmune encephalomyelitis (EAE) in SJL/J mice. The expression of L-selectin, VLA-4 and LFA-1 on spleen cells of EAE animals treated with TNFbp or saline was examined by FACS analysis. The expression of VCAM-1 and ICAM-1 was investigated by immunochemistry in spinal cord tissue of SJL/J mice with EAE. In animals sensitized for EAE and treated with TNFbp, the expression of VCAM-1 in the central nervous system as well as VLA-4 on spleen cells was clearly diminished. Reduction in VCAM-1 staining and VLA-4 expression corresponded to inhibition of inflammation in the spinal cord and to prevention of clinical signs of EAE. The results have also shown that myelin basic protein responses as well as non-antigen-specific responses were not diminished in animals treated with TNFbp. The findings suggest that TNFbp might prevent EAE development by modulating the expression of VCAM-1 and VLA-4.
Collapse
MESH Headings
- Animals
- CD4-CD8 Ratio
- Carrier Proteins/pharmacology
- Disease Models, Animal
- Down-Regulation/drug effects
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Immunophenotyping
- Integrin alpha4beta1
- Integrins/biosynthesis
- Intercellular Adhesion Molecule-1/biosynthesis
- L-Selectin/biosynthesis
- Lymphocyte Function-Associated Antigen-1/biosynthesis
- Mice
- Receptors, Lymphocyte Homing/biosynthesis
- Receptors, Tumor Necrosis Factor
- Receptors, Tumor Necrosis Factor, Type I
- Spleen/cytology
- Spleen/metabolism
- T-Lymphocytes/classification
- T-Lymphocytes/immunology
- Tumor Necrosis Factor Decoy Receptors
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Vascular Cell Adhesion Molecule-1/biosynthesis
Collapse
Affiliation(s)
- K Selmaj
- Department of Neurology, Medical Academy of Lodz, Poland.
| | | | | | | | | | | |
Collapse
|