1
|
Defects of CTLA-4 Are Associated with Regulatory T Cells in Myasthenia Gravis Implicated by Intravenous Immunoglobulin Therapy. Mediators Inflamm 2020; 2020:3645157. [PMID: 32148437 PMCID: PMC7042523 DOI: 10.1155/2020/3645157] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Myasthenia gravis (MG) is a CD4+ T cell-dependent autoimmune disease resulting from aberrant immune response mediated by circulating autoantibodies at the neuromuscular junction. Intravenous immunoglobulin (IVIg) is an expensive and commonly used immunotherapeutic approach to treat patients with MG. The mechanisms of actions involved in IVIg treatment, however, remain to be investigated. In an effort to examine the roles of various subsets of CD4+ T cells in the periphery blood of MG and uncover the mechanisms that contribute to the therapeutical effects of IVIg, we first demonstrated that a subset of CD4+ T cells, CTLA-4-expressing regulatory T (Treg) cells, were underrepresented and functionally defective in MG patients. The dynamic profiling during the IVIg therapy course further revealed an inverse relationship between the frequency of CTLA-4+ Treg and the quantitative MG (QMG) score that represents disease severity. Our mechanistic studies indicated that IVIg expands CTLA-4-Treg cells via modulating antigen-presenting dendritic cells (DCs). To determine the molecular defects of CTLA-4 in abnormities of Treg in MG patients, we demonstrated hypermethylation at -658 and -793 CpGs of CTLA-4 promoter in MG Tregs. Interestingly, IVIg therapy significantly reduced the methylation level at these two sites in MG patients. Overall, our study may suggest a role of CTLA-4 in functionally defected Treg cells in MG and its actions involved in IVIg therapy.
Collapse
|
2
|
Chen W, Meng QF, Sui JK, Wang YJ, Li XL, Liu S, Li H, Wang CC, Li CH, Li YB. Ginsenoside Rb1: The new treatment measure of myasthenia gravis. Int Immunopharmacol 2016; 41:136-143. [DOI: 10.1016/j.intimp.2016.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/06/2016] [Accepted: 08/24/2016] [Indexed: 02/08/2023]
|
3
|
Astilbin ameliorates experimental autoimmune myasthenia gravis by decreased Th17 cytokines and up-regulated T regulatory cells. J Neuroimmunol 2016; 298:138-45. [PMID: 27609287 DOI: 10.1016/j.jneuroim.2016.07.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/16/2016] [Accepted: 07/19/2016] [Indexed: 02/07/2023]
Abstract
Astilbin, a major bioactive compound extracted from Rhizoma smilacis glabrae (RSG), has been reported to possess immunosuppressive properties. Our study first evaluated the effect of astilbin on experimental autoimmune myasthenia gravis (EAMG) in Lewis rats. The results showed that astilbin could attenuate the severity of EAMG by decreasing antigen-specific autoantibodies with up-regulation of regulatory T cells and down-regulation of Th17 cells. In addition to, astilbin also reduced the efficiency of the antigen presenting cells on which the expression of MHC class II decreased. These results suggest that astilbin might be a candidate drug for immunoregulation of EAMG, and provide us new treatment ideas for human myasthenia gravis (MG).
Collapse
|
4
|
Losen M, Martinez-Martinez P, Molenaar PC, Lazaridis K, Tzartos S, Brenner T, Duan RS, Luo J, Lindstrom J, Kusner L. Standardization of the experimental autoimmune myasthenia gravis (EAMG) model by immunization of rats with Torpedo californica acetylcholine receptors--Recommendations for methods and experimental designs. Exp Neurol 2015; 270:18-28. [PMID: 25796590 PMCID: PMC4466156 DOI: 10.1016/j.expneurol.2015.03.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/06/2015] [Accepted: 03/10/2015] [Indexed: 12/21/2022]
Abstract
Myasthenia gravis (MG) with antibodies against the acetylcholine receptor (AChR) is characterized by a chronic, fatigable weakness of voluntary muscles. The production of autoantibodies involves the dysregulation of T cells which provide the environment for the development of autoreactive B cells. The symptoms are caused by destruction of the postsynaptic membrane and degradation of the AChR by IgG autoantibodies, predominantly of the G1 and G3 subclasses. Active immunization of animals with AChR from mammalian muscles, AChR from Torpedo or Electrophorus electric organs, and recombinant or synthetic AChR fragments generates a chronic model of MG, termed experimental autoimmune myasthenia gravis (EAMG). This model covers cellular mechanisms involved in the immune response against the AChR, e.g. antigen presentation, T cell-help and regulation, B cell selection and differentiation into plasma cells. Our aim is to define standard operation procedures and recommendations for the rat EAMG model using purified AChR from the Torpedo californica electric organ, in order to facilitate more rapid translation of preclinical proof of concept or efficacy studies into clinical trials and, ultimately, clinical practice.
Collapse
Affiliation(s)
- Mario Losen
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.
| | - Pilar Martinez-Martinez
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Peter C Molenaar
- Division Neuroscience, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | | | - Socrates Tzartos
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, PR China
| | - Jie Luo
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Jon Lindstrom
- Department of Neuroscience, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Linda Kusner
- Department of Pharmacology & Physiology, The George Washington University, Washington, DC, USA
| |
Collapse
|
5
|
Tüzün E, Allman W, Ulusoy C, Yang H, Christadoss P. Novel animal models of acetylcholine receptor antibody-related myasthenia gravis. Ann N Y Acad Sci 2012; 1274:133-9. [DOI: 10.1111/j.1749-6632.2012.06773.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
6
|
Abstract
Etanercept is a tumor necrosis factor (TNF) blocking agent that has been shown to suppress ongoing experimental autoimmune myasthenia gravis (EAMG). This subcutaneously administered TNF blocking agent has also been shown to be safe and effective in several rheumatological diseases including rheumatoid arthritis, psoriasis, and ankylosing spondylitis. A prospective clinical pilot trial of etanercept in corticosteroid-dependent autoimmune myasthenia gravis (MG) patients supports further study into etanercept as a treatment for MG.
Collapse
Affiliation(s)
- Julie Rowin
- University of Illinois at Chicago, Department of Neurology and Rehabilitation, Section of Neuromuscular Disease, Neuropsychiatric Institute, Chicago IL. 60612-7330, USA.
| |
Collapse
|
7
|
Stanovnik B, Bratusek U, Recnik S, Svete J, Meden A. Transformations of Methyl 2-Benzoylamino-2-oxobutanoate. The Synthesis of Oxazolo[4,5-c]quinoline and 1-Substituted 1H-Imidazole-4-carboxylate Derivatives. HETEROCYCLES 2003. [DOI: 10.3987/com-03-9722] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
8
|
Duan RS, Wang HB, Yang JS, Scallon B, Link H, Xiao BG. Anti-TNF-alpha antibodies suppress the development of experimental autoimmune myasthenia gravis. J Autoimmun 2002; 19:169-74. [PMID: 12473237 DOI: 10.1006/jaut.2002.0618] [Citation(s) in RCA: 40] [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
To understand the role of TNF-alpha in the induction of experimental autoimmune myasthenia gravis (EAMG) and detect a possible effect of anti-TNF-alpha antibodies in the treatment of EAMG, anti-TNF-alpha antibodies were administrated intraperitoneally to Lewis rats twice per week for 5 weeks from the day of immunization with Torpedo AChR and complete Freund's adjuvant (CFA). Administration of anti-TNF-alpha antibodies resulted in lower incidence of EAMG, and in delayed onset and only mild muscle weakness compared with control EAMG rats. These mild clinical signs were accompanied by lower AChR-specific lymphocyte proliferation, down-regulated IFN-gamma and IL-10, and up-regulated TGF-beta. The lower levels of anti-AChR IgG, Ig2a and IgG2b and decreased anti-AChR IgG affinity were found in rats treated with anti-TNF-alpha antibodies. These results demonstrate that anti-TNF-alpha antibodies can suppress the induction and development of EAMG.
Collapse
Affiliation(s)
- Rui-Sheng Duan
- Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
9
|
Osterberg J, Haglund U. Effect of linomide on gut immune cell distribution and on TNF-alpha in plasma and ascites: an experimental study in the septic rat. Shock 2002; 18:471-5. [PMID: 12412629 DOI: 10.1097/00024382-200211000-00015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A significant reduction of the pan T lymphocytes as well as CD4+ and CD8 subsets of cells in the gut mucosa of the septic rats has previously been demonstrated. In contrast, the populations of major histocompatibility complex (MHC) class II-positive cells and macrophages increased. The aim of this study was to evaluate if the immunomodulator Linomide influenced the immune cell distribution in the small intestinal mucosa in sepsis and, furthermore, if these changes coincide with changes in the concentration of tumor necrosis factor-alpha (TNF-alpha) in plasma or ascites. Polymicrobial sepsis was induced in rats by cecal ligation and puncture (CLP). Three different experimental groups were used: CLP, Linomide p.o. + CLP, and Linomide i.p.+ CLP, with adequate controls. Specimens were taken from the small bowel for immunohistologic staining and grading of mucosal injury. The following monoclonal antibodies were used: W3/25, OX8, R73, OX6, and ED1. All slides were examined by one "blinded" examiner. Mucosal injury was graded from 0 to 5. The immunostained tissues were also analyzed by an automatic color-based image system. All controls had a normal appearance of the mucosa (grade 0-1), whereas the septic animals had a median grade of III (II-IV) mucosal injury. Linomide i.p. + CLP decreased mucosal damage to median I (0-IV, P < 0.05). Linomide had no effects on the immune cell distribution in controls. In CLP rats, a significant reduction in both CD4+ and CD8+ T lymphocytes as well as an increased number of macrophages and MHC class II-positive cells was seen in the villi as compared with sham-operated controls (P < 0.05). Linomide attenuated these changes for CD8+ and T lymphocytes and macrophages. Sepsis caused increased concentrations of TNF-alpha in portal blood and ascites 3 h from CLP induction. This increase was attenuated by Linomide.
Collapse
|
10
|
Weiss L, Barak V, Zeira M, Abdul-Hai A, Raibstein I, Reich S, Hirschfeld E, Gross D, Slavin S. Cytokine production in Linomide-treated nod mice and the potential role of a Th (1)/Th(2) shift on autoimmune and anti-inflammatory processes. Cytokine 2002; 19:85-93. [PMID: 12182843 DOI: 10.1006/cyto.2002.1956] [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: 11/22/2022]
Abstract
Linomide prevents the development of autoimmune insulitis and insulin-deficient diabetes mellitus in female NOD mice. Linomide prevents development of autoimmune manifestations in other experimentally induced and spontaneous autoimmune diseases as well, but the mechanism of action is unknown. The present report summarizes our investigations on the effect of Linomide on different functional T cell subsets in NOD mice analyzed according to their cytokine profile. Supernatants from cultured splenocytes and peritoneal cells taken from Linomide-treated mice contained lower levels of TNFalpha, IL-1 beta, IFN gamma and IL-12 versus higher levels of IL-4, IL-6 and IL-10 in comparison with supernatants from cultures of untreated mice. Our results suggest that regulation of autoimmunity following oral Linomide administration in NOD mice induces a shift from Th(1) to Th(2) phenotype response, thereby preventing the development of diabetes by active cytokine-induced immunoregulation of T cell subsets, including downregulation of Th(1) and upregulation of Th(2).
Collapse
Affiliation(s)
- Lola Weiss
- Departments of Bone Marrow Transplantation & Cancer Immunotherapy, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zou LP, Abbas N, Volkmann I, Nennesmo I, Levi M, Wahren B, Winblad B, Hedlund G, Zhu J. Suppression of experimental autoimmune neuritis by ABR-215062 is associated with altered Th1/Th2 balance and inhibited migration of inflammatory cells into the peripheral nerve tissue. Neuropharmacology 2002; 42:731-9. [PMID: 11985832 DOI: 10.1016/s0028-3908(02)00015-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The therapeutic effects of ABR-215062, which is a new immunoregulator derived from Linomide, have been evaluated in experimental autoimmune neuritis (EAN), a CD4(+) T cell-mediated animal model of Guillain-Barré syndrome in man. In previous studies, we reported that Linomide suppressed the clinical EAN and myelin antigen-reactive T and B cell responses. Here EAN induced in Lewis rats by inoculation with peripheral nerve myelin P0 protein peptide 180-199 and Freund's complete adjuvant was strongly suppressed by ABR-215062 administered daily subcutaneously from the day of inoculation. ABR-215062 dose-dependently reduced the incidence of EAN, ameliorated clinical signs and inhibited P0 peptide 180-199-specific T cell responses as well as also the decreased inflammation and demyelination in the peripheral nerves. The suppression of clinical EAN was associated with inhibition of the inflammatory cytokines IFN-gamma and TNF-alpha, as well as the enhancement of anti-inflammatory cytokine IL-4 in lymph node cells and periphery nerve tissues, respectively, in a dose-dependent manner. These effects indicate that ABR-215062 may mediate its effects by regulation of Th1/Th2 cytokine balance and suggest that ABR-215062 is potentially a new chemical entity for effective treatment of autoimmune diseases.
Collapse
MESH Headings
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Cell Movement/drug effects
- Cell Movement/immunology
- Dose-Response Relationship, Drug
- Dose-Response Relationship, Immunologic
- Hydroxyquinolines/chemistry
- Hydroxyquinolines/pharmacology
- Hydroxyquinolines/therapeutic use
- Injections, Subcutaneous
- Interferon-gamma/biosynthesis
- Male
- Neuritis, Autoimmune, Experimental/immunology
- Neuritis, Autoimmune, Experimental/pathology
- Neuritis, Autoimmune, Experimental/prevention & control
- Peripheral Nerves/drug effects
- Peripheral Nerves/immunology
- Peripheral Nerves/metabolism
- Peripheral Nerves/pathology
- Quinolones
- Rats
- Rats, Inbred Lew
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/drug effects
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Tumor Necrosis Factor-alpha/biosynthesis
Collapse
Affiliation(s)
- L-P Zou
- Division of Geriatric Medicine, Department of Clinical Neuroscience, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Gross DJ, Weiss L, Reibstein I, Hedlund G, Dahlén E, Rapoport MJ, Slavin S. The immunomodulator Linomide: role in treatment and prevention of autoimmune diabetes mellitus. Int Immunopharmacol 2001; 1:1131-9. [PMID: 11407307 DOI: 10.1016/s1567-5769(01)00042-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Insulin-dependent diabetes mellitus (IDDM) is considered to be an autoimmune disorder characterized by destruction of the pancreatic beta-cells by auto-reacting lymphocytes. An attractive therapeutic approach to this disease would be to abrogate the autoimmune process at an early stage, thus preserving a critical mass of pancreatic beta-cells necessary for maintenance of normal glucose tolerance. Linomide (quinoline-3-carboxamide, Roquinimex, LS 2616), is a novel, orally absorbed, immunomodulatory drug that has been shown to be effective in various models of autoimmunity without causing non-specific immunosuppression. In this review, we describe the efficacy of Linomide for ameliorating the autoimmune process and diabetes in the non-obese diabetic (NOD) model of IDDM when administered at early stages of the disease. We also show that advanced disease in the NOD mouse can be treated effectively by combining Linomide with therapeutic modalities designed to increase pancreatic beta-cell mass. Subsequent clinical studies have shown that Linomide preserves beta-cell function in individuals with new-onset IDDM. Based on these data, Linomide or derivatives thereof might be useful for treatment of human IDDM.
Collapse
Affiliation(s)
- D J Gross
- Department of Endocrinology and Metabolism, Hadassah University Hospital, P.O. Box 12000, 91120, Jerusalem, Israel.
| | | | | | | | | | | | | |
Collapse
|
13
|
Hedlund G, Link H, Zhu J, Xiao BG. Effects of Linomide on immune cells and cytokines inhibit autoimmune pathologies of the central and peripheral nervous system. Int Immunopharmacol 2001; 1:1123-30. [PMID: 11407306 DOI: 10.1016/s1567-5769(01)00041-8] [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: 11/22/2022]
Abstract
Linomide (roquinimex, LS 2616) is a quinoline-3-carboxamide with pleiotropic immune modulating capacity and it has therapeutic effects in several experimental animal models of autoimmune diseases. Linomide has been evaluated in clinical trials for multiple sclerosis, and was indeed shown to have disease inhibitory effects. However, due to unexpected side effects recorded in patients treated with Linomide, premature termination of clinical trials was required. The basic mechanism(s) of action of Linomide in inducing beneficial effects in autoimmune diseases is still elusive. Some experimental evidence indicates that Linomide influences the regulation of the cytokine profile, resulting in the inhibition of autoimmune and inflammation pathologies. This review focuses on Linomide applied in models for autoimmune and inflammation pathologies of the central and the peripheral nervous system, and summarises its very encouraging disease inhibitory effects and their potential pharmacological basis. The beneficial effects recorded with Linomide in both experimental and clinical trials emphasise the possible value of substances with Linomide-like activity for clinical use in autoimmune and inflammation pathologies in the near future.
Collapse
Affiliation(s)
- G Hedlund
- Active Biotech Research AB, Box 724, S-220 07 Lund, Sweden.
| | | | | | | |
Collapse
|
14
|
Poussin MA, Goluszko E, Hughes TK, Duchicella SI, Christadoss P. Suppression of experimental autoimmune myasthenia gravis in IL-10 gene-disrupted mice is associated with reduced B cells and serum cytotoxicity on mouse cell line expressing AChR. J Neuroimmunol 2000; 111:152-60. [PMID: 11063833 DOI: 10.1016/s0165-5728(00)00385-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To analyze the role of interleukin-10 (IL-10) in experimental autoimmune myasthenia gravis (EAMG) pathogenesis, we induced clinical EAMG in C57BL/6 and IL-10 gene-knockout (KO) mice. IL-10 KO mice had a lower incidence and severity of EAMG, with less muscle acetylcholine receptor (AChR) loss. AChR-immunized IL-10 KO mice showed a significantly higher AChR-specific proliferative response, altered cytokine response, lower number of class II-positive cells and B-cells, but a greater CD5(+)CD19(+) population than C57BL/6 mice. The lower clinical incidence in IL-10 KO could be explained not by a reduction of the quantity, but by a possible difference in the pathogenicity of anti-AChR antibodies.
Collapse
MESH Headings
- Adjuvants, Immunologic/genetics
- Animals
- Antigens, CD19/analysis
- Autoantibodies/blood
- B-Lymphocytes/chemistry
- B-Lymphocytes/cytology
- B-Lymphocytes/immunology
- Blood Proteins/immunology
- CD5 Antigens/analysis
- Cell Division/immunology
- Cell Line
- Cytotoxins/immunology
- Epitopes/immunology
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/immunology
- Immunization
- Immunodominant Epitopes/immunology
- In Vitro Techniques
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-6/biosynthesis
- Interleukin-6/immunology
- Lymphocyte Count
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Skeletal/chemistry
- Muscle, Skeletal/immunology
- Myasthenia Gravis, Autoimmune, Experimental/genetics
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Receptors, Cholinergic/genetics
- Receptors, Cholinergic/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- M A Poussin
- Department of Microbiology and Immunology, University of Texas Medical Branch, 77555-1070, Galveston, TX, USA
| | | | | | | | | |
Collapse
|
15
|
Wang HB, Li H, Shi FD, Chambers BJ, Link H, Ljunggren HG. Tumor necrosis factor receptor-1 is critically involved in the development of experimental autoimmune myasthenia gravis. Int Immunol 2000; 12:1381-8. [PMID: 11007755 DOI: 10.1093/intimm/12.10.1381] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor receptor-1 (TNFR1, CD120a) has been implicated in the pathogenesis of several experimental models of T cell-mediated autoimmune disorders, but its role in antibody-mediated autoimmune diseases has not been addressed. Experimental autoimmune myasthenia gravis (EAMG), an autoantibody-mediated T cell-dependent neuromuscular disorder, represents an animal model for myasthenia gravis in human. To investigate the role of TNFR1 in the pathogenesis of EAMG, TNFR1(-/-) and wild-type mice were immunized with TORPEDO: acetylcholine receptor (AChR) in complete Freund's adjuvant. TNFR1(-/-) mice failed to develop EAMG. Lymphoid cells from TNFR1(-/-) mice produced low amounts of T(h)1 (IFN-gamma, IL-2 and IL-12)-type cytokines, but elevated levels of T(h)2 (IL-4 and IL-10)-type cytokines compared with lymphoid cells of wild-type mice. Accordingly, the levels of anti-AChR IgG2 antibodies were severely reduced and the level of anti-AChR IgG1 antibodies were moderately reduced. Co-injection of recombinant mouse IL-12 with AChR in adjuvant restored T cell responses to AChR and promoted development of EAMG in TNFR1(-/-) mice. These results demonstrate that the TNF/TNFR1 system is required for the development of EAMG. The lack of a functional TNF/TNFR1 system can, at least in part, be substituted by IL-12 at the stage of initial priming with AChR and adjuvant.
Collapse
Affiliation(s)
- H B Wang
- Experimental Neurology Unit, Division of Neurology, Karolinska Institutet, Huddinge University Hospital, 14186 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
16
|
Selective disruption of interleukin 4 autocrine-regulated loop by a tyrosine kinase inhibitor restricts activity of T-helper 2 cells. Blood 2000. [DOI: 10.1182/blood.v95.12.3816] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Interleukin (IL) 4 is a potent immunomodulatory cytokine secreted by T-helper 2 (Th2) cells and Th2 mast cells that promotes the commitment of cells. However, unregulated production and release of IL-4 can exacerbate allergic reactions and increase susceptibility to infectious organisms and viruses. Here, we present evidence that AG-490, a Janus tyrosine kinase (JAK) 2-JAK3 inhibitor, effectively blocked IL-4 gene expression and secretion in the Th2 cell line D10 that was not occurring after anti-CD3 antibody stimulation, whereas AG-490 had no inhibitory effect on production of other Th2 cytokines or cytokines synthesized by the corresponding Th1 cell line clone 29. AG-490 potently inhibited IL-4–mediated proliferation of both D10 and the IL-4–dependent cell line CT.4S. Moreover, AG-490 markedly inhibited IL-4 activation of JAK3 and blocked the downstream activation of signal transducer and activator of transcription 6, as judged by tyrosine phosphorylation, DNA binding, and transcription assays. In contrast, AG-490 did not affect tumor necrosis factor activation of NF-κB at similar concentrations of drug. These data suggest that tyrosine kinase inhibitors that inhibit JAK3 may have previously unrecognized and selective clinical potential as immunotherapeutic drugs to treat Th2-mediated diseases driven by IL-4.
Collapse
|
17
|
Selective disruption of interleukin 4 autocrine-regulated loop by a tyrosine kinase inhibitor restricts activity of T-helper 2 cells. Blood 2000. [DOI: 10.1182/blood.v95.12.3816.012k29_3816_3822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin (IL) 4 is a potent immunomodulatory cytokine secreted by T-helper 2 (Th2) cells and Th2 mast cells that promotes the commitment of cells. However, unregulated production and release of IL-4 can exacerbate allergic reactions and increase susceptibility to infectious organisms and viruses. Here, we present evidence that AG-490, a Janus tyrosine kinase (JAK) 2-JAK3 inhibitor, effectively blocked IL-4 gene expression and secretion in the Th2 cell line D10 that was not occurring after anti-CD3 antibody stimulation, whereas AG-490 had no inhibitory effect on production of other Th2 cytokines or cytokines synthesized by the corresponding Th1 cell line clone 29. AG-490 potently inhibited IL-4–mediated proliferation of both D10 and the IL-4–dependent cell line CT.4S. Moreover, AG-490 markedly inhibited IL-4 activation of JAK3 and blocked the downstream activation of signal transducer and activator of transcription 6, as judged by tyrosine phosphorylation, DNA binding, and transcription assays. In contrast, AG-490 did not affect tumor necrosis factor activation of NF-κB at similar concentrations of drug. These data suggest that tyrosine kinase inhibitors that inhibit JAK3 may have previously unrecognized and selective clinical potential as immunotherapeutic drugs to treat Th2-mediated diseases driven by IL-4.
Collapse
|
18
|
Teleshova N, Matusevicius D, Kivisäkk P, Mustafa M, Pirskanen R, Link H. Altered expression of costimulatory molecules in myasthenia gravis. Muscle Nerve 2000; 23:946-53. [PMID: 10842273 DOI: 10.1002/(sici)1097-4598(200006)23:6<946::aid-mus16>3.0.co;2-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To characterize the involvement of costimulatory pathways in the pathogenesis of myasthenia gravis (MG), a multiparameter flow cytometry assay was adopted to enumerate blood mononuclear cells (MNC) expressing CD28, CD80, CD86, CD40, and CD40L molecules in patients with MG and healthy subjects. Patients with MG had lower percentages of CD8(+)CD28(+) cells, augmented percentages of CD4(+)CD80(+), CD4(+)CD86(+), CD8(+)CD80(+), CD8(+)CD86(+), CD14(+)CD80(+), and CD14(+)CD86(+) cells, and similar levels of cells expressing CD40 and CD40L and of B cells expressing CD80 and CD86 compared to the controls. Patients with early onset of MG (<40 years) had lower percentages of CD3(+)CD86(+), CD4(+)CD86(+), CD8(+)CD86(+) T cells and CD20(+)CD86(+) B cells compared to those with late onset (>40 years). There was a positive correlation between the patients' age and percentages of CD86(+) cells. The data indicate that the CD28/CD80-CD86 costimulatory pathway is involved in MG. The high percentages of CD80 and CD86 positive T cells and monocytes may reflect persistent activation of T and B cells, whereas the low CD28 expression may be the result of chronic exposure to CD80 and CD86. These molecules could be the focus for new and improved immunomodulating therapies of MG.
Collapse
Affiliation(s)
- N Teleshova
- Department of Neurology, Neuroimmunology Unit, Karolinska Institutet, Huddinge University Hospital, S-14186 Huddinge, Sweden.
| | | | | | | | | | | |
Collapse
|
19
|
Wang HB, Shi FD, Li H, van der Meide PH, Ljunggren HG, Link H. Role for interferon-gamma in rat strains with different susceptibility to experimental autoimmune myasthenia gravis. Clin Immunol 2000; 95:156-62. [PMID: 10779409 DOI: 10.1006/clim.2000.4850] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Experimental autoimmune myasthenia gravis (EAMG) is caused by autoantibodies against the nicotinic acetylcholine receptor (AChR) at the neuromuscular postsynaptic membrane and represents an animal model of myasthenia gravis in human. Recent studies highlighted the roles of TH1 cytokines (IFN-gamma, IL-12), rather than TH2 cytokines (IL-4), in the pathogenesis of EAMG by using homozygous (-/-) knockout mice with an EAMG-susceptible genetic background. To further evaluate a role for IFN-gamma, we injected recombinant rat IFN-gamma (rrIFN-gamma) at the time of immunization with AChR in complete Freund's adjuvant to EAMG-susceptible Lewis rats and EAMG-resistant Wistar Furth (WF) rats. RrIFN-gamma enhanced Lewis rat EAMG. The exacerbated muscular weakness was associated with higher levels of anti-AChR IgG and enhanced TNF-alpha responses. Anti-AChR IgG antibody levels were augmented to a similar extent as in Lewis rats, however, the identical immunization and IFN-gamma injection induced only mild and transient EAMG in WF rats due to the default TH3 phenotype development and inherent low TH1 responses. We conclude that IFN-gamma plays a major role in the pathogenesis of EAMG in the Lewis rat, but fails to break disease resistance in the WF rat.
Collapse
Affiliation(s)
- H B Wang
- Experimental Neurology Unit, Huddinge University Hospital, Stockholm, S-141 86, Sweden.
| | | | | | | | | | | |
Collapse
|
20
|
Stegall T, Krolick KA. A monoclonal lewis rat myocyte line that responds to interferon-gamma: responsiveness with the potential to influence subsequent interactions with the immune system. Clin Immunol 2000; 94:125-32. [PMID: 10637097 DOI: 10.1006/clim.1999.4821] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In order to begin asking questions about immunopathology associated with the model of the neuromuscular disease experimental autoimmune myasthenia gravis, a monoclonal myocyte line, LE1, has been prepared from the Lewis rat. The LE1 myocyte clone was selected from among several clones produced based on its ease of maintenance in culture and for the stability of its phenotype, which is very similar to that reported for in vivo muscle and other cultured myocyte lines. Thus, LE1 cells were observed to produce, constitutively, the myocyte-associated neural cell adhesion molecule (CD56), the intracellular adhesion molecule (ICAM-1), and the acetylcholine receptor. LE1 cells were also observed to constitutively secrete relatively low levels of IL-6 and TGF-beta. Moreover, the LE1 cell line may be of use for predicting muscle responses to various immune mediators. For example, the inflammatory cytokine interferon-gamma (IFN-gamma) has been recently reported by others to play a role in experimental myasthenia gravis. Thus, it was of interest that LE1 cells could be activated by IFN-gamma to express increased levels of immunopathologically relevant membrane molecules such as ICAM-1 and Class II major histocompatibility molecules (i.e., RT-1B).
Collapse
Affiliation(s)
- T Stegall
- Department of Microbiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78284-7758, USA
| | | |
Collapse
|
21
|
Stegall T, Krolick KA. Myocytes respond to both interleukin-4 and interferon-gamma: cytokine responsiveness with the potential to influence the severity and course of experimental myasthenia gravis. Clin Immunol 2000; 94:133-9. [PMID: 10637098 DOI: 10.1006/clim.1999.4822] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Messenger RNA that encodes for interleukin-15 (IL-15) has been reported to be constitutively expressed in skeletal muscle, although the protein product is not generally observed. Furthermore, interferon-gamma (IFN-gamma) has been reported to exacerbate symptoms of experimental myasthenia gravis (EAMG). Therefore, since IL-15 is an activator of IFN-gamma-producing cells, the hypothesis that drove the study reported below proposes that muscle is not a passive participant in the development of disease symptoms in EAMG and, in fact, plays a very important active role by producing immunomodulating factors that can influence the eventual immunopathological impact of the immune system on muscle. Tests of this hypothesis, made using a monoclonal skeletal myocyte line from the Lewis rat, have indicated that myocytes produce IL-15 protein following exposure to interleukin-4 (IL-4), an interesting paradox in light of the usual anti-inflammatory role played by IL-4. Furthermore, the level of IL-15 also can be regulated by IFN-gamma itself. Although yet to be confirmed in vivo, IFN-gamma has been shown to be capable of activating cultured myocytes in a variety of ways that could influence the ongoing autoimmune response associated with EAMG.
Collapse
Affiliation(s)
- T Stegall
- Department of Microbiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78284-7758, USA
| | | |
Collapse
|
22
|
Abstract
Messenger RNA that encodes for monocyte chemotactic protein-1 (MCP-1), as well as its protein product, was observed to be constitutively expressed at low levels in a monoclonal Lewis rat skeletal muscle cell line (LE1). Immunohistochemical analyses of sections of skeletal muscle yielded similar results. Since interferon-gamma (IFN-gamma) has been reported to have a likely role in determining the severity of symptoms in the neuromuscular autoimmune disease experimental myasthenia gravis (EAMG), the hypothesis tested and proven true in these studies was that IFN-gamma would up-regulate the production of MCP-1 in LE1 cells. It was also observed that muscle-derived MCP-1 could be up-regulated in vivo in rats receiving a monoclonal anti-acetylcholine receptor antibody (mAb35), a potent inducer of symptoms of EAMG. Therefore, it is concluded that muscle may contribute to disease progression by producing factors that influence activities of the immune system.
Collapse
Affiliation(s)
- S M Reyes-Reyna
- Department of Microbiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78284, USA
| | | |
Collapse
|
23
|
Shalev M, Ko A, Gelderman MP, Fortin E, Reed G, Slavin S, Gery I. Multiformic modulation of endotoxin effects by linomide. Clin Immunol 1999; 93:250-5. [PMID: 10600336 DOI: 10.1006/clim.1999.4800] [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] [Indexed: 11/22/2022]
Abstract
Linomide is a potent immunomodulator that either enhances or suppresses certain immunological processes. Of particular interest is this compound's capacity to inhibit a variety of organ-specific autoimmune diseases. Here, we report on the effects of linomide on several immunological reactions elicited by endotoxin (LPS), both in vivo and in vitro. In rats and mice linomide inhibited the elicitation of endotoxin-induced uveitis (EIU), an acute inflammatory eye disease that develops within 24 h following footpad injection of LPS. Linomide also inhibited the production of TNF-alpha and IL-6 by LPS-stimulated rat and mouse macrophage monolayers. On the other hand, treatment with linomide significantly increased the levels of IL-1beta (mice and less in rats), IL-6 (rats), and TNF-alpha (mice) in serum samples collected 2 h following injection with LPS. The increased production of proinflammatory cytokines in linomide-treated mice was also indicated by the enhanced lethal effect of LPS in these mice. The finding of elevated levels of these cytokines in animals with suppressed EIU is also in line with previous observations of an inverse relationship between EIU severity and levels of TNF-alpha. Data recorded here underscore the unique capacity of linomide to both enhance and suppress the immune system.
Collapse
Affiliation(s)
- M Shalev
- The National Eye Institute, NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Schiefer D, Gottstein C, Diehl V, Engert A. [Anti-angiogenesis: a new approach to tumor therapy?]. MEDIZINISCHE KLINIK (MUNICH, GERMANY : 1983) 1999; 94:570-9. [PMID: 10554516 DOI: 10.1007/bf03044955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
BACKGROUND The overall mortality due to metastatic cancer has not or only minimally been reduced in spite of intensive research and many innovations in the field of conventional antineoplastic therapy in the past decade. In the last years it has become a fact that tumor growth is angiogenesis-dependent. Therefore, inhibitors of angiogenesis are a new class of antineoplastic substances with a novel mechanism of action that might be a powerful complement to conventional cytostatic therapy. SUBSTANCES AND CLINICAL TRIALS: Inhibitors of tumor-angiogenesis which have entered clinical trials, with their results published until December 1998 are discussed here. Most results originate from phase-I or phase-II clinical trials. They are discussed and compared in respect to toxicity and response. Also some substances with high therapeutic potential which are still in preclinical testing are discussed. RESULTS Many of the investigated angiogenesis inhibitors demonstrated anti-tumor effects in phase-I or phase-II clinical trials. The commonest manifestation was stable disease, followed by partial remissions. In a few cases, complete remissions were observed. The toxicities of these substances differ both in type and degree of side effects. CONCLUSION Some antiangiogenic drugs appear to be promising candidates for a clinical use in the therapy of solid tumors. Further conclusions can only be drawn after evaluation of the results of ongoing phase-III clinical trials.
Collapse
Affiliation(s)
- D Schiefer
- Klinik I für Innere Medizin, Universität zu Köln
| | | | | | | |
Collapse
|
25
|
Ismaïli L, Refouvelet B, Robert JF. Synthesis of new pyrazolo[4,3-c]quinolin-3-one derivatives and some oxazolo[4,5-c]quinoline-2,4-diones. J Heterocycl Chem 1999. [DOI: 10.1002/jhet.5570360323] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
Zhang GX, Xiao BG, Bai XF, van der Meide PH, Örn A, Link H. Mice with IFN-γ Receptor Deficiency Are Less Susceptible to Experimental Autoimmune Myasthenia Gravis. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.7.3775] [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
IFN-γ can either adversely or beneficially affect certain experimental autoimmune diseases. To study the role of IFN-γ in the autoantibody-mediated experimental autoimmune myasthenia gravis (EAMG), an animal model of myasthenia gravis in humans, IFN-γR-deficient (IFN-γR−/−) mutant C57BL/6 mice and congenic wild-type mice were immunized with Torpedo acetylcholine receptor (AChR) plus CFA. IFN-γR−/− mice exhibited significantly lower incidence and severity of muscle weakness, lower anti-AChR IgG Ab levels, and lower Ab affinity to AChR compared with wild-type mice. Passive transfer of serum from IFN-γR−/− mice induced less muscular weakness compared with serum from wild-type mice. In contrast, numbers of lymph node cells secreting IFN-γ and of those expressing IFN-γ mRNA were strongly augmented in the IFN-γR−/− mice, reflecting a failure of negative feedback circuits. Cytokine studies by in situ hybridization revealed lower levels of lymphoid cells expressing AChR-reactive IL-1β and TNF-α mRNA in AChR + CFA-immunized IFN-γR−/− mice compared with wild-type mice. No differences were found for AChR-reactive cells expressing IL-4, IL-10, or TGF-β mRNA. These results indicate that IFN-γ promotes systemic humoral responses in EAMG by up-regulating the production and the affinity of anti-AChR autoantibodies, thereby contributing to susceptibility to EAMG in C57BL/6-type mice.
Collapse
Affiliation(s)
- Guang-Xian Zhang
- *Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | - Bao-Guo Xiao
- *Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | - Xue-Feng Bai
- *Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | - Anders Örn
- ‡Microbiology and Tumor Biology Center, Karolinska Institute, Stockholm, Sweden
| | - Hans Link
- *Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Hutchings P, Hedlund G, Dawe K, Howlett S, Cooke A. Effect of the synthetic immunomodulator, linomide, on experimental models of thyroiditis. Immunology 1999; 96:340-7. [PMID: 10233714 PMCID: PMC2326756 DOI: 10.1046/j.1365-2567.1999.00713.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The drug Linomide is an immunomodulator showing marked down-regulation of several experimental autoimmune diseases. In this study, its effect on three different experimental models of thyroid disease and on spontaneous infiltration of salivary glands (sialoadenitis), was investigated. Although very effective at preventing thyroid infiltrates in mice immunized with mouse thyroglobulin and complete Freund's adjuvant and in spontaneous models of thyroiditis and sialoadenitis, it completely failed to modify experimental autoimmune thyroiditis (EAT) induced in mice immunized with mouse thyroglobulin and lipopolysaccharide. There was no significant shift in the observed isotypes of anti-mouse thyroglobulin antibodies and only anti-mouse thyroglobulin antibodies in the spontaneous model were completely down-modulated by the drug. One surprising fact to emerge was that Linomide-treated donor mice, although protected from thyroid lesions themselves, were still able to transfer EAT showing that they must have been effectively primed while being treated with Linomide. It is possible that the drug down modulated EAT by interfering with the trafficking of primed effector cells.
Collapse
Affiliation(s)
- P Hutchings
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | | | | |
Collapse
|
28
|
Zhu J, Bai XF, Hedlund G, Björk J, Bakhiet M, Van Der Meide PH, Link H. Linomide suppresses experimental autoimmune neuritis in Lewis rats by inhibiting myelin antigen-reactive T and B cell responses. Clin Exp Immunol 1999; 115:56-63. [PMID: 9933420 PMCID: PMC1905200 DOI: 10.1046/j.1365-2249.1999.00768.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Linomide (quinoline-3-carboxamide) is a synthetic immunomodulator that suppresses several experimental autoimmune diseases. Here we report the effects of Linomide on experimental autoimmune neuritis (EAN), a CD4+ T cell-mediated animal model of acute Guillain-Barré syndrome (GBS) in humans. EAN induced in Lewis rats by inoculation with bovine peripheral nervous system (PNS) myelin and Freund's complete adjuvant was strongly suppressed by Linomide administered daily subcutaneously from the day of inoculation. Linomide dose-dependently delayed the interval between immunization and onset of clinical EAN, as well as the severity of EAN symptoms. These clinical effects were associated with dose-dependent down-modulation of PNS antigen-induced T and B cell responses and with suppression of the proinflammatory cytokines IL-12, interferon-gamma (IFN-gamma) and tumour necrosis factor-alpha (TNF-alpha) mRNA. In PNS sections, Linomide suppressed IL-12 and TNF-alpha, and up-regulated IL-10 mRNA expression. These findings suggest that Linomide could be useful in certain T cell-dependent autoimmune diseases.
Collapse
Affiliation(s)
- J Zhu
- Pharmacia & Upjohn, Lund Research Centre and The Wallenberg Laboratory, Lund University, Sweden
| | | | | | | | | | | | | |
Collapse
|
29
|
Li H, Shi FD, Bai X, Huang Y, Diab A, He B, Link H. Cytokine and chemokine mRNA expressing cells in muscle tissues of experimental autoimmune myasthenia gravis. J Neurol Sci 1998; 161:40-6. [PMID: 9879680 DOI: 10.1016/s0022-510x(98)00181-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
In-situ hybridization with labeled oligonucleotide probes was applied to explore cytokine and chemokine mRNA expression in sections of striated muscle, the target organ in experimental autoimmune myasthenia gravis (EAMG), induced in Lewis rats by immunization with acetylcholine receptor (AChR) and complete Freund's adjuvant (CFA). A transient burst of TNF-alpha, IL-1beta and IL-6 mRNA expressing cells was detected during the early phase of EAMG. This cytokine pattern was related to muscular infiltration of macrophages. Levels of IL-4, IL-10, IFN-gamma, cytolysin and TGF-beta mRNA expressing cells were low and observed mainly during the early phase of EAMG. C-C chemokine RANTES, MCP, MIP-1alpha and MIP-2 mRNA expressing cells were not detected over the course of EAMG. The low and transient expression of cytokines in EAMG muscle tissues suggests that the immune effector responses are unlikely operated by infiltrating cells in muscle. Muscular infiltrations in EAMG are unlikely due to local accumulation of C-C chemokines.
Collapse
Affiliation(s)
- H Li
- Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhu J, Diab A, Mustafa M, Levi M, Wahren B, Björk J, Hedlund G. Linomide suppresses chronic-relapsing experimental autoimmune encephalomyelitis in DA rats. J Neurol Sci 1998; 160:113-20. [PMID: 9849793 DOI: 10.1016/s0022-510x(98)00244-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Linomide (quinoline-3-carboxamide) is a synthetic immunomodulator that suppresses several experimental autoimmune diseases. Here we report the effects of Linomide on chronic progressive and/or relapsing experimental autoimmune encephalomyelitis (PR-EAE), a CD4+ T cell mediated animal model of multiple sclerosis (MS). PR-EAE induced in DA rats by inoculation with homogenized guinea pig spinal cord and Freund's complete adjuvant, was strongly suppressed by Linomide administered daily subcutaneously from the day of inoculation. Linomide dose-dependently delayed the interval between immunization and onset of clinical PR-EAE, reduced severity and relapse of clinical PR-EAE, and shortened clinical PR-EAE. These clinical effects were associated with the down-modulation of CNS antigen-induced T cell responses and production of proinflammatory cytokines (IFN-gamma and TNF-alpha) as well as with upregulation of IL-4 (except in spleen MNC), IL-10 and TGF-beta in both spleen MNC and the spinal cord. These effects indicate that Linomide can suppress PR-EAE and may mediate its suppressive effects by regulation of cytokines.
Collapse
Affiliation(s)
- J Zhu
- Division of Neurology, Karolinska Institute, Huddinge University Hospital, Sweden.
| | | | | | | | | | | | | |
Collapse
|
31
|
Erälinna JP, Röyttä M, Hukkanen V, Zinhu D, Salmi AA, Salonen R. Selective downregulation of Th1 response by Linomide reduces autoimmunity but increases susceptibility to viral infection in BALB/c and SJL mice. J Neuroimmunol 1998; 88:165-76. [PMID: 9688338 DOI: 10.1016/s0165-5728(98)00115-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Susceptibility to autoimmunity has been associated with polarization of Th1/Th2 balance in immune system towards the Th1-type of reactivity. We report here that orally administered quinoline-3-carboxamide (Linomide) selectively downregulates Th1 response in BALB/c and SJL mice, leading to reduction of autoimmunity in the BALB/c and SJL models of experimental allergic encephalomyelitis (EAE). This was shown by prevention of EAE in Th1 responding SJL mice and partial downregulation of EAE in Th2-prone BALB/c mice. In a BALB/c model of EAE, in which infection with Semliki Forest A7 virus (SFV-A7) is used for enhancement of autoimmunity, clinical signs of EAE were reduced while mortality due to viral infection in the CNS was enhanced. Selective downregulation of the Th1 response by Linomide also rendered initially resistant SJL mice susceptible to SFV-A7 CNS infection. This was shown by immunohistochemical detection of extensive deposits of viral antigen in numerous perivascular foci within the CNS and abolished virus antigen-specific lymphocyte reactivity in Linomide-treated SJL mice. In addition, analysis of spleen cell cytokine mRNA production profile revealed decreased number of IFN-gamma producing cells in both SJL and BALB/c mice, reduced number of IL-12p40 producing cells in SJL and increased number of 12p40 producing cells in BALB/c mice along with slightly increased IL-4 production in both strains of mice. These results indicate that oral treatment with Linomide induces selective downregulation of Th1 reactivity causing reduction of autoimmunity and increased susceptibility to SFV-A7 CNS infection. Selective downregulation of Th1 response is a desired effect in the treatment of autoimmune diseases but our results suggest that the benefits have to be balanced against the possible loss in immunoprotection against pathogens.
Collapse
Affiliation(s)
- J P Erälinna
- Department of Virology, University of Turku, Finland.
| | | | | | | | | | | |
Collapse
|
32
|
Xiao BG, Bail XF, Zhang GX, Hedlund G, Link H. Linomide-mediated protection of oligodendrocytes is associated with inhibition of nitric oxide production and IL-1beta expression in Lewis rat glial cells. Neurosci Lett 1998; 249:17-20. [PMID: 9672378 DOI: 10.1016/s0304-3940(98)00371-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Linomide is a synthetic immunomodulator that down-regulates autoimmune response without inducing systemic immunosuppression. Linomide effectively inhibits severe experimental autoimmune diseases, like experimental allergic encephalomyelitis (EAE), a model of multiple sclerosis (MS). Here we report that Linomide suppresses nitric oxide (NO) production by microglia and astrocytes derived from newborn rats and prevented oligodendrocyte damage. Linomide strongly inhibited interleukin (IL) 1 betamRNA expression on glial cells, suggesting a potential mechanism for inhibition of NO production by Linomide. These results demonstrate that Linomide-mediated inhibition of NO production by glial cells could explain the preventive and therapeutic effects of Linomide in EAE and perhaps also MS. However, Linomide at higher dose [correction of doss] (10(-5) M) resulted in direct oligodendrocyte damage.
Collapse
Affiliation(s)
- B G Xiao
- Division of Neurology, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
33
|
Bai XF, Shi FD, Zhu J, Xiao BG, Hedlund G, Link H. Linomide-induced suppression of experimental autoimmune neuritis is associated with down-regulated macrophage functions. J Neuroimmunol 1997; 76:177-84. [PMID: 9184648 DOI: 10.1016/s0165-5728(97)00051-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Experimental autoimmune neuritis (EAN) is a T-cell mediated autoimmune disease of the peripheral nervous system, in which macrophages and T-cells feature prominently in nerve lesions. EAN represents a counterpart to Guillain-Barré syndrome in humans. In the present study, we investigated the in vitro and in vivo effects of Linomide (LS-2616, quinoline-3-carboxamide), a synthetic immunomodulatory compound, on macrophages in relation to EAN. Linomide strongly suppressed IFN-gamma and lipopolysaccharide (LPS)-induced IL-1 beta, TNF-alpha and IL-6 mRNA expression in macrophages in vitro as demonstrated by in situ hybridisation. Linomide administered daily subcutaneously from the day of inoculation completely prevented the development of clinical symptoms of EAN. Linomide administered from day 9 post immunisation (p.i.) significantly suppressed clinical EAN. Macrophages from Linomide-treated EAN rats showed decreased IL-1 beta, TNF-alpha and IL-6 mRNA expression in response to IFN-gamma and LPS. LPS-induced nitric oxide production by macrophages was also suppressed by Linomide in vitro. Linomide, however, does not affect macrophage death and release of lactate dehydrogenase. We conclude that Linomide may exert its actions in EAN and perhaps also in other autoimmune disease models, by suppressing macrophage functions.
Collapse
Affiliation(s)
- X F Bai
- Division of Neurology, Karolinska Institute, Huddinge Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|