151
|
Nandakumar KS, Holmdahl R. Arthritis induced with cartilage-specific antibodiesis IL-4-dependent. Eur J Immunol 2006; 36:1608-18. [PMID: 16688680 DOI: 10.1002/eji.200535633] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
It is widely believed that IL-4 exerts its influence by profiling the immune response during priming and expansion of immune cells, and thereby modulates the outcome of chronic inflammation. In the present investigation, collagen antibody-induced arthritis (CAIA) was used to delineate the role of IL-4 in a T cell-independent inflammatory phase. Mice predisposed to Th2 cytokines (BALB/c and STAT4-deficient mice) developed a more severe arthritis than mice biased towards Th1 cytokines (C57BL/6 and STAT6-deficient mice). Reduced incidence of CAIA was observed in IL-4-deficient mice compared to control littermates. Infiltrating cells in the paws of IL-4-sufficient mice had increased osteoclast activity and tumor necrosis factor (TNF)-alpha and interleukin (IL)-1beta secretion. Massive infiltration of granulocytes and joint and cartilage damage were present in arthritic paws. Depletion of IL-4 suppressed CAIA, which was abrogated by IFN-gamma neutralization. IL-1R- and IL-1RTNFR-deficient mice were completely resistant to CAIA. Thus, IL-4 promotes an antibody-mediated and TNF-alpha/IL-1beta-dependent inflammation in vivo.
Collapse
|
152
|
Polman C, Schellekens H, Killestein J. Neutralizing antibodies to interferon-beta may persist after cessation of therapy: what impact could they have? Mult Scler 2006; 12:245-6. [PMID: 16764335 DOI: 10.1191/135248506ms1284ed] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
153
|
Sharif MN, Sosic D, Rothlin CV, Kelly E, Lemke G, Olson EN, Ivashkiv LB. Twist mediates suppression of inflammation by type I IFNs and Axl. ACTA ACUST UNITED AC 2006; 203:1891-901. [PMID: 16831897 PMCID: PMC2118370 DOI: 10.1084/jem.20051725] [Citation(s) in RCA: 193] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Type I interferons (IFNs) are pleiotropic cytokines with antiviral and immunomodulatory properties. The immunosuppressive actions of type I IFNs are poorly understood, but IFN-mediated suppression of TNFα production has been implicated in the regulation of inflammation and contributes to the effectiveness of type I IFNs in the treatment of certain autoimmune and inflammatory diseases. In this study, we investigated mechanisms by which type I IFNs suppress induction of TNFα production by immune complexes, Fc receptors, and Toll-like receptors. Suppression of TNFα production was mediated by induction and activation of the Axl receptor tyrosine kinase and downstream induction of Twist transcriptional repressors that bind to E box elements in the TNF promoter and suppress NF-κB–dependent transcription. Twist expression was activated by the Axl ligand Gas6 and by protein S and apoptotic cells. These results implicate Twist proteins in regulation of TNFα production by antiinflammatory factors and pathways, and provide a mechanism by which type I IFNs and Axl receptors suppress inflammatory cytokine production.
Collapse
Affiliation(s)
- M Nusrat Sharif
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
154
|
Yamamoto K, Kawamura I, Ito J, Mitsuyama M. Modification of allergic inflammation in murine model of rhinitis by different bacterial ligands: involvement of mast cells and dendritic cells. Clin Exp Allergy 2006; 36:760-9. [PMID: 16776677 DOI: 10.1111/j.1365-2222.2006.02488.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND It has been suggested that airway bacterial infections exacerbate allergic disorders, and bacterial components in the air affect allergic inflammation via Toll-like receptors expressed on mast cells and dendritic cells in the airway mucosa. OBJECTIVE Peptidoglycan (PGN) is a major component of the bacterial cell wall. We investigated the effect of PGN on the effector phase of allergic inflammation, in comparison with the effect of CpG-oligodeoxynucleotides (CpG), which is known to be a Th1 adjuvant. METHODS Ovalbumin (OVA)-sensitized mice were challenged intranasally with OVA alone or OVA together with PGN or CpG. Nasal allergic symptoms and eosinophilia were scored, and the OVA-specific cytokine response was examined in the cells of cervical lymph nodes and nasal mucosa. Bone marrow-derived mast cells (BMMCs) and dendritic cells (BMDCs) were stimulated with PGN or CpG in vitro, and the expression level of cytokines and chemokines was examined by RT-PCR. In addition, the expression level of chemokines was examined by RT-PCR in mast cells of OVA-sensitized mice challenged with OVA alone or OVA together with PGN or CpG. RESULTS PGN exposure exacerbated the nasal allergic symptoms and eosinophilia, whereas CpG exposure suppressed them. In addition, PGN exposure increased the OVA-specific IL-4 response in the cells, whereas CpG exposure decreased it. On the other hand, there were no significant differences in the OVA-specific IFN-gamma response. PGN but not CpG induced the expression of thymus and activation-regulated chemokine (TARC) and macrophage/monocyte-derived chemokine (MDC) in both BMMCs and mast cells of mice sensitized and challenged with OVA. CpG but not PGN induced the expression of IFN-beta and interferon-inducible protein-10 (IP-10) in BMDCs, and histamine did not influence this effect. CONCLUSION These results demonstrate that PGN exposure exacerbates allergic inflammation mainly via mast cells, whereas CpG exposure suppresses allergic inflammation mainly via dendritic cells.
Collapse
Affiliation(s)
- K Yamamoto
- Department of Microbiology, Head and Neck Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | | | | |
Collapse
|
155
|
Liu Y, Teige I, Birnir B, Issazadeh-Navikas S. Neuron-mediated generation of regulatory T cells from encephalitogenic T cells suppresses EAE. Nat Med 2006; 12:518-25. [PMID: 16633347 DOI: 10.1038/nm1402] [Citation(s) in RCA: 238] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Accepted: 04/04/2006] [Indexed: 12/31/2022]
Abstract
Neurons have been neglected as cells with a major immune-regulatory function because they do not express major histocompatibility complex class II. Our data show that neurons are highly immune regulatory, having a crucial role in governing T-cell response and central nervous system (CNS) inflammation. Neurons induce the proliferation of activated CD4+ T cells through B7-CD28 and transforming growth factor (TGF)-beta1-TGF-beta receptor signaling pathways, resulting in amplification of T-cell receptor signaling through phosphorylated ZAP-70, interleukin (IL)-2 and IL-9. The interaction between neurons and T cells results in the conversion of encephalitogenic T cells to CD25+ TGF-beta1+ CTLA-4+ FoxP3+ T regulatory (Treg) cells that suppress encephalitogenic T cells and inhibit experimental autoimmune encephalomyelitis. Suppression is dependent on cytotoxic T lymphocyte antigen (CTLA)-4 but not TGF-beta1. Autocrine action of TGF-beta1, however, is important for the proliferative arrest of Treg cells. Blocking the B7 and TGF-beta pathways prevents the CNS-specific generation of Treg cells. These findings show that generation of neuron-dependent Treg cells in the CNS is instrumental in regulating CNS inflammation.
Collapse
MESH Headings
- Animals
- Antigens, CD
- Antigens, Differentiation/immunology
- B7-1 Antigen/immunology
- CTLA-4 Antigen
- Cell Line
- Cell Proliferation
- Coculture Techniques
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Mice
- Mice, Inbred Strains
- Mice, Transgenic
- Neurons/cytology
- Neurons/physiology
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction/physiology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta1
Collapse
Affiliation(s)
- Yawei Liu
- Neuroinflammation Unit, Institute for Experimental Medical Science, University of Lund, BMC, I13, 221 84 Lund, Sweden
| | | | | | | |
Collapse
|
156
|
Qin H, Wilson CA, Lee SJ, Benveniste EN. IFN‐β‐induced SOCS‐1 negatively regulates CD40 gene expression in macrophages and microglia. FASEB J 2006; 20:985-7. [PMID: 16571771 DOI: 10.1096/fj.05-5493fje] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Costimulation between T cells and antigen-presenting cells is required for adaptive immune responses. CD40, a costimulatory molecule, is expressed in macrophages and microglia. The aberrant expression of CD40 is involved in human diseases including multiple sclerosis, rheumatoid arthritis, and Alzheimer's disease. CD40 expression is induced by a variety of stimuli, including IFN-gamma and lipopolysaccharide (LPS). In this study, we describe the molecular basis by which IFN-beta, a cytokine with immunomodulatory properties, regulates CD40 gene expression. IFN-beta induces CD40 expression in macrophages and microglia at the transcriptional level, and GAS elements in the CD40 promoter are required for IFN-beta-induced CD40 promoter activity. The critical role of signal transducers and activators of transcription-1alpha (STAT-1alpha) in this response was confirmed by utilizing primary microglia from STAT-1alpha deficient mice. IFN-beta induces suppressor of cytokine signaling-1 (SOCS-1) gene expression, which inhibits cytokine signaling by inhibiting activation of STAT proteins. The ectopic expression of SOCS-1 abrogates IFN-beta-mediated STAT-1alpha activation and inhibits IFN-beta-induced CD40 expression. IFN-beta-induced recruitment of STAT-1alpha and RNA Pol II and permissive histone modifications on the CD40 promoter are also inhibited by SOCS-1 overexpression. These novel results indicate that IFN-beta-induced SOCS-1 plays an important role in the negative regulation of IFN-beta-induced CD40 gene expression.
Collapse
Affiliation(s)
- Hongwei Qin
- Department of Cell Biology, University of Alabama at Birmingham, 1918 University Blvd., MCLM 395, Birmingham, Alabama 35294-0005, USA
| | | | | | | |
Collapse
|
157
|
Runström A, Leanderson T, Ohlsson L, Axelsson B. Inhibition of the development of chronic experimental autoimmune encephalomyelitis by laquinimod (ABR-215062) in IFN-beta k.o. and wild type mice. J Neuroimmunol 2006; 173:69-78. [PMID: 16472873 DOI: 10.1016/j.jneuroim.2005.11.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 10/25/2022]
Abstract
Laquinimod is a novel oral immunomodulatory substance, which is currently developed for the treatment of multiple sclerosis (MS). The ability of laquinimod to inhibit disease development was investigated in chronic experimental autoimmune encephalomyelitis (chEAE) in IFN-beta k.o. mice and wild type mice. Laquinimod was shown to inhibit both disease development and histopathological changes in the CNS. Furthermore, laquinimod was found to be independent of endogenous IFN-beta for its effect in chEAE. When laquinimod was combined with exogenous IFN-beta, a synergistic disease inhibitory effect was seen. These findings using laquinimod in preclinical disease models for MS emphasize the potential of laquinimod in the future treatment of MS also in patients that do not respond to IFN-beta monotherapy. Furthermore, the results indicate that laquinimod may favourably be combined with IFN-beta.
Collapse
|
158
|
Treschow AP, Teige I, Nandakumar KS, Holmdahl R, Issazadeh-Navikas S. Stromal cells and osteoclasts are responsible for exacerbated collagen-induced arthritis in interferon-beta-deficient mice. ACTA ACUST UNITED AC 2006; 52:3739-48. [PMID: 16320324 DOI: 10.1002/art.21496] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Clinical trials using interferon-beta (IFNbeta) in the treatment of rheumatoid arthritis have shown conflicting results. We undertook this study to understand the mechanisms of IFNbeta in arthritis at a physiologic level. METHODS Collagen-induced arthritis (CIA) was induced in IFNbeta-deficient and control mice. The role of IFNbeta was investigated in both the priming and effector phases of the disease. The effect of IFNbeta deficiency on synovial cells, macrophages, and fibroblasts from preimmunized mice was analyzed by flow cytometry, immunohistochemistry, and enzyme-linked immunosorbent assay. Differences in osteoclast maturation were determined in situ by histology of arthritic and naive paws and by in vitro maturation studies of naive bone marrow cells. The importance of IFNbeta-producing fibroblasts was determined by transferring fibroblasts into mice at the time of CIA immunization. RESULTS Mice lacking IFNbeta had a prolonged disease with a higher incidence compared with control mice. IFNbeta deficiency was found to influence the effector phase, but not the priming phase, of arthritis. Compared with control mice, IFNbeta-deficient mice had greater infiltration of CD11b+ cells and greater production of tumor necrosis factor alpha in vivo, and their macrophages and fibroblasts were both more activated in vitro. Moreover, IFNbeta-deficient mice generated a greater number of osteoclasts in vitro, and mice immunized to induce arthritis, but not naive mice, had a greater number of osteoclasts in vivo compared with control mice. Importantly, IFNbeta-competent fibroblasts were able to ameliorate arthritis in IFNbeta-deficient recipients. CONCLUSION Our data indicate that IFNbeta is involved in regulating the activation state of osteoclasts and stromal cells, including macrophages and fibroblasts, but that it has little effect on T cells.
Collapse
|
159
|
Galligan CL, Murooka TT, Rahbar R, Baig E, Majchrzak-Kita B, Fish EN. Interferons and viruses: signaling for supremacy. Immunol Res 2006; 35:27-40. [PMID: 17003507 PMCID: PMC7091094 DOI: 10.1385/ir:35:1:27] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/28/2022]
Abstract
Interferon (IFN)-alpha and IFN-beta are critical mediators of host defense against microbial challenges, directly interfering with viral infection and influencing both the innate and adaptive immune responses. IFNs exert their effects in target cells through the activation of a cell-surface receptor, leading to a cascade of signaling events that determine transcriptional and translation regulation. Understanding the circuitry associated with IFN-mediated signal transduction that leads to a specific biological outcome has been a major focus of our laboratory. Through the efforts of graduate students, postdoctoral fellows, a skilled research technologist, and important collaborations with investigators elsewhere, we have provided some insights into the complexity of the IFN system-and the elegance and simplicity of how protein-protein interactions define biological function.
Collapse
Affiliation(s)
- C. L. Galligan
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| | - T. T. Murooka
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| | - R. Rahbar
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| | - E. Baig
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| | - B. Majchrzak-Kita
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| | - E. N. Fish
- Department of Immunology, University of Toronto, Toronto, Canada
- Toronto General Research Institute, University Health Network, 67 College Street Rm. 424, M5G 2M1 Toronto, Ontario Canada
| |
Collapse
|
160
|
Schwarting A, Paul K, Tschirner S, Menke J, Hansen T, Brenner W, Kelley VR, Relle M, Galle PR. Interferon-β: A Therapeutic for Autoimmune Lupus in MRL-Faslpr Mice. J Am Soc Nephrol 2005; 16:3264-72. [PMID: 16221871 DOI: 10.1681/asn.2004111014] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Type I interferons are associated with lupus. Genes that are regulated by IFN-alpha are upregulated in pediatric lupus patients. Gene deletion of the IFN-alpha/beta receptor in experimental lupus-like NZB mice results in reduced disease activity. Conversely, IFN-beta is a well-established treatment in multiple sclerosis, another autoimmune disease. For determining whether IFN-beta treatment is harmful or beneficial in lupus, MRL-Fas(lpr) mice were injected with this type I IFN. Treatment was initiated in MRL-Fas(lpr) mice with mild and advanced disease. IFN-beta was highly effective in prolonging survival and ameliorating the clinical (renal function, proteinuria, splenomegaly, and skin lesions), serologic (autoantibodies and cytokines), and histologic parameters of the lupus-like disease in mice that had mild and advanced disease. Several underlying mechanisms of IFN-beta therapy involving cellular (decreased T cell proliferation and infiltration of leukocytes into the kidney) and humoral (decrease in IgG3 isotypes) immune responses and a reduction in nephrogenic cytokines were identified. In conclusion, IFN-beta treatment of lupus nephritis in MRL-Fas(lpr) mice is remarkably beneficial and suggests that IFN-beta may be an appealing therapeutic candidate for subtypes of human lupus.
Collapse
Affiliation(s)
- Andreas Schwarting
- First Department of Medicine, Johannes-Gutenberg University of Mainz, Langenbeckstrasse 1, Mainz 55131, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Katakura K, Lee J, Rachmilewitz D, Li G, Eckmann L, Raz E. Toll-like receptor 9-induced type I IFN protects mice from experimental colitis. J Clin Invest 2005; 115:695-702. [PMID: 15765149 PMCID: PMC1051992 DOI: 10.1172/jci22996] [Citation(s) in RCA: 361] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Accepted: 12/07/2004] [Indexed: 12/22/2022] Open
Abstract
Experimental colitis is mediated by inflammatory or dysregulated immune responses to microbial factors of the gastrointestinal tract. In this study we observed that administration of Toll-like receptor 9 (TLR9) agonists suppressed the severity of experimental colitis in RAG1-/- but not in SCID mice. This differential responsiveness between phenotypically similar but genetically distinct animals was related to a partial blockade in TLR9 signaling and defective production of type I IFN (i.e., IFN-alpha/beta) in SCID mice upon TLR9 stimulation. The addition of neutralization antibodies against type I IFN abolished the antiinflammatory effects induced by TLR9 agonists, whereas the administration of recombinant IFN-beta mimicked the antiinflammatory effects induced by TLR9 agonists in this model. Furthermore, mice deficient in the IFN-alpha/beta receptor exhibited more severe colitis than wild-type mice did upon induction of experimental colitis. These results indicate that TLR9-triggered type I IFN has antiinflammatory functions in colitis. They also underscore the important protective role of type I IFN in intestinal homeostasis and suggest that strategies to modulate innate immunity may be of therapeutic value for the treatment of intestinal inflammatory conditions.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Cells, Cultured
- Colitis/chemically induced
- Colitis/immunology
- Colon/cytology
- Colon/immunology
- Colon/pathology
- Culture Media, Conditioned
- Cytokines/immunology
- DNA-Activated Protein Kinase
- DNA-Binding Proteins/agonists
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dextran Sulfate/administration & dosage
- Dextran Sulfate/toxicity
- Disease Models, Animal
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Indicators and Reagents/administration & dosage
- Indicators and Reagents/toxicity
- Interferon-alpha/immunology
- Interferon-beta/immunology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Mice, SCID
- Myeloid Differentiation Factor 88
- Nuclear Proteins
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptor, Interferon alpha-beta
- Receptors, Cell Surface/agonists
- Receptors, Cell Surface/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- Toll-Like Receptor 9
Collapse
Affiliation(s)
- Kyoko Katakura
- Department of Medicine, UCSD, La Jolla, California 92093-0663, USA
| | | | | | | | | | | |
Collapse
|
162
|
Hahm B, Trifilo MJ, Zuniga EI, Oldstone MBA. Viruses evade the immune system through type I interferon-mediated STAT2-dependent, but STAT1-independent, signaling. Immunity 2005; 22:247-57. [PMID: 15723812 DOI: 10.1016/j.immuni.2005.01.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 12/10/2004] [Accepted: 01/19/2005] [Indexed: 11/26/2022]
Abstract
Understanding, treating, and preventing diseases caused by immunosuppression and/or persistent infections remain both a major challenge in biomedical research and an important health goal. For a virus or any infectious agent to persist, it must utilize strategies to suppress or evade the host's immune response. Here, we report that two dissimilar viruses employ a common maneuver to cause a profound immunosuppression. Measles virus (MV) and lymphocytic choriomeningitis virus (LCMV) interfere with dendritic cell (DC) development and expansion in vivo and in vitro. The underlying mechanism for this is through the generation of type I interferon (IFN) that acts via a signal transducer and activator of a transcription (STAT)2-dependent, but STAT1-independent, pathway. Thus, viruses subvert the known antiviral effect of type I IFN through STAT2-specific signaling to benefit their survival. These observations have implications for understanding and developing therapies to treat diseases caused by immunosuppression and/or persistent infections.
Collapse
Affiliation(s)
- Bumsuk Hahm
- Division of Virology, Department of Neuropharmacology, Department of Infectology , The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | |
Collapse
|
163
|
Abstract
The significance of type I interferons (IFN-alpha/beta) in biology and medicine renders research on their activities continuously relevant to our understanding of normal and abnormal (auto) immune responses. This relevance is bolstered by discoveries that unambiguously establish IFN-alpha/beta, among the multitude of cytokines, as dominant in defining qualitative and quantitative characteristics of innate and adaptive immune processes. Recent advances elucidating the biology of these key cytokines include better definition of their complex signaling pathways, determination of their importance in modifying the effects of other cytokines, the role of Toll-like receptors in their induction, their major cellular producers, and their broad and diverse impact on both cellular and humoral immune responses. Consequently, the role of IFN-alpha/beta in the pathogenesis of autoimmunity remains at the forefront of scientific inquiry and has begun to illuminate the mechanisms by which these molecules promote or inhibit systemic and organ-specific autoimmune diseases.
Collapse
|
164
|
Matheu V, Treschow A, Teige I, Navikas V, Issazadeh-Navikas S. Local therapy with CpG motifs in a murine model of allergic airway inflammation in IFN-beta knock-out mice. Respir Res 2005; 6:25. [PMID: 15748290 PMCID: PMC555575 DOI: 10.1186/1465-9921-6-25] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Accepted: 03/05/2005] [Indexed: 12/02/2022] Open
Abstract
Background CpG oligodeoxynucleotides (CpG-ODN) are capable of inducing high amounts of type I IFNs with many immunomodulatory properties. Furthermore, type-I IFNs have been proposed to play a key role in mediating effects of CpG-ODN. The precise role of IFN-β in the immunomodulatory effects of CpG-ODN is not known. Objective Here, we aimed to elucidate the role of IFN-β in the anti-allergic effect of CpG motifs. Methods We assessed the immune response in OVA-primed/OVA-challenged IFN-β knockout (-/-) mice compared to wild type (WT) control, after intranasal and systemic treatment with synthetic CpG motifs. Results Vaccination with CpG-ODN reduced the number of cells in airways of OVA-sensitized WT but not IFN-β-/- mice. Although airway eosinophilia was reduced in both treated groups, they were significantly higher in IFN-β-/- mice. Other inflammatory cells, such as lymphocytes and macrophages were enhanced in airways by CpG treatment in IFN-β-/- mice. The ratio of IFN-γ/IL-4 cytokines in airways was significantly skewed to a Th1 response in WT compared to IFN-β-/- group. In contrast, IL-4 and IgE were reduced with no differences between groups. Ag-specific T-cell proliferation, Th1-cytokines such as IFN-γ, IL-2 and also IL-12 were significantly lower in IFN-β-/- mice. Surprisingly, we discovered that intranasal treatment of mice with CpG-ODN results in mild synovitis particularly in IFN-β-/- mice. Conclusion Our results indicate that induction of Th1 response by therapy with CpG-ODN is only slightly and partially dependent on IFN-β, while IFN-β is not an absolute requirement for suppression of airway eosinophilia and IgE. Furthermore, our finding of mild synovitis is a warning for possible negative effects of CpG-ODN vaccination.
Collapse
Affiliation(s)
- Victor Matheu
- Section of Medical Inflammation Research, Department of Cell & Molecular Biology; Lund University; Sweden
- Fundación Rafael Clavijo de Investigación Biomédica, Tenerife, Spain
| | - Alexandra Treschow
- Section of Medical Inflammation Research, Department of Cell & Molecular Biology; Lund University; Sweden
| | - Ingrid Teige
- Section of Medical Inflammation Research, Department of Cell & Molecular Biology; Lund University; Sweden
| | - Vaidrius Navikas
- Section of Medical Inflammation Research, Department of Cell & Molecular Biology; Lund University; Sweden
| | - Shohreh Issazadeh-Navikas
- Section of Medical Inflammation Research, Department of Cell & Molecular Biology; Lund University; Sweden
| |
Collapse
|
165
|
Raivich G, Banati R. Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. ACTA ACUST UNITED AC 2005; 46:261-81. [PMID: 15571769 DOI: 10.1016/j.brainresrev.2004.06.006] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2004] [Indexed: 12/23/2022]
Abstract
Microglia and macrophages, one a brain-resident, the other a mostly hematogenous cell type, represent two related cell types involved in the brain pathology in multiple sclerosis and its autoimmune animal model, the experimental allergic encephalomyelitis. Together, they perform a variety of different functions: they are the primary sensors of brain pathology, they are rapidly recruited to sites of infection, trauma or autoimmune inflammation in experimental allergic encephalomyelitis and multiple sclerosis and they are competent presenters of antigen and interact with T cells recruited to the inflamed CNS. They also synthesise a variety of molecules, such as cytokines (TNF, interleukins), chemokines, accessory molecules (B7, CD40), complement, cell adhesion glycoproteins (integrins, selectins), reactive oxygen radicals and neurotrophins, that could exert a damaging or a protective effect on adjacent axons, myelin and oligodendrocytes. The current review will give a detailed summary on their cellular response, describe the different classes of molecules expressed and their attribution to the blood derived or brain-resident macrophages and then discuss how these molecules contribute to the neuropathology. Recent advances using chimaeric and genetically modified mice have been particularly telling about the specific, overlapping and nonoverlapping roles of macrophages and microglia in the demyelinating disease. Interestingly, they point to a crucial role of hematogenous macrophages in initiating inflammation and myelin removal, and that of microglia in checking excessive response and in the induction and maintenance of remission.
Collapse
Affiliation(s)
- Gennadij Raivich
- Department of Anatomy, Obstetrics and Gynaecology, Perinatal Brain Repair Centre, University College London, Chenies Mews 86-96 WC1E 6HX London, UK.
| | | |
Collapse
|
166
|
Palucka AK, Blanck JP, Bennett L, Pascual V, Banchereau J. Cross-regulation of TNF and IFN-alpha in autoimmune diseases. Proc Natl Acad Sci U S A 2005; 102:3372-7. [PMID: 15728381 PMCID: PMC552921 DOI: 10.1073/pnas.0408506102] [Citation(s) in RCA: 406] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cytokines, most particularly TNF and type I IFN (IFN-alphabeta), have been long considered essential elements in the development of autoimmunity. Identification of TNF in the pathogenesis of rheumatoid arthritis and TNF antagonist therapy represent successes of immunology. IFN-alphabeta plays a major role in systemic lupus erythematosus (SLE), a prototype autoimmune disease characterized by a break of tolerance to nuclear components. Here, we show that TNF regulates IFN-alpha production in vitro at two levels. First, it inhibits the generation of plasmacytoid dendritic cells (pDCs), a major producer of IFN-alphabeta, from CD34+ hematopoietic progenitors. Second, it inhibits IFN-alpha release by immature pDCs exposed to influenza virus. Neutralization of endogenous TNF sustains IFN-alpha secretion by pDCs. These findings are clinically relevant, as five of five patients with systemic juvenile arthritis treated with TNF antagonists display overexpression of IFN-alpha-regulated genes in their blood leukocytes. These results, therefore, might provide a mechanistic explanation for the development of anti-dsDNA antibodies and lupus-like syndrome in patients undergoing anti-TNF therapy.
Collapse
Affiliation(s)
- A Karolina Palucka
- Baylor Institute for Immunology Research, 3434 Live Oak Street, Dallas, TX 75214, USA
| | | | | | | | | |
Collapse
|
167
|
Dikopoulos N, Bertoletti A, Kröger A, Hauser H, Schirmbeck R, Reimann J. Type I IFN negatively regulates CD8+ T cell responses through IL-10-producing CD4+ T regulatory 1 cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:99-109. [PMID: 15611232 DOI: 10.4049/jimmunol.174.1.99] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Pleiotropic, immunomodulatory effects of type I IFN on T cell responses are emerging. We used vaccine-induced, antiviral CD8(+) T cell responses in IFN-beta (IFN-beta(-/-))- or type I IFN receptor (IFNAR(-/-))-deficient mice to study immunomodulating effects of type I IFN that are not complicated by the interference of a concomitant virus infection. Compared with normal B6 mice, IFNAR(-/-) or IFN-beta(-/-) mice have normal numbers of CD4(+) and CD8(+) T cells, and CD25(+)FoxP3(+) T regulatory (T(R)) cells in liver and spleen. Twice as many CD8(+) T cells specific for different class I-restricted epitopes develop in IFNAR(-/-) or IFN-beta(-/-) mice than in normal animals after peptide- or DNA-based vaccination. IFN-gamma and TNF-alpha production and clonal expansion of specific CD8(+) T cells from normal and knockout mice are similar. CD25(+)FoxP3(+) T(R) cells down-modulate vaccine-primed CD8(+) T cell responses in normal, IFNAR(-/-), or IFN-beta(-/-) mice to a comparable extent. Low IFN-alpha or IFN-beta doses (500-10(3) U/mouse) down-modulate CD8(+) T cells priming in vivo. IFNAR- and IFN-beta-deficient mice generate 2- to 3-fold lower numbers of IL-10-producing CD4(+) T cells after polyclonal or specific stimulation in vitro or in vivo. CD8(+) T cell responses are thus subjected to negative control by both CD25(+)FoxP3(+) T(R) cells and CD4(+)IL-10(+) T(R1) cells, but only development of the latter T(R) cells depends on type I IFN.
Collapse
Affiliation(s)
- Nektarios Dikopoulos
- Department of Medical Microbiology and Immunology, University of Ulm, Helmholtzstrasse 8/1, D-89081 Ulm, Germany.
| | | | | | | | | | | |
Collapse
|
168
|
Luo H, Yu G, Tremblay J, Wu J. EphB6-null mutation results in compromised T cell function. J Clin Invest 2005; 114:1762-73. [PMID: 15599401 PMCID: PMC535066 DOI: 10.1172/jci21846] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Accepted: 10/05/2004] [Indexed: 11/17/2022] Open
Abstract
So far, there is very limited knowledge about the role of Eph kinases, the largest family of receptor tyrosine kinases, in the immune system. Here, using EphB6(-/-) mice, we demonstrated that in vitro and in vivo T cell responses such as lymphokine secretion, proliferation, and the development of delayed-type skin hypersensitivity and experimental autoimmune encephalitis in EphB6(-/-) mice were compromised. On the other hand, humoral immune responses, such as serum levels of different Ig isotypes and IgG response to tetanus toxoid, were normal in these mice. Mechanistically, we showed that EphB6 migrated to the aggregated TCRs and rafts after TCR activation. Further downstream, in the absence of EphB6, ZAP-70 activation, LAT phosphorylation, the association of PLCgamma1 with SLP-76, and p44/42 MAPK activation were diminished. Thus, we have shown that EphB6 is pivotal in T cell function.
Collapse
MESH Headings
- Animals
- Antigens/metabolism
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Biotinylation
- CD28 Antigens/biosynthesis
- CD3 Complex/biosynthesis
- Cell Proliferation
- Cytokines/biosynthesis
- Dose-Response Relationship, Drug
- Exons
- Female
- Flow Cytometry
- Gene Deletion
- Green Fluorescent Proteins/metabolism
- Immunoglobulin Class Switching
- Lectins, C-Type
- Leukocytes/metabolism
- Ligands
- Lymphocytes/metabolism
- Mice
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Models, Genetic
- Mutation
- Polymerase Chain Reaction
- Receptor, EphB6/genetics
- Receptor, EphB6/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Interleukin-2/biosynthesis
- Signal Transduction
- Spleen/metabolism
- T-Lymphocytes/metabolism
- Thymus Gland/metabolism
- Time Factors
Collapse
Affiliation(s)
- Hongyu Luo
- Laboratory of Immunology, Centre de Recherché, Notre Dame Hospital, Centre Hospitalier de l'Université de Montréal, Pavilion DeSève, 1560 Sherbrooke Street East, Montréal, Quebec H2L 4M1, Canada
| | | | | | | |
Collapse
|
169
|
Alt C, Duvefelt K, Franzén B, Yang Y, Engelhardt B. Gene and protein expression profiling of the microvascular compartment in experimental autoimmune encephalomyelitis in C57Bl/6 and SJL mice. Brain Pathol 2005; 15:1-16. [PMID: 15779231 PMCID: PMC8095736 DOI: 10.1111/j.1750-3639.2005.tb00094.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Dysfunction of the blood-brain barrier (BBB) is a hallmark of inflammatory diseases of the central nervous system (CNS) such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). The molecular mechanisms leading to BBB breakdown are not well understood. In order to find molecules involved in this process, we used oligonucleotide microarrays and proteomics to analyze gene and protein expression of the microvascular compartment isolated from brains of C57Bl/6 and SJL/N mice afflicted with EAE and the microvascular compartment isolated from healthy controls. Out of the 6500 known genes and expressed sequence tags (ESTs) studied, expression of 288 genes was found to be changed. Of these genes 128 were altered in the microvascular compartment in both EAE models. Six proteins were identified to be present at altered levels. In addition to the expected increased expression of genes coding for molecules involved in leukocyte recruitment, genes not yet ascribed to EAE pathogenesis were identified. Thus, proteomics and gene array screens of the microvascular compartment are valid approaches, that can be used to define novel candidate molecules involved in EAE pathogenesis at the level of the BBB.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/physiology
- Brain/blood supply
- Cerebrovascular Circulation/genetics
- Electrophoresis, Gel, Two-Dimensional
- Electrophoresis, Polyacrylamide Gel
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Gene Expression
- Gene Expression Profiling
- Image Processing, Computer-Assisted
- Immunohistochemistry
- In Situ Hybridization
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Microcirculation/physiology
- Oligonucleotide Array Sequence Analysis
- Protein Array Analysis
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
- Carsten Alt
- Max‐Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kristina Duvefelt
- Gene and Protein Technology, Molecular Sciences, Local Discovery Research Area CNS & Pain Control, Sweden
- Division of Neurology, Neurotec, Karolinska Institute, Huddinge University Hospital, Stockholm, Sweden (present address)
| | - Bo Franzén
- Gene and Protein Technology, Molecular Sciences, Local Discovery Research Area CNS & Pain Control, Sweden
| | - Yang Yang
- Analytical Development, AstraZeneca R&D Södertälje, Sweden
| | - Britta Engelhardt
- Max‐Planck Institute for Molecular Biomedicine, Münster, Germany
- Theodor‐Kocher Institute, University of Bern, Switzerland
| |
Collapse
|
170
|
Abstract
For a long time, the family of type I interferons (IFN-alpha/beta) has received little attention outside the fields of virology and tumor immunology. In recent years, IFN-alpha/beta regained the interest of immunologists, due to the phenotypic and functional characterization of IFN-alpha/beta-producing cells, the definition of novel immunomodulatory functions and signaling pathways of IFN-alpha/beta, and the observation that IFN-alpha/beta not only exerts antiviral effects but is also relevant for the pathogenesis or control of certain bacterial and protozoan infections. This review summarizes the current knowledge on the production and function of IFN-alpha/beta during non-viral infections in vitro and in vivo.
Collapse
Affiliation(s)
- Christian Bogdan
- Institute of Medical Microbiology and Hygiene, Department of Microbiology and Hygiene, University of Freiburg, Freiburg, Germany.
| | | | | |
Collapse
|
171
|
Hadjilambreva G, Mix E, Rolfs A, Müller J, Strauss U. Neuromodulation by a cytokine: interferon-beta differentially augments neocortical neuronal activity and excitability. J Neurophysiol 2004; 93:843-52. [PMID: 15385586 DOI: 10.1152/jn.01224.2003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The immunomodulatory cytokine interferon-beta (IFN-beta) is used in the treatment of autoimmune diseases such as multiple sclerosis. However, the effect of IFN-beta on neuronal functions is currently unknown. Intracellular recordings were conducted on somatosensory neurons of neocortical layers 2/3 and 5 exposed to IFN-beta. The excitability of neurons was increased by IFN-beta (10-10,000 U/ml) in two kinetically distinct, putatively independent manners. First IFN-beta reversibly influenced the subthreshold membrane response by raising the membrane resistance R(M) 2.5-fold and the membrane time constant tau 1.7-fold dose-dependently. The effect required permanent exposure to IFN-beta and was reduced in magnitude if the extracellular K+ was lowered. However, the membrane response to IFN-beta in the subthreshold range was prevented by ZD7288 (a specific blocker of I(h)) but not by Ni2+, carbachol, or bicuculline, pointing to a dependence on an intact I(h). Second, IFN-beta enhanced the rate of action potential firing. This effect was observed to develop for >1 h when the cell was exposed to IFN-beta for 5 min or >5 min and showed no reversibility (< or =210 min). Current-discharge (F-I) curves revealed a shift (prevented by bicuculline) as well as an increase in slope (prevented by carbachol and Ni2+). Layer specificity was not observed with any of the described effects. In conclusion, IFN-beta influences the neuronal excitability in neocortical pyramidal neurons in vitro, especially under conditions of slightly increased extracellular K+. Our blocker experiments indicate that changes in various ionic conductances with different voltage dependencies cause different IFN-beta influences on sub- and suprathreshold behavior, suggesting a more general intracellular process induced by IFN-beta.
Collapse
Affiliation(s)
- Gergana Hadjilambreva
- Department of Neurology, Neurobiological Laboratory, University of Rostock, Gehlsheimer Strasse 20, 18147 Rostock, Germany
| | | | | | | | | |
Collapse
|
172
|
Ivashkiv LB. Type I interferon modulation of cellular responses to cytokines and infectious pathogens: potential role in SLE pathogenesis. Autoimmunity 2004; 36:473-9. [PMID: 14984024 DOI: 10.1080/08916930310001605882] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Type I interferons (IFNs) are pleiotropic cytokines that have been implicated in the pathogenesis of systemic lupus erythematosus (SLE). A key aspect of type I IFN biology is that previous exposure to type I IFNs alters subsequent cellular responses to extracellular stimuli. Type I IFNs may either prime cells for stronger responses to viruses, bacterial pathogens and cytokines such as IL-6 and IFN-gamma, or may suppress cellular responses to LPS and TNFalpha. Herein, we review type I IFN signal transduction via the Jak-STAT pathway, and mechanisms by which type I IFNs prime or suppress responses to environmental factors. We develop a hypothesis that type I IFN-dependent priming/enhancement of cellular responses to pro-inflammatory cytokines such as IFNgamma and IL-6 contributes to pathogenesis of SLE. In addition, cross-regulation between type I IFNs and TNFalpha and its potential role in SLE pathogenesis is discussed.
Collapse
Affiliation(s)
- Lionel B Ivashkiv
- Department of Medicine, Hospital for Special Surgery, Weill Medical College of Cornell University, 535 East 70th Street, New York, NY 10021, USA.
| |
Collapse
|
173
|
Abstract
Inflammatory diseases affect a significant portion of the population worldwide and have been intensely studied for several decades. The advent of transgenic technology has allowed researchers to study individual gene contributions to the pathogenesis of these diseases. This has been done using standard inflammatory disease models in transgenic animals and by identifying novel models through the spontaneous generation of disease in the transgenic animal. Recent advances have been made in the understanding of rheumatoid arthritis, pulmonary inflammation, multiple sclerosis and inflammatory bowel disease through the use of transgenic animals in models of human inflammatory disease.
Collapse
Affiliation(s)
- Carrie M Brodmerkel
- Incyte Corporation, Building 400, Rte 141 and Henry Clay Road, Wilmington, DE 19880, USA
| | | |
Collapse
|
174
|
Abstract
We propose a model where autoimmunity can be viewed as a dynamic system driven by opposite vectors IFN-alpha/beta and TNF. These cytokines drive differentiation of distinct types of DCs, TNF-DCs, or IFN-DCs, which present different antigens leading to distinct autoimmune responses. When balanced, both cytokines synergize in protective immunity. When one of the cytokines prevails, autoimmunity occurs, Type I interferons (IFN-alpha/beta) playing a major role in systemic lupus erythematosus (SLE) and TNF playing a major role in rheumatoid arthritis. This model complements the Type 1/Type 2 paradigm. Therefore, immunity can be viewed as a dynamic system driven by two sets of opposite vectors: IFN-alpha/beta/TNF and IFN-gamma/IL-4.
Collapse
Affiliation(s)
- Jacques Banchereau
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204 USA
| | | | | |
Collapse
|