1
|
Wang Y, Zhang J, Luo P, Zhu J, Feng J, Zhang HL. Tumor necrosis factor-α in Guillain-Barré syndrome, friend or foe? Expert Opin Ther Targets 2016; 21:103-112. [PMID: 27817222 DOI: 10.1080/14728222.2017.1258402] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ying Wang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Jingdian Zhang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Peijuan Luo
- Norman Bethune Health Science Center, Jilin University, Changchun, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Jiachun Feng
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| | - Hong-Liang Zhang
- Neuroscience Center, Department of Neurology, the First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Lehmann HC, Hughes RAC, Kieseier BC, Hartung HP. Recent developments and future directions in Guillain-Barré syndrome. J Peripher Nerv Syst 2013; 17 Suppl 3:57-70. [PMID: 23279434 DOI: 10.1111/j.1529-8027.2012.00433.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Guillain-Barré syndrome (GBS) encompasses a spectrum of acquired neuropathic conditions characterized by inflammatory demyelinating or axonal peripheral neuropathy with acute onset. Clinical and experimental studies in the past years have led to substantial progress in epidemiology, pathogenesis of GBS variants, and identification of prognostic factors relevant to treatment. In this review we provide an overview and critical assessment of the most recent developments and future directions in GBS research.
Collapse
Affiliation(s)
- Helmar C Lehmann
- Department of Neurology, Heinrich-Heine-University, Medical School, Moorenstrasse 5, Düsseldorf, Germany
| | | | | | | |
Collapse
|
3
|
The role of cytokines in Guillain-Barré syndrome. J Neurol 2010; 258:533-48. [PMID: 21104265 DOI: 10.1007/s00415-010-5836-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 10/13/2010] [Accepted: 11/09/2010] [Indexed: 12/17/2022]
Abstract
Cytokines play an important role in the pathogenesis of autoimmune diseases including Guillain-Barré syndrome (GBS) and its animal model experimental autoimmune neuritis (EAN). In this article, we reviewed the current knowledge of the role of cytokines such as TNF-α, IFN-γ, IL-1β, IL-6, IL-12, IL-18, IL-23, IL-17, IL-10, IL-4 and chemokines in GBS and EAN as unraveled by studies both in the clinic and the laboratory. However, these studies occasionally yield conflicting results, highlighting the complex role that cytokines play in the disease process. Efforts to modulate cytokine function in GBS and other autoimmune disease have shown efficiency indicating that cytokines are important therapeutic targets.
Collapse
|
4
|
Upregulation of erythropoietin in rat peripheral nervous system with experimental autoimmune neuritis. Brain Res 2010; 1333:82-90. [DOI: 10.1016/j.brainres.2010.03.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 03/18/2010] [Accepted: 03/21/2010] [Indexed: 01/21/2023]
|
5
|
Kraft AD, McPherson CA, Harry GJ. Heterogeneity of microglia and TNF signaling as determinants for neuronal death or survival. Neurotoxicology 2009; 30:785-93. [PMID: 19596372 PMCID: PMC3329780 DOI: 10.1016/j.neuro.2009.07.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/01/2009] [Indexed: 12/30/2022]
Abstract
Microglia do not constitute a single, uniform cell population, but rather comprise cells with varied phenotypes, some which are beneficial and others that may require active regulatory control. Thus, gaining a better understanding of the heterogeneity of resident microglia responses will contribute to any interpretation regarding the impact of any such response in the brain. Microglia are the primary source of the pro-inflammatory cytokine, tumor necrosis factor (TNF) that can initiate various effects through the activation of membrane receptors. The TNF p55 receptor contains a death domain and activation normally leads to cellular apoptosis; however, under specific conditions, receptor activation can also lead to the activation of NF-kappaB and contribute to cell survival. These divergent outcomes have been linked to receptor localization with receptor internalization leading to cell death and membrane localization supporting cell survival. A second TNF receptor, TNF p75 receptor, is normally linked to cell growth and survival, however, it can cooperate with the p55 receptor and contribute to cell death. Thus, while an elevation in TNFalpha in the brain is often considered an indicator of microglia activation and neuroinflammation, a number of factors come into play to determine the final outcome. Data are reviewed demonstrating that heterogeneity in morphological response of microglia and the expression of TNFalpha and TNF receptors are critical in identifying and characterizing neurotoxic events as they relate to neuroinflammation, neuronal damage and in stimulating neuroprotection.
Collapse
Affiliation(s)
- Andrew D. Kraft
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - Christopher A McPherson
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| | - G. Jean Harry
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Dept of Health and Human Services, Research Triangle Park, NC
| |
Collapse
|
6
|
Lu MO, Duan RS, Quezada HC, Chen ZG, Mix E, Jin T, Yang X, Ljunggren HG, Zhu J. Aggravation of experimental autoimmune neuritis in TNF-α receptor 1 deficient mice. J Neuroimmunol 2007; 186:19-26. [PMID: 17428547 DOI: 10.1016/j.jneuroim.2007.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2006] [Revised: 02/14/2007] [Accepted: 02/16/2007] [Indexed: 01/19/2023]
Abstract
The role of tumor necrosis factor (TNF)-alpha and its receptors in the pathogenesis of experimental autoimmune neuritis (EAN) induced by P0 peptide 180-199 in TNFR1 (p55) deficient (TNFR1-/-) mice was investigated. Compared to wild type EAN mice, TNFR1-/- EAN mice developed significantly more severe clinical signs, in parallel with enhanced numbers of inflammatory infiltrating cells in peripheral nerves and splenic P0-reactive T cell proliferation, as well as increased obviously MHC class II and CCR3 expression on the macrophages in the cauda equina. Our data indicated that TNF-alpha might have anti-inflammatory effect preventing the development of EAN in this mouse model.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Enzyme-Linked Immunosorbent Assay/methods
- Flow Cytometry/methods
- Histocompatibility Antigens Class II/metabolism
- Immunization/methods
- Interferon-gamma/metabolism
- Interleukin-4/metabolism
- Leukocytes, Mononuclear/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin P0 Protein
- Neuritis, Autoimmune, Experimental/chemically induced
- Neuritis, Autoimmune, Experimental/genetics
- Neuritis, Autoimmune, Experimental/pathology
- Neuritis, Autoimmune, Experimental/physiopathology
- Receptors, CCR3
- Receptors, Chemokine/metabolism
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Schwann Cells/pathology
- Severity of Illness Index
- Thymidine/pharmacokinetics
Collapse
Affiliation(s)
- Ming-Ou Lu
- Division of Neurodegeneration and Neuroinflammation, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Taylor JM, Pollard JD. Soluble TNFR1 inhibits the development of experimental autoimmune neuritis by modulating blood–nerve-barrier permeability and inflammation. J Neuroimmunol 2007; 183:118-24. [PMID: 17196669 DOI: 10.1016/j.jneuroim.2006.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Revised: 11/23/2006] [Accepted: 11/27/2006] [Indexed: 11/18/2022]
Abstract
The role of TNFalpha/LTalpha during EAN induced by active immunization with peripheral nerve myelin was examined by administering a recombinant soluble chimeric form of human TNF receptor 1 (TNFR1-IgG). TNFalpha and LTalpha do not directly contribute to neurological deficit during EAN since treatment with TNFR1-IgG after onset failed to alter the course of disease. Prophylaxis with a single dose of TNFR1-IgG delayed the onset of EAN and was accompanied initially by inhibition of blood-nerve-barrier permeability and inflammation. Subsequently, the number of infiltrating macrophages and blood-nerve-barrier permeability increased but the disease symptoms remained mild for five days (on average a limp tail) after which severe EAN developed. The antibody titer to peripheral nerve myelin was unaltered by prophylaxis with TNFR1-IgG. The markedly altered tempo of disease onset after TNFR1-IgG prophylaxis indicates that TNFalpha and/or LTalpha have a key role in the development of blood-nerve-barrier permeability and the coupling of macrophage activation and recruitment to peripheral nerve pathology during EAN.
Collapse
MESH Headings
- Animals
- Antibodies/administration & dosage
- Antibodies/blood
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/physiopathology
- Capillary Permeability/drug effects
- Capillary Permeability/physiology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Inflammation/etiology
- Inflammation/prevention & control
- Macrophages/drug effects
- Male
- Myelin Sheath/immunology
- Rats
- Rats, Inbred Lew
- Receptors, Tumor Necrosis Factor, Type I/administration & dosage
- Time Factors
Collapse
Affiliation(s)
- Jude Matthew Taylor
- Neurology Laboratory, Department of Medicine (D06), University of Sydney, Australia.
| | | |
Collapse
|
8
|
Kurz M, Pischel H, Hartung HP, Kieseier BC. Tumor necrosis factor-alpha-converting enzyme is expressed in the inflamed peripheral nervous system. J Peripher Nerv Syst 2005; 10:311-8. [PMID: 16221290 DOI: 10.1111/j.1085-9489.2005.10309.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is considered to play a critical role in the pathogenesis of immune-mediated inflammatory demyelinating disorders of the peripheral nervous system (PNS). Processing of membrane-bound inactive pro-TNF-alpha into the active soluble cytokine is mediated by a sheddase, the so-called TNF-alpha-converting enzyme (TACE), a member of the A Disintegrin and Metalloproteinase (ADAM) family. We explored the expression of TACE (ADAM-17) in sciatic nerves from Lewis rats with experimental autoimmune neuritis (EAN), an animal model of the Guillain-Barré syndrome (GBS), an immune-mediated polyradiculoneuropathy. To extend our study to human disease, sural nerve biopsies from GBS patients were investigated by immunohistochemistry. In EAN, T lymphocytes could be defined as the cellular source of ADAM-17 with peak expression levels at maximum clinical disease severity. Similarly, in human sural nerves, ADAM-17-expressing T cells could be localized primarily within the epi- and perineurium, whereas in control sections from patients with non-inflammatory neuropathies, no expression could be depicted. Our findings indicate that ADAM-17 is upregulated during EAN and expressed in nerves of GBS patients and thus may contribute to the pathogenesis of inflammatory demyelination of the PNS.
Collapse
MESH Headings
- ADAM Proteins/metabolism
- ADAM17 Protein
- Animals
- Antigens, CD/metabolism
- Cell Count/methods
- Disease Models, Animal
- Ectodysplasins
- Female
- Freund's Adjuvant
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Enzymologic/physiology
- Guillain-Barre Syndrome/metabolism
- Guillain-Barre Syndrome/pathology
- Humans
- Immunohistochemistry/methods
- Membrane Proteins/metabolism
- Neuritis, Autoimmune, Experimental/chemically induced
- Neuritis, Autoimmune, Experimental/complications
- Neuritis, Autoimmune, Experimental/metabolism
- Rats
- Rats, Inbred Lew
- Sciatic Neuropathy/etiology
- Sciatic Neuropathy/metabolism
- Sural Nerve/metabolism
- Time Factors
- Tumor Necrosis Factors/metabolism
Collapse
Affiliation(s)
- Martin Kurz
- Department of Neurology, Research Group for Clinical and Experimental Neuroimmunology, Heinrich-Heine-University, Duesseldorf, Germany
| | | | | | | |
Collapse
|
9
|
Apoptotic Cell Death in Experimental Autoimmune Encephalomyelitis. EXPERIMENTAL MODELS OF MULTIPLE SCLEROSIS 2005. [PMCID: PMC7122661 DOI: 10.1007/0-387-25518-4_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Particularly in the vulnerable CNS with a low capacity for regeneration specialized mechanisms must be active for the fast and gentle elimination of dysregulated autoaggressive immune cells. In EAE, local apoptosis of autoimmune T-cells has been identified as a safe means for the removal of these unwanted cells. T-cell apoptosis in situ followed by phagocytic clearance of apoptotic remnants by glia assures a minimum of detrimental bystander damage to the local parenchyma and down-regulates the local inflammatory reaction. The pharmacological augmentation of local apoptosis of inflammatory effector cells might gain therapeutic importance also in human neuroimmunological diseases such as multiple sclerosis.
Collapse
|
10
|
Correale J, Villa A. The neuroprotective role of inflammation in nervous system Injuries. J Neurol 2004; 251:1304-16. [PMID: 15592725 DOI: 10.1007/s00415-004-0649-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 09/02/2004] [Indexed: 12/16/2022]
Abstract
The contribution of inflammation to the pathogenesis of several nervous system disorders has long been established. Other observations, however, indicate that both inflammatory cells and mediators may also have beneficial functions, assisting in repair and recovery processes. There is compelling evidence to indicate that in the injured nervous system, as in other tissues, macrophages are needed at an early stage after injury in order for healing to take place. Likewise, activated T cells of a particular specificity can reduce the spread of damage. This neuroprotective effect of T cells may be caused, at least in part, by the production of neurotrophic factors such as neurotrophin-3 or brain-derived neurotrophic factor. Interestingly, recent findings indicate that immune cells are able to produce a variety of neurotrophic factors which promote neuronal survival and may also mediate anti-inflammatory effects. Numerous cytokines are induced after nervous system injuries. Some cytokines, such as TNF-alpha, IL-1 and IFN-gamma, are well known for their promotion of inflammatory responses. However, these cytokines also have immunosuppressive functions and their subsequent expression also assists in repair or recovery processes, suggesting a dual role for some pro-inflammatory cytokines. This should be clarified, as it may be crucial in the design of therapeutic strategies to target specific cytokine(s). Finally, there is a growing body of evidence to show that autoreactive IgM antibodies may constitute an endogenous system of tissue repair, and therefore prove of value as a therapeutic strategy. Available evidence would appear to indicate that the inflammatory response observed in several neurological conditions is more complex than previously thought. Therefore, the design of more effective therapies depends on a clear delineation of the beneficial and detrimental effects of inflammation.
Collapse
Affiliation(s)
- Jorge Correale
- Raúl Carrea Institute for Neurological Research, FLENI, Montañeses 2325, 1428, Buenos Aires, Argentina.
| | | |
Collapse
|
11
|
Weishaupt A, Kreiss M, Gold R, Herrmann T. Modulation of experimental autoimmune encephalomyelitis by administration of cells expressing antigenic peptide covalently linked to MHC class II. J Neuroimmunol 2004; 152:11-9. [PMID: 15223233 DOI: 10.1016/j.jneuroim.2004.03.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2004] [Revised: 03/08/2004] [Accepted: 03/08/2004] [Indexed: 10/26/2022]
Abstract
The MHC class II molecule RT1Bl covalently linked with gpMBP-71-90 was expressed in P80 cells (mouse mastocytoma P815 expressing rat-CD80) and i.v. injection ameleriorated active and adoptive transfer (AT) experimental autoimmune encephalomyelitis (EAE) in Lewis rats. Spinal cord of animals with AT-EAE showed significant increase of apoptotic T-cells at maximum of disease after injection of P80-RT1Bl-MBP-71-90 but not of P80RT1Bl or P80 cells. The data demonstrate a possible therapeutic effect on EAE by provision of T-cell receptor (TCR) and costimulatory signals by genetically engineered antigen presenting cells (APC) and suggest induction of T-cell apoptosis as important mechanism of action.
Collapse
Affiliation(s)
- Andreas Weishaupt
- Department of Neurology, Clinical Research Group for Multiple Sclerosis and Neuroimmunology, Julius-Maximilians Universität, Josef-Schneider-Strasse 11, 97080 Würzburg, Germany
| | | | | | | |
Collapse
|
12
|
Wiendl H, Hohlfeld R. Therapeutic approaches in multiple sclerosis: lessons from failed and interrupted treatment trials. BioDrugs 2003; 16:183-200. [PMID: 12102646 DOI: 10.2165/00063030-200216030-00003] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The therapy for multiple sclerosis (MS) has changed dramatically over the past decade. Recent immunobiological findings and current pathophysiological concepts together with advances in biotechnology, improvements in clinical trial design and development of magnetic resonance imaging have led to a variety of evaluable therapeutic approaches in MS. However, in contrast to the successfully introduced and established immunomodulatory therapies (e.g. interferon-beta and glatiramer acetate), there have been a remarkable number of therapeutic failures as well. Despite convincing immunological concepts, impressive data from animal models and promising results from phase I/II studies, the drugs and strategies investigated showed no benefit or even turned out to have unexpectedly severe adverse effects. Although to date there is no uniformly accepted model for MS, there is agreement on the significance of inflammatory events mediated by autoreactive T cells in the CNS. These can be modified therapeutically at the individual steps of a hypothetical pathogenetic cascade. Crucial corners like: the prevalence and peripheral activation of CNS-autoreactive T cells in the periphery;adhesion and penetration of T cells into the CNS;local activation and proliferation and;de- and remyelination processes can be targeted through their putative mediators. Like a 'specificity pyramid', therapeutic approaches therefore cover from general immunosuppression up to specific targeting of T-cell receptor peptide major histocompatibility (MHC) complex. We discuss in detail clinical MS trials that failed or were discontinued for other reasons. These trials include cytokine modulators [tumour necrosis factor (TNF)-alpha antagonists, interleukin-10, interleukin-4, transforming growth factor-beta2], immunosuppressive agents (roquinimex, gusperimus, sulfasalazine, cladribine), inducers of remyelination [intravenous immunoglobulins (IVIg)], antigen-derived therapies [oral tolerance, altered peptide ligands (APL), MHC-Peptide blockade], T cell and T-cell receptor directed therapies (T cell vaccination, T-cell receptor peptide vaccination), monoclonal antibodies against leucocyte differentiation molecules (anti-CD3, anti-CD4), and inactivation of circulating T cells (extracorporeal photopheresis). The main conclusions that can be drawn from these 'negative' experiences are as follows. Theoretically promising agents may paradoxically increase disease activity (lenercept, infliximab), be associated with unforeseen adverse effects (e.g. roquinimex) or short-term favourable trends may reverse with prolonged follow-up (e.g. sulfasalzine). One should not be too enthusiastic about successful trials in animal models (TNFalpha blockers; oral tolerance; remyelinating effect of IVIg) nor be irritated by non-scientific media hype (deoxyspergualine; bone marrow transplantation). More selectivity can imply less efficacy (APL, superselective interventions like T-cell receptor vaccination) and antigen-related therapies can stimulate rather than inhibit encephalitogenic cells. Failed strategies are of high importance for a critical revision of assumed immunopathological mechanisms, their neuroimaging correlates, and for future trial design. Since failed trials add to our growing understanding of multiple sclerosis, 'misses' are nearly as important to the scientific process as the 'hits'.
Collapse
Affiliation(s)
- Heinz Wiendl
- Department of Neurology, School of Medicine, University of Tuebingen, Tuebingen, Germany.
| | | |
Collapse
|
13
|
Mäurer M, Toyka KV, Gold R. Immune mechanisms in acquired demyelinating neuropathies: lessons from animal models. Neuromuscul Disord 2002; 12:405-14. [PMID: 12062260 DOI: 10.1016/s0960-8966(01)00302-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The peripheral nervous system (PNS) is the target for a heterogenous immune attack mediated by T-cells, B-cells, and macrophages. The interaction of the humoral and cellular immune system with the structural components in the peripheral nervous system may determine the extent of inflammation and possibly repair mechanisms. The animal model experimental autoimmune neuritis (EAN) allows detailed study of the various effector pathways and tests novel therapeutic strategies in vivo. Unexpectedly, involvement of the immune system is also found in animal models for inherited neuropathies and in its human counterpart Charcot-Marie-Tooth (CMT) disease, suggesting an autoimmune reaction triggered by the genetically determined demyelinating disorder. A better understanding of immune regulation and its failure in the peripheral nervous system may help to develop more specific and more effective immunotherapies.
Collapse
Affiliation(s)
- Mathias Mäurer
- Department of Neurology, Section of Developmental Neurobiology, Julius-Maximilians-Universität, Würzburg, Germany
| | | | | |
Collapse
|
14
|
Lee Y, Shin T. Expression of constitutive endothelial and inducible nitric oxide synthase in the sciatic nerve of Lewis rats with experimental autoimmune neuritis. J Neuroimmunol 2002; 126:78-85. [PMID: 12020959 DOI: 10.1016/s0165-5728(02)00058-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study examined the expression of constitutive endothelial nitric oxide synthase (eNOS) and inducible NOS (iNOS) in the sciatic nerve of Lewis rats with experimental autoimmune neuritis (EAN). Western blot analysis showed that both eNOS and iNOS expressions in the sciatic nerves of rats increased significantly during the peak stage of EAN, but declined thereafter. Only minimal amounts of these enzymes were identified in normal rat sciatic nerves. Immunohistochemical studies showed that eNOS was increased in vascular endothelial cells and Schwann cells, but not in inflammatory cells, during the peak stage of EAN. However, iNOS was found mainly in inflammatory macrophages in sciatic nerve EAN lesions.These findings suggest that, depending on the stage of peripheral nervous system autoimmune disease, the increased expressions of both eNOS and iNOS might be involved in either the production of detrimental effects during the induction stage of EAN or in the recovery from EAN paralysis.
Collapse
Affiliation(s)
- Yongduk Lee
- Department of Veterinary Medicine, Graduate School (Brain Korea 21 Project), Cheju National University, 690-756, Republic of Korea
| | | |
Collapse
|
15
|
Abstract
The liver has emerged as an organ with distinct immunological properties. In this review, we summarize evidence that shows that the liver can remove apoptotic, or non-apoptotic but activated, CD8+ T cells from the circulation and induce apoptosis in these activated T cells by either active or passive mechanisms. Hepatitis viruses, particularly hepatitis C virus, often establish persistent infection. We review evidence that suggests that these viruses exploit intrahepatic tolerance mechanisms to protect themselves from immune attack.
Collapse
Affiliation(s)
- Sun Park
- The David H Smith Center for VaccineBiology and Immunology, The University of Rochester, Rochester, New York 14642, USA
| | | | | | | |
Collapse
|
16
|
Stienekemeier M, Falk K, Rötzschke O, Weishaupt A, Schneider C, Toyka KV, Gold R, Strominger JL. Vaccination, prevention, and treatment of experimental autoimmune neuritis (EAN) by an oligomerized T cell epitope. Proc Natl Acad Sci U S A 2001; 98:13872-7. [PMID: 11717443 PMCID: PMC61134 DOI: 10.1073/pnas.241504598] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2001] [Indexed: 11/18/2022] Open
Abstract
Using a polypeptide oligomer harboring 16 repeats of the neuritogenic epitope (aa 58-73) of myelin P2 protein separated by spacers, enhancement of the immune response to the P2 protein, an important neuritogenic autoantigen in experimental autoimmune neuritis (EAN), was attempted. In contrast to a previous study with PLP-16-mer antigen-specific response of T cells was attenuated at all doses examined to a variable degree. Treatment of Lewis rats with the P2-16-mer up to 2 months before immunization with P2(53-78) (vaccination) or after immunization but before appearance of disease (prevention) had a strong tolerizing effect against the induction of EAN on immunization with P2(53-78). Moreover, rats injected with 200 microg of the P2-16-mer i.v. on day 11 after disease induction, at which time the initial signs of disease had appeared, were almost completely protected against progression of clinical disease, whereas animals treated with the same amount of monomeric control peptide developed severe disease (treatment). Similar results were obtained by i.v. treatment of adoptive-transfer EAN with the P2-16-mer. The lack of clinical signs of disease after 16-mer therapy could be correlated with a reduced proliferative response of P2(53-78)-specific lymph node cells. The frequency of apoptotic T cells in sciatic nerve or in lymph node cells, however, was not increased by the 16-mer treatment, suggesting that induction of anergy or other forms of peripheral tolerance may be responsible for the effect. Thus, the oligomerized P2 peptide antigen was highly effective in all three treatment modalities examined in this specific autoreactive T cell-mediated immune response.
Collapse
MESH Headings
- Animals
- Apoptosis
- Autoantigens/immunology
- Autoantigens/therapeutic use
- Cell Division
- Cell Line
- Down-Regulation
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/therapeutic use
- Immunotherapy/methods
- Lymph Nodes/immunology
- Myelin P2 Protein/immunology
- Myelin P2 Protein/therapeutic use
- Neuritis, Autoimmune, Experimental/immunology
- Neuritis, Autoimmune, Experimental/prevention & control
- Neuritis, Autoimmune, Experimental/therapy
- Oligopeptides/immunology
- Oligopeptides/therapeutic use
- Peptide Fragments/immunology
- Peptide Fragments/therapeutic use
- Rats
- Rats, Inbred Lew
- Solubility
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Vaccination/methods
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/therapeutic use
Collapse
Affiliation(s)
- M Stienekemeier
- Department of Neurology, Clinical Research Group for Multiple Sclerosis and Neuroimmunology, Julius-Maximilians Universität, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Weishaupt A, Brück W, Hartung T, Toyka KV, Gold R. Schwann cell apoptosis in experimental autoimmune neuritis of the Lewis rat and the functional role of tumor necrosis factor-alpha. Neurosci Lett 2001; 306:77-80. [PMID: 11403962 DOI: 10.1016/s0304-3940(01)01877-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Schwann cell (SC) apoptosis may be a critical factor challenging nerve remyelination and regeneration in experimental autoimmune neuritis (EAN) in the Lewis rat. We therefore analyzed the fate of SC during high-dose antigen therapy of adoptive transfer-(AT-) EAN using rhP2 protein. P2 antigen therapy was associated with an increase of tumor necrosis factor (serum levels 1 h after intravenous (i.v.) injection and an augmentation of T-cell apoptosis. Antigen specific therapy had no clear effect on SC apoptosis. The effects on SC apoptosis were determined by morphological criteria or by in situ tailing (IST) followed by immunocytochemical analysis. Secondly, we neutralized TNF-alpha, released in abundance by antigen treatment but only in small concentrations during natural disease course. We found that the addition of a TNF-alpha neutralizing antiserum resulted in a significant decrease in the rate of SC apoptosis in vivo compared to animals treated with control antigen rhP0 or with rhP2 only.
Collapse
Affiliation(s)
- A Weishaupt
- Department of Neurology, Clinical Research Group for Multiple Sclerosis and Neuroimmunology, Julius-Maximilians Universität Würzburg, Josef-Schneider-Strasse 11, D-97080, Würzburg, Germany.
| | | | | | | | | |
Collapse
|
18
|
Kiefer R, Kieseier BC, Stoll G, Hartung HP. The role of macrophages in immune-mediated damage to the peripheral nervous system. Prog Neurobiol 2001; 64:109-27. [PMID: 11240209 DOI: 10.1016/s0301-0082(00)00060-5] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophage-mediated segmental demyelination is the pathological hallmark of autoimmune demyelinating polyneuropathies, including the demyelinating form of Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy. Macrophages serve a multitude of functions throughout the entire pathogenetic process of autoimmune neuropathy. Resident endoneurial macrophages are likely to act as local antigen-presenting cells by their capability to express major histocompatibility complex antigens and costimulatory B7-molecules, and may thus be critical in triggering the autoimmune process. Hematogenous infiltrating macrophages then find their way into the peripheral nerve together with T-cells by the concerted action of adhesion molecules, matrix metalloproteases and chemotactic signals. Within the nerve, macrophages regulate inflammation by secreting several pro-inflammatory cytokines including IL-1, IL-6, IL-12 and TNF-alpha. Autoantibodies are likely to guide macrophages towards their myelin or primarily axonal targets, which then attack in a complement-dependent and receptor-mediated manner. In addition, non-specific tissue damage occurs through the secretion of toxic mediators and cytokines. Later, macrophages contribute to the termination of inflammation by promoting T-cell apoptosis and expressing anti-inflammatory cytokines including TGF-beta1 and IL-10. During recovery, they are tightly involved in allowing Schwann cell proliferation, remyelination and axonal regeneration to proceed. Macrophages, thus, play dual roles in autoimmune neuropathy, being detrimental in attacking nervous tissue but also salutary, when aiding in the termination of the inflammatory process and the promotion of recovery.
Collapse
Affiliation(s)
- R Kiefer
- Department of Neurology, Westfälische Wilhelms-Universität, Albert-Schweitzer-Strasse 33, D-48129 Münster, Germany.
| | | | | | | |
Collapse
|
19
|
Weishaupt A, Jander S, Brück W, Kuhlmann T, Stienekemeier M, Hartung T, Toyka KV, Stoll G, Gold R. Molecular mechanisms of high-dose antigen therapy in experimental autoimmune encephalomyelitis: rapid induction of Th1-type cytokines and inducible nitric oxide synthase. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:7157-63. [PMID: 11120847 DOI: 10.4049/jimmunol.165.12.7157] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
High-dose Ag administration induces apoptotic death of autoreactive T cells and is an effective therapy of experimental autoimmune diseases of the nervous system. To explore the role of cytokines in Ag-specific immunotherapy, we analyzed mRNA induction and protein expression for the proinflammatory cytokines TNF-alpha and IFN-gamma, the anti-inflammatory cytokine IL-10, and the cytokine-inducible NO synthase (iNOS) during high-dose Ag therapy of adoptive transfer experimental autoimmune encephalomyelitis (AT-EAE) in the Lewis rat. Using semiquantitative and competitive RT-PCR, we found 5- to 6-fold induction of TNF-alpha mRNA and 3-fold induction of IFN-gamma mRNA in the spinal cord that occurred within 1 h after i.v. injection of Ag and was accompanied by a 2-fold increase of iNOS mRNA. Both IFN-gamma and iNOS mRNA remained elevated for at least 6 h, whereas TNF-alpha mRNA was already down-regulated 6 h after Ag injection. A comparable time course was found for circulating serum levels of TNF-alpha and IFN-gamma. IL-10 mRNA levels did not change significantly following Ag injection. Neutralization of TNF-alpha by anti-TNF-alpha antiserum in vivo led to a significant decrease in the rate of T cell and oligodendrocyte apoptosis induced by high-dose Ag administration, but did not change the beneficial clinical effect of Ag therapy. Our data suggest profound activation of proinflammatory but not of anti-inflammatory cytokine gene expression by high-dose Ag injection. Functionally, TNF-alpha contributes to increased apoptosis of both autoaggressive T cells and oligodendrocytes in the target organ and may thereby play a dual role in this model of Ag-specific therapy of CNS autoimmune diseases.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Autoantigens/administration & dosage
- Autoantigens/therapeutic use
- Cell Movement/immunology
- Cytokines/biosynthesis
- Cytokines/blood
- Disease Progression
- Dose-Response Relationship, Immunologic
- Drug Administration Schedule
- Encephalomyelitis, Autoimmune, Experimental/enzymology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Enzyme Induction/genetics
- Enzyme Induction/immunology
- Female
- Gene Expression Regulation/immunology
- Guinea Pigs
- Immune Sera/administration & dosage
- Injections, Intravenous
- Myelin Basic Protein/administration & dosage
- Myelin Basic Protein/immunology
- Myelin Basic Protein/therapeutic use
- Nitric Oxide Synthase/biosynthesis
- Nitric Oxide Synthase/genetics
- Nitric Oxide Synthase Type II
- RNA, Messenger/biosynthesis
- Rats
- Rats, Inbred Lew
- Spinal Cord/enzymology
- Spinal Cord/immunology
- Spinal Cord/pathology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- A Weishaupt
- Department of Neurology, Neuroimmunology Branch and Clinical Research Group for Multiple Sclerosis, Julius-Maximilians Universität, Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|