101
|
Farooqi N, Gran B, Constantinescu CS. Are current disease-modifying therapeutics in multiple sclerosis justified on the basis of studies in experimental autoimmune encephalomyelitis? J Neurochem 2010; 115:829-44. [DOI: 10.1111/j.1471-4159.2010.06982.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
102
|
Christophi GP, Hudson CA, Gruber R, Christophi CP, Massa PT. Promoter-specific induction of the phosphatase SHP-1 by viral infection and cytokines in CNS glia. J Neurochem 2010; 105:2511-23. [PMID: 18331586 DOI: 10.1111/j.1471-4159.2008.05337.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have previously shown that the protein tyrosine phosphatase SHP-1 is highly expressed in CNS glia and is an important modulator of cytokine signaling. As such, mice genetically lacking SHP-1 display constitutive myelin abnormalities, severe virus-induced demyelinating disease, and defects in innate anti-viral responses in the CNS. In this study, we show the differential distribution of the SHP-1 promoter-specific transcripts and demonstrate that several cytokines significantly induce SHP-1 expression in CNS glia. Consistent with these cytokine effects, infection with a neurotropic virus both in vitro and in vivo up-regulates SHP-1 transcripts and protein in CNS cells. Using CNS glial cultures of gene knockout mice, we show that interferons-beta and interferons-gamma act through STAT-1 and interferon regulatory factor-1 to induce the SHP-1 promoter I transcripts. Conversely, interferons-beta and IL-6 act through STAT-3 to induce SHP-1 promoter II transcripts. This study demonstrates that interferons and other cytokines associated with virus infections in the CNS can significantly induce the expression of SHP-1 through STAT-1/3 activity and provides a better understanding of the molecular mechanisms regulating cytokine-induced expression important for multiple homeostatic functions of SHP-1 in the CNS.
Collapse
Affiliation(s)
- George P Christophi
- Department of Neurology, SUNY Upstate Medical University, Syracuse, New York 13210, USA
| | | | | | | | | |
Collapse
|
103
|
Field R, Campion S, Warren C, Murray C, Cunningham C. Systemic challenge with the TLR3 agonist poly I:C induces amplified IFNalpha/beta and IL-1beta responses in the diseased brain and exacerbates chronic neurodegeneration. Brain Behav Immun 2010; 24:996-1007. [PMID: 20399848 PMCID: PMC3334265 DOI: 10.1016/j.bbi.2010.04.004] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 04/12/2010] [Accepted: 04/12/2010] [Indexed: 11/28/2022] Open
Abstract
The role of inflammation in the progression of neurodegenerative disease remains unclear. We have shown that systemic bacterial insults accelerate disease progression in animals and in patients with Alzheimer's disease. Disease exacerbation is associated with exaggerated CNS inflammatory responses to systemic inflammation mediated by microglia that become 'primed' by the underlying neurodegeneration. The impact of systemic viral insults on existing neurodegenerative disease has not been investigated. Polyinosinic:polycytidylic acid (poly I:C) is a toll-like receptor-3 (TLR3) agonist and induces type I interferons, thus mimicking inflammatory responses to systemic viral infection. In the current study we hypothesized that systemic challenge with poly I:C, during chronic neurodegenerative disease, would amplify CNS inflammation and exacerbate disease. Using the ME7 model of prion disease and systemic challenge with poly I:C (12 mg/kg i.p.) we have shown an amplified expression of IFN-alpha and beta and of the pro-inflammatory genes IL-1beta and IL-6. Similarly amplified expression of specific IFN-dependent genes confirmed that type I IFNs were secreted and active in the brain and this appeared to have anti-inflammatory consequences. However, prion-diseased animals were susceptible to heightened acute sickness behaviour and acute neurological impairments in response to poly I:C and this treatment also accelerated disease progression in diseased animals without effect in normal animals. Increased apoptosis coupled with double-stranded RNA-dependent protein kinase (PKR) and Fas transcription suggested activation of interferon-dependent, pro-apoptotic pathways in the brain of ME7+poly I:C animals. That systemic poly I:C accelerates neurodegeneration has implications for the control of systemic viral infection during chronic neurodegeneration and indicates that type I interferon responses in the brain merit further study.
Collapse
Affiliation(s)
- Robert Field
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Suzanne Campion
- Weatherall Institute of Molecular Medicine, University of Oxford. Oxford, OX3 9DS, UK
| | - Colleen Warren
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Carol Murray
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
104
|
Prinz M, Kalinke U. New lessons about old molecules: how type I interferons shape Th1/Th17-mediated autoimmunity in the CNS. Trends Mol Med 2010; 16:379-86. [PMID: 20591737 DOI: 10.1016/j.molmed.2010.06.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/14/2010] [Accepted: 06/03/2010] [Indexed: 01/02/2023]
Abstract
Type I interferons (IFN-alpha and IFN-beta) were discovered more than five decades ago and are widely used for the treatment of human autoimmune diseases such as multiple sclerosis (MS). Despite their highly beneficial features, the precise mechanism of action remains speculative. Given the frequent side effects of IFN-alpha/beta therapy, understanding its action in an in vivo setting is vital to further improve this therapeutic approach. Major advances in our understanding of the IFN biology have recently been made and are particularly based on the combination of powerful genome-wide expression analysis in humans with gene-targeting techniques available for basic research. The recent discovery of a novel T-cell subset, Th17 cells, sheds new light on type I IFNs in MS.
Collapse
Affiliation(s)
- Marco Prinz
- Department of Neuropathology, University of Freiburg, Breisacher Str. 64, D-79106 Freiburg, Germany.
| | | |
Collapse
|
105
|
Croxford AL, Kurschus FC, Waisman A. Mouse models for multiple sclerosis: historical facts and future implications. Biochim Biophys Acta Mol Basis Dis 2010; 1812:177-83. [PMID: 20600870 DOI: 10.1016/j.bbadis.2010.06.010] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2009] [Revised: 06/04/2010] [Accepted: 06/16/2010] [Indexed: 11/19/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory and demyelinating condition of the CNS, characterized by perivascular infiltrates composed largely of T lymphocytes and macrophages. Although the precise cause remains unknown, numerous avenues of research support the hypothesis that autoimmune mechanisms play a major role in the development of the disease. Pathologically similar lesions to those seen in MS can be induced in laboratory rodents by immunization with CNS-derived antigens. This form of disease induction, broadly termed experimental autoimmune encephalomyelitis, is frequently the starting point in MS research with respect to studying pathogenesis and creating novel treatments. Many different EAE models are available, each mimicking a particular facet of MS. These models all have common ancestry, and have developed from a single concept of immunization with self-antigen. We will discuss the major changes in immunology research, which have shaped the EAE models we use today, and discuss how current animal models of MS have resulted in successful treatments and more open questions for researchers to address.
Collapse
MESH Headings
- Animals
- Autoantigens/history
- Autoantigens/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/history
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Gene Targeting/history
- History, 20th Century
- History, 21st Century
- Humans
- Mice
- Multiple Sclerosis/etiology
- Multiple Sclerosis/immunology
- Multiple Sclerosis/therapy
- Th17 Cells/immunology
Collapse
Affiliation(s)
- Andrew L Croxford
- Institute for Molecualr Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Germany.
| | | | | |
Collapse
|
106
|
Khorooshi R, Owens T. Injury-induced type I IFN signaling regulates inflammatory responses in the central nervous system. THE JOURNAL OF IMMUNOLOGY 2010; 185:1258-64. [PMID: 20562259 DOI: 10.4049/jimmunol.0901753] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Innate glial response is critical for the induction of inflammatory mediators and recruitment of leukocytes to sites of the injury in the CNS. We have examined the involvement of type I IFN signaling in the mouse hippocampus following sterile injury (transection of entorhinal afferents). Type I IFNs signal through a receptor (IFNAR), which involves activation of IFN regulatory factor (IRF)9, leading to the induction of IFN-stimulated genes including IRF7, that in turn enhances the induction of type I IFN. Axonal transection induced upregulation of IRF7 and IRF9 in hippocampus. Induction of IRF7 and IRF9 mRNAs was IFNAR dependent. Double-labeling immunofluorescence showed that IRF7 selectively was induced in Mac-1/CD11b(+) macrophages/microglia in hippocampus after axonal transection. IRF7 mRNA was also detected in microglia sorted by flow cytometry. Lack of type I IFN signaling resulted in increased leukocyte infiltration into the lesion-reactive hippocampus. Axonal lesion-induced CXCL10 gene expression was abrogated, whereas matrix metalloproteinase 9 mRNA was elevated in IFNAR-deficient mice. Our findings point to a role for type I IFN signaling in regulation of CNS response to sterile injury.
Collapse
Affiliation(s)
- Reza Khorooshi
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
107
|
|
108
|
Zhang X, Markovic-Plese S. Interferon beta inhibits the Th17 cell-mediated autoimmune response in patients with relapsing-remitting multiple sclerosis. Clin Neurol Neurosurg 2010; 112:641-5. [PMID: 20570038 DOI: 10.1016/j.clineuro.2010.04.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 04/19/2010] [Accepted: 04/28/2010] [Indexed: 01/15/2023]
Abstract
Interferon (IFN)beta has been used over the past decades as an effective first-line therapy against relapsing-remitting multiple sclerosis (RR MS), however its in vivo operative mechanisms of action are not fully understood. Current advances in our understanding of the development of the autoimmune response, including its induction by a recently discovered Th17 cell lineage, may allow us to identify the biomarkers of this effective therapy. Our in vitro human studies have characterized IFNbeta's immunoregulatory effects on Th17 cell differentiation. IFNbeta inhibited IL-1beta, IL-23 and transforming growth factor (TGF)-beta (which induce Th17 cell differentiation), and induced IL-27, IL-12p35 and IL-10 (which suppress it) in dendritic cells (DCs) and B-cells. The effect on IL-1beta, IL-23 and IL-27 production in DCs was mediated by the up-regulation of Toll-like receptor (TLR)7 and its downstream signaling molecules. IFNbeta's direct effect on naïve T-cells suppressed in vitro Th17 differentiation by inhibiting Th17 cell lineage markers (retinoic acid-related orphan nuclear hormone receptor (ROR)c, IL-17A, IL-23R and CCR6), and by inducing IL-10 production by CD4 cells, which constrains Th17 cell expansion. Our results have identified novel therapeutic mechanisms of IFNbeta, which inhibit Th17 cell differentiation in the context of the autoimmune response in MS.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
109
|
Yen JH, Kong W, Ganea D. IFN-beta inhibits dendritic cell migration through STAT-1-mediated transcriptional suppression of CCR7 and matrix metalloproteinase 9. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:3478-86. [PMID: 20190134 PMCID: PMC2877494 DOI: 10.4049/jimmunol.0902542] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
IFN-beta is an approved therapeutic option for the treatment of multiple sclerosis. The molecular mechanisms underlying the effects of IFN-beta in multiple sclerosis are not fully understood. Migration of dendritic cells (DCs) from the inflammatory site to draining lymph nodes for Ag presentation and activation of naive T cells and to the CNS for reactivation of encephalitogenic T cells requires CCR7 and matrix metalloproteinase (MMP)-9 expression. This article reports for the first time that IFN-beta inhibits CCR7 expression and MMP-9 production in mature DCs and reduces their migratory capacity. The effect of IFN-beta is mediated through STAT-1. In vivo treatment with IFN-beta results in lower numbers of DCs migrating to the draining lymph node following exposure to FITC and in reduced expression of CCR7 and MMP-9 in splenic CD11c(+) DCs following LPS administration. IFN-beta and IFN-gamma share the same properties in terms of their effects on CCR7, MMP-9, and DC migration, but they have opposite effects on IL-12 production. In addition, IFN-beta-treated DCs have a significantly reduced capacity for activating CD4(+) T cells and generating IFN-gamma-producing Th1 cells. The suppression of mature DC migration through negative regulation of CCR7 and MMP-9 expression represents a novel mechanism for the therapeutic effect of IFN-beta.
Collapse
Affiliation(s)
- Jui-Hung Yen
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Weimin Kong
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Doina Ganea
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
110
|
The TLR7 agonist, imiquimod, increases IFN-β production and reduces the severity of experimental autoimmune encephalomyelitis. J Neuroimmunol 2010; 221:107-11. [DOI: 10.1016/j.jneuroim.2010.01.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Revised: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 02/06/2023]
|
111
|
Merson TD, Binder MD, Kilpatrick TJ. Role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination of the CNS. Neuromolecular Med 2010; 12:99-132. [PMID: 20411441 DOI: 10.1007/s12017-010-8112-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2009] [Accepted: 02/26/2010] [Indexed: 12/11/2022]
Abstract
As the resident innate immune cells of the central nervous system (CNS), microglia fulfil a critical role in maintaining tissue homeostasis and in directing and eliciting molecular responses to CNS damage. The human disease Multiple Sclerosis and animal models of inflammatory demyelination are characterized by a complex interplay between degenerative and regenerative processes, many of which are regulated and mediated by microglia. Cellular communication between microglia and other neural and immune cells is controlled to a large extent by the activity of cytokines. Here we review the role of cytokines as mediators and regulators of microglial activity in inflammatory demyelination, highlighting their importance in potentiating cell damage, promoting neuroprotection and enhancing cellular repair in a context-dependent manner.
Collapse
Affiliation(s)
- Tobias D Merson
- Florey Neuroscience Institutes, Centre for Neuroscience, University of Melbourne, Parkville, VIC, 3010, Australia.
| | | | | |
Collapse
|
112
|
Suppression of EAE by oral tolerance is independent of endogenous IFN‐β whereas treatment with recombinant IFN‐β ameliorates EAE. Immunol Cell Biol 2010; 88:468-76. [DOI: 10.1038/icb.2009.111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
113
|
Isaksson M, Ardesjö B, Rönnblom L, Kämpe O, Lassmann H, Eloranta ML, Lobell A. Plasmacytoid DC promote priming of autoimmune Th17 cells and EAE. Eur J Immunol 2009; 39:2925-35. [PMID: 19637225 DOI: 10.1002/eji.200839179] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
EAE, an animal model for MS, is a Th17 and Th1-cell-mediated autoimmune disease, but the mechanisms leading to priming of encephalitogenic T cells in autoimmune neuroinflammation are poorly understood. To investigate the role of plasmacytoid DC (pDC) in the initiation of autoimmune Th17- and Th1-cell responses and EAE, we depleted pDC with anti-pDC Ag-1 (anti-PDCA1) mAb prior to immunization of C57BL/6 mice with myelin oligodendrocyte glycoprotein (MOG). pDC-depleted mice developed less severe clinical and histopathological signs of EAE than control mice, which demonstrates a promoting role for pDC in the initiation of EAE. The levels of type I IFN were much lower in the sera from anti-PDCA1-treated mice. However, neutralization of type I IFN ameliorated the early phase of EAE but did not alter the severity of disease. Thus, only a minor part of the EAE-promoting effect of pDC appears to be mediated by IFN-alpha/beta secretion. The numbers of MOG-specific Th17 cells, but not Th1 cells, were lower in spleen from anti-PDCA1-treated mice compared with controls. In contrast, pDC depletion a week after MOG immunization resulted in more severe clinical signs of EAE. In conclusion, we demonstrate that pDC promote initiation of MOG-induced Th17-cell responses and EAE.
Collapse
Affiliation(s)
- Magnus Isaksson
- Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
114
|
Mackensen F, Max R, Becker MD. Interferons and their potential in the treatment of ocular inflammation. Clin Ophthalmol 2009; 3:559-66. [PMID: 19898628 PMCID: PMC2770867 DOI: 10.2147/opth.s3308] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Indexed: 12/31/2022] Open
Abstract
Since their discovery in the 1950s interferons have been the scope of investigation in many diseases as therapeutic as well as pathogenetic factors. We know they have immune stimulatory and immune regulatory effects. This apparently counter-intuitive mechanism can be summarized as immunomodulatory action and seems to be very effective in a number of ocular inflammatory diseases. We review the current knowledge of interferons in immunity and autoimmunity and show their use in clinical ophthalmologic practice.
Collapse
Affiliation(s)
- Friederike Mackensen
- Interdisciplinary Uveitis Center, Dept. of Ophthalmology, University of Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany.
| | | | | |
Collapse
|
115
|
Koning N, Uitdehaag BMJ, Huitinga I, Hoek RM. Restoring immune suppression in the multiple sclerosis brain. Prog Neurobiol 2009; 89:359-68. [PMID: 19800386 DOI: 10.1016/j.pneurobio.2009.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 08/26/2009] [Accepted: 09/28/2009] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis is a very disabling inflammatory demyelinating disease of the brain of unknown etiology. Current therapies can reduce new lesion development and partially prevent clinical disease activity, but none can halt the progression, or cure the disease. We will review current therapeutic strategies, which are mostly discussed in literature in terms of their effective inhibition of T cells. However, we argue that many of these treatments also influence the myeloid compartment. Interestingly, recent evidence indicates that myelin phagocytosis by infiltrated macrophages and activated microglia is not just a hallmark of multiple sclerosis, but also a key determinant of lesion development and disease progression. We reason that severe side effects and/or insufficient effectiveness of current treatments necessitates the search for novel therapeutic targets, and postulate that these should aim at manipulation of the activation and phagocytic capacity of macrophages and microglia. We will discuss three candidate targets with high potential, namely the complement receptor 3, CD47-SIRPalpha interaction as well as CD200-CD200R interaction. Blocking the actions of complement receptor 3 could inhibit myelin phagocytosis, as well as migration of myeloid cells into the brain. CD47 and CD200 are known to inhibit macrophage/microglia activation through binding to their receptors SIRPalpha and CD200R, expressed on phagocytes. Triggering these receptors may thus dampen the inflammatory response. Our recent findings indicate that the CD200-CD200R interaction is the most specific and hence probably best-suited target to suppress excessive macrophage and microglia activation, and restore immune suppression in the brain of patients with multiple sclerosis.
Collapse
Affiliation(s)
- Nathalie Koning
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
116
|
Matheu V, Barrios Y, Arnau MR, Navikas V, Issazadeh-Navikas S. Similar response in male and female B10.RIII mice in a murine model of allergic airway inflammation. Inflamm Res 2009; 59:263-9. [PMID: 19779803 DOI: 10.1007/s00011-009-0094-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2009] [Revised: 09/07/2009] [Accepted: 09/08/2009] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Several reports have been published on the gender differences associated with allergies in mice. GOAL In the present study we investigate the influence of gender on allergy response using a strain of mice, B10.RIII, which is commonly used in the collagen-induced arthritis murine model. METHODS Both male and female B10.RIII young mice were immunized with OVA and challenged four times with OVA intranasally. Samples were taken 24 h after the last challenge, and eosinophils in bronchoalveolar lavage (BAL) and parenchyma, Th-2 cytokines in BAL, total and antigen-specific IgE in sera, and antigen-specific T-cell proliferation were measured. RESULTS Immunization in both male and female B10.RIII mice with OVA elicited a classical Th2-type response. Results showed no significant differences among male and female mice. Also a high eosinophilia in BAL fluid and parenchyma was produced in both genders without any significant differences. However, the deviation of both parameters was higher in young males compared to young females. CONCLUSIONS Gender differences, classically associated with some strains of mice, are not reproducible in B10.RIII mice. Gender differences in murine models of allergic airway inflammation are probably strain-dependent.
Collapse
Affiliation(s)
- Victor Matheu
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.
| | | | | | | | | |
Collapse
|
117
|
Ramgolam VS, Sha Y, Jin J, Zhang X, Markovic-Plese S. IFN-beta inhibits human Th17 cell differentiation. THE JOURNAL OF IMMUNOLOGY 2009; 183:5418-27. [PMID: 19783688 DOI: 10.4049/jimmunol.0803227] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IFN-beta-1a has been used over the past 15 years as a primary therapy for relapsing-remitting multiple sclerosis (MS). However, the immunomodulatory mechanisms that provide a therapeutic effect against this CNS inflammatory disease are not yet completely elucidated. The effect of IFN-beta-1a on Th17 cells, which play a critical role in the development of the autoimmune response, has not been extensively studied in humans. We have investigated the effect of IFN-beta-1a on dendritic cells (DCs) and naive CD4(+)CD45RA(+) T cells derived from untreated MS patients and healthy controls in the context of Th17 cell differentiation. We report that IFN-beta-1a treatment down-regulated the expression of IL-1beta and IL-23p19 in DCs, whereas it induced the gene expression of IL-12p35 and IL-27p28. We propose that IFN-beta-1a-mediated up-regulation of the suppressor of cytokine signaling 3 expression, induced via STAT3 phosphorylation, mediates IL-1beta and IL-23 down-regulation, while IFN-beta-1a-induced STAT1 phosphorylation induces IL-27p28 expression. CD4(+)CD45RA(+) naive T cells cocultured with supernatants from IFN-beta-1a-treated DCs exhibited decreased gene expression of the Th17 cell markers retinoic acid-related orphan nuclear hormone receptor c (RORc), IL-17A, and IL-23R. A direct IFN-beta-1a treatment of CD45RA(+) T cells cultured in Th17-polarizing conditions also down-regulated RORc, IL-17A, and IL-23R, but up-regulated IL-10 gene expression. Studies of the mechanisms involved in the Th17 cell differentiation suggest that IFN-beta-1a inhibits IL-17 and induces IL-10 secretion via activated STAT1 and STAT3, respectively. IFN-beta's suppression of Th17 cell differentiation may represent its most relevant mechanism of selective suppression of the autoimmune response in MS.
Collapse
Affiliation(s)
- Vinod S Ramgolam
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
118
|
Baranzini SE. Systems-based medicine approaches to understand and treat complex diseases. The example of multiple sclerosis. Autoimmunity 2009; 39:651-62. [PMID: 17178562 DOI: 10.1080/08916930601061686] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Systems medicine is an emerging concept that acknowledges the complexity of a multitude of non-linear interactions among molecular and physiological variables. Under this new paradigm, rather than a collection of symptoms, diseases are seen as the product of deviations from a robust steady state compatible with life. This concept requires the incorporation of mathematics and physics to the more classical arsenal of physiology and molecular biology with which physicians are trained today. This review explores the diverse types of information that can be accumulated towards the understanding of multiple sclerosis (MS), a complex autoimmune disease that targets the central nervous system (CNS). The challenge of data integration and modeling of dynamical systems is discussed in the context of disease susceptibility and response to treatment. A theoretical framework that supports the use of combination therapy is also presented.
Collapse
Affiliation(s)
- Sergio E Baranzini
- Department of Neurology, School of Medicine, University of California, San Francisco, 513 Parnassus Avenue Room S-256, San Francisco, CA 94143-0435, USA.
| |
Collapse
|
119
|
Li M, Liu X, Zhou Y, Su SB. Interferon-lambdas: the modulators of antivirus, antitumor, and immune responses. J Leukoc Biol 2009; 86:23-32. [PMID: 19304895 DOI: 10.1189/jlb.1208761] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
IFN-lambdas, including IFN-lambda1, IFN-lambda2, and IFN-lambda3, also known as IL-29, IL-28A, or IL-28B, are a newly described group of cytokines distantly related to the type I IFNs and IL-10 family members. The IFN-lambdaR complex consists of a unique ligand-binding chain, IFN-lambdaR1 (also designated IL-28Ralpha), and an accessory chain, IL-10R2, which is shared with receptors for IL-10-related cytokines. IFN-lambdas signal through the IFN-lambdaR and activate pathways of JAK-STATs and MAPKs to induce antiviral, antiproliferative, antitumor, and immune responses. In this review, we summarize recent findings about the biology of IFN-lambdas and their pathophysiological roles in viral infection, cancer, and immune responses of the innate and adaptive arms.
Collapse
Affiliation(s)
- Mingcai Li
- Institute of Inflammation and Immune Diseases, Shantou University Medical College, Shantou, China
| | | | | | | |
Collapse
|
120
|
Interferon beta induces mature dendritic cell apoptosis through caspase-11/caspase-3 activation. Blood 2009; 114:1344-54. [PMID: 19531658 DOI: 10.1182/blood-2008-12-196592] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although interferon beta (IFNbeta) decreases relapse rate and disease activity in multiple sclerosis (MS), the mechanisms involved have not been elucidated. The present study is the first report on the apoptotic effect of IFNbeta in mature, but not immature, myeloid dendritic cells (DCs). Both exogenous IFNbeta added to DCs matured through exposure to proinflammatory cytokines and endogenous IFNbeta secreted after lipopolysaccharide stimulation induced DC cell death. Apoptosis of mature DCs required both NF-kappaB and STAT-1 activation, and was mediated through the induction of caspase-11 expression and activation of caspase-3. In vivo, we observed increased caspase-11 expression and a significant decrease in the number of splenic DCs after lipopolysaccharide administration in wt but not in STAT-1-deficient mice. Since mature DCs are major contributors to the inflammatory response and essential partners in the induction of adaptive immunity, IFNbeta-dependent elimination of activated DCs could play an essential role in re-establishing homeostasis, and might represent a new molecular mechanism for the therapeutic effect of IFNbeta in MS.
Collapse
|
121
|
Mannie MD, Abbott DJ, Blanchfield JL. Experimental autoimmune encephalomyelitis in Lewis rats: IFN-beta acts as a tolerogenic adjuvant for induction of neuroantigen-dependent tolerance. THE JOURNAL OF IMMUNOLOGY 2009; 182:5331-41. [PMID: 19380780 DOI: 10.4049/jimmunol.0803756] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytokine-Ag fusion proteins represent a novel approach for induction of Ag-specific tolerance and may constitute an efficient therapy for autoimmune disease. This study addressed whether a fusion protein containing rat IFN-beta and the encephalitogenic 73-87 determinant of myelin basic protein (i.e., the neuroantigen, or NAg) could prevent or treat experimental autoimmune encephalomyelitis (EAE) in Lewis rats. The optimal structure of the fusion protein was comprised of the rat IFN-beta cytokine as the N-terminal domain with an enterokinase (EK) linker to the NAg domain. Both cytokine and NAg domains had full biological activity. Subcutaneous administration of 1 nmol of IFNbeta-NAg fusion protein in saline on days -21, -14, and -7 before encephalitogenic challenge on day 0 resulted in a substantial attenuation of EAE. In contrast, administration of IFN-beta or NAg alone did not affect susceptibility to EAE. The covalent attachment of IFN-beta and NAg was not necessary, because separate injections of IFN-beta and NAg at adjacent sites were as effective as injection of IFNbeta-NAg for prevention of disease. When treatment was initiated after disease onset, the rank order of inhibitory activity was as follows: the IFNbeta-NAg fusion protein > or = a mixture of IFN-beta plus NAg > IFN-beta > NAg. The novel finding that IFN-beta acts as a tolerogenic adjuvant as well as a tolerogenic fusion partner may have significance for development of tolerogenic vaccines.
Collapse
Affiliation(s)
- Mark D Mannie
- Department of Microbiology and Immunology, East Carolina University, Brody School of Medicine, Greenville, NC 27834, USA
| | | | | |
Collapse
|
122
|
Goriely S, Cavoy R, Goldman M. Interleukin-12 family members and type I interferons in Th17-mediated inflammatory disorders. Allergy 2009; 64:702-9. [PMID: 19383026 DOI: 10.1111/j.1398-9995.2009.02039.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cytokines produced by antigen-presenting cells govern the fate of helper T-cell responses. Herein, we review the impact of interleukin (IL)-23 and IL-27 on the outcome of T-helper (Th) 17 cell responses and discuss their impact in the pathogenesis of T-cell-mediated inflammatory disorders of autoimmune or allergic origin. We then discuss how type I interferons might influence the course of autoimmune diseases by tipping the balance between IL-12 family members.
Collapse
Affiliation(s)
- S Goriely
- Institute for Medical Immunology, Université Libre de Bruxelles, Charleroi, Belgium
| | | | | |
Collapse
|
123
|
Tzima S, Victoratos P, Kranidioti K, Alexiou M, Kollias G. Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-beta production. THE JOURNAL OF EXPERIMENTAL MEDICINE 2009. [PMID: 19398754 DOI: 10.1084/jem.200815822715044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heme oxygenase-1 (HO-1) is a key cytoprotective, antioxidant, and antiinflammatory molecule. The pathophysiological functions of HO-1 have been associated with its enzymatic activities in heme catabolism. We have examined the immune functions of HO-1 by its conditional ablation in myeloid cells (HO-1(M-KO) mice). We demonstrate that myeloid HO-1 is required for the activation of interferon (IFN) regulatory factor (IRF) 3 after Toll-like receptor 3 or 4 stimulation, or viral infection. HO-1-deficient macrophages show reduced expression of IFN-beta and of primary IRF3 target genes encoding RANTES, IP-10 and MCP-1. In the presence of polyI:C, myeloid HO-1 knockout mice infected with Listeria monocytogenes, a model dependent on IFN-beta production, showed enhanced bacterial clearance and survival, whereas control mice succumbed to infection. Moreover, after induction of experimental autoimmune encephalomyelitis, mice with myeloid-specific HO-1 deficiency developed a higher incidence and an exacerbated, nonremitting clinical disease correlating with persistent activation of antigen-presenting cells, enhanced infiltration of Th17 cells, and a nonregressing myelin-specific T cell reactivity. Notably, these defects were rectified by exogenous administration of IFN-beta, confirming that HO-1 functions directly upstream of this critical immune pathway. These results uncover a novel direct function for myeloid HO-1 in the regulation of IFN-beta production, establishing HO-1 as a critical early mediator of the innate immune response.
Collapse
Affiliation(s)
- Sotiria Tzima
- Biomedical Sciences Research Center Alexander Fleming, Vari 166-72, Greece
| | | | | | | | | |
Collapse
|
124
|
Tzima S, Victoratos P, Kranidioti K, Alexiou M, Kollias G. Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-beta production. ACTA ACUST UNITED AC 2009; 206:1167-79. [PMID: 19398754 PMCID: PMC2715044 DOI: 10.1084/jem.20081582] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Heme oxygenase–1 (HO-1) is a key cytoprotective, antioxidant, and antiinflammatory molecule. The pathophysiological functions of HO-1 have been associated with its enzymatic activities in heme catabolism. We have examined the immune functions of HO-1 by its conditional ablation in myeloid cells (HO-1M-KO mice). We demonstrate that myeloid HO-1 is required for the activation of interferon (IFN) regulatory factor (IRF) 3 after Toll-like receptor 3 or 4 stimulation, or viral infection. HO-1–deficient macrophages show reduced expression of IFN-β and of primary IRF3 target genes encoding RANTES, IP-10 and MCP-1. In the presence of polyI:C, myeloid HO-1 knockout mice infected with Listeria monocytogenes, a model dependent on IFN-β production, showed enhanced bacterial clearance and survival, whereas control mice succumbed to infection. Moreover, after induction of experimental autoimmune encephalomyelitis, mice with myeloid-specific HO-1 deficiency developed a higher incidence and an exacerbated, nonremitting clinical disease correlating with persistent activation of antigen-presenting cells, enhanced infiltration of Th17 cells, and a nonregressing myelin-specific T cell reactivity. Notably, these defects were rectified by exogenous administration of IFN-β, confirming that HO-1 functions directly upstream of this critical immune pathway. These results uncover a novel direct function for myeloid HO-1 in the regulation of IFN-β production, establishing HO-1 as a critical early mediator of the innate immune response.
Collapse
Affiliation(s)
- Sotiria Tzima
- Biomedical Sciences Research Center Alexander Fleming, Vari 166-72, Greece
| | | | | | | | | |
Collapse
|
125
|
Christensen JE, Simonsen S, Fenger C, Sørensen MR, Moos T, Christensen JP, Finsen B, Thomsen AR. Fulminant lymphocytic choriomeningitis virus-induced inflammation of the CNS involves a cytokine-chemokine-cytokine-chemokine cascade. THE JOURNAL OF IMMUNOLOGY 2009; 182:1079-87. [PMID: 19124751 DOI: 10.4049/jimmunol.182.2.1079] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Intracerebral inoculation of immunocompetent mice with lymphocytic choriomeningitis virus (LCMV) normally results in fatal CD8+ T cell mediated meningoencephalitis. However, in CXCL10-deficient mice, the virus-induced CD8+ T cell accumulation in the neural parenchyma is impaired, and only 30-50% of the mice succumb to the infection. Similar results are obtained in mice deficient in the matching chemokine receptor, CXCR3. Together, these findings point to a key role for CXCL10 in regulating the severity of the LCMV-induced inflammatory process. For this reason, we now address the mechanisms regulating the expression of CXCL10 in the CNS of LCMV-infected mice. Using mice deficient in type I IFN receptor, type II IFN receptor, or type II IFN, as well as bone marrow chimeras expressing CXCL10 only in resident cells or only in bone marrow-derived cells, we analyzed the up-stream regulation as well as the cellular source of CXCL10. We found that expression of CXCL10 initially depends on signaling through the type I IFN receptor, while late expression and up-regulation requires type II IFN produced by the recruited CD8+ T cells. Throughout the infection, the producers of CXCL10 are exclusively resident cells of the CNS, and astrocytes are the dominant expressors in the neural parenchyma, not microglial cells or recruited bone marrow-derived cell types. These results are consistent with a model suggesting a bidirectional interplay between resident cells of the CNS and the recruited virus-specific T cells with astrocytes as active participants in the local antiviral host response.
Collapse
Affiliation(s)
- Jeanette E Christensen
- University of Copenhagen, Department of International Health, Immunology and Microbiology, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
126
|
Bjurstöm H, Wang J, Wang J, Ericsson I, Bengtsson M, Liu Y, Kumar-Mendu S, Issazadeh-Navikas S, Birnir B. GABA, a natural immunomodulator of T lymphocytes. J Neuroimmunol 2008; 205:44-50. [PMID: 18954912 DOI: 10.1016/j.jneuroim.2008.08.017] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 08/27/2008] [Accepted: 08/27/2008] [Indexed: 11/16/2022]
Abstract
gamma-aminobutyric acid (GABA) is the main neuroinhibitory transmitter in the brain. Here we show that GABA in the extracellular space may affect the fate of pathogenic T lymphocytes entering the brain. We examined in encephalitogenic T cells if they expressed functional GABA channels that could be activated by the low (nM-1 microM), physiological concentrations of GABA present around neurons in the brain. The cells expressed the alpha1, alpha4, beta2, beta3, gamma1 and delta GABAA channel subunits and formed functional, extrasynaptic-like GABA channels that were activated by 1 microM GABA. 100 nM and higher GABA concentrations decreased T cell proliferation. The results are consistent with GABA being immunomodulatory.
Collapse
Affiliation(s)
- Helen Bjurstöm
- Lund University, Diabetic Centre, CRC, Department of Clinical Sciences, Malmö, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
127
|
Qin H, Niyongere SA, Lee SJ, Baker BJ, Benveniste EN. Expression and functional significance of SOCS-1 and SOCS-3 in astrocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:3167-76. [PMID: 18713987 PMCID: PMC2836124 DOI: 10.4049/jimmunol.181.5.3167] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Astrocytes play a number of important physiological roles in CNS homeostasis. Inflammation stimulates astrocytes to secrete cytokines and chemokines that guide macrophages/microglia and T cells to sites of injury/inflammation. Herein, we describe how these processes are controlled by the suppressor of cytokine signaling (SOCS) proteins, a family of proteins that negatively regulate adaptive and innate immune responses. In this study, we describe that the immunomodulatory cytokine IFN-beta induces SOCS-1 and SOCS-3 expression in primary astrocytes at the transcriptional level. SOCS-1 and SOCS-3 transcriptional activity is induced by IFN-beta through IFN-gamma activation site (GAS) elements within their promoters. Studies in STAT-1alpha-deficient astrocytes indicate that STAT-1alpha is required for IFN-beta-induced SOCS-1 expression, while STAT-3 small interfering RNA studies demonstrate that IFN-beta-induced SOCS-3 expression relies on STAT-3 activation. Specific small interfering RNA inhibition of IFN-beta-inducible SOCS-1 and SOCS-3 in astrocytes enhances their proinflammatory responses to IFN-beta stimulation, such as heightened expression of the chemokines CCL2 (MCP-1), CCL3 (MIP-1alpha), CCL4 (MIP-1beta), CCL5 (RANTES), and CXCL10 (IP-10), and promoting chemotaxis of macrophages and CD4(+) T cells. These results indicate that IFN-beta induces SOCS-1 and SOCS-3 in primary astrocytes to attenuate its own chemokine-related inflammation in the CNS.
Collapse
Affiliation(s)
- Hongwei Qin
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | | | | | | | | |
Collapse
|
128
|
Prinz M, Schmidt H, Mildner A, Knobeloch KP, Hanisch UK, Raasch J, Merkler D, Detje C, Gutcher I, Mages J, Lang R, Martin R, Gold R, Becher B, Brück W, Kalinke U. Distinct and nonredundant in vivo functions of IFNAR on myeloid cells limit autoimmunity in the central nervous system. Immunity 2008; 28:675-86. [PMID: 18424188 DOI: 10.1016/j.immuni.2008.03.011] [Citation(s) in RCA: 319] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 01/27/2008] [Accepted: 03/04/2008] [Indexed: 12/22/2022]
Abstract
The action of type I interferons in the central nervous system (CNS) during autoimmunity is largely unknown. Here, we demonstrate elevated interferon beta concentrations in the CNS, but not blood, of mice with experimental autoimmune encephalomyelitis (EAE), a model for CNS autoimmunity. Furthermore, mice devoid of the broadly expressed type I IFN receptor (IFNAR) developed exacerbated clinical disease accompanied by a markedly higher inflammation, demyelination, and lethality without shifting the T helper 17 (Th17) or Th1 cell immune response. Whereas adoptive transfer of encephalitogenic T cells led to enhanced disease in Ifnar1(-/-) mice, newly created conditional mice with B or T lymphocyte-specific IFNAR ablation showed normal EAE. The engagement of IFNAR on neuroectodermal CNS cells had no protective effect. In contrast, absence of IFNAR on myeloid cells led to severe disease with an enhanced effector phase and increased lethality, indicating a distinct protective function of type I IFNs during autoimmune inflammation of the CNS.
Collapse
Affiliation(s)
- Marco Prinz
- Department of Neuropathology, University of Freiburg, D-79106 Freiburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Abstract
Although interferon-beta is the most popular treatment for multiple sclerosis, its mechanism of action remains enigmatic. In this issue of Immunity, Prinz et al. (2008) elucidate an intriguing portrait of the pleiotropic effects of type 1 interferons in taming brain inflammation.
Collapse
Affiliation(s)
- Robert C Axtell
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
130
|
Guo B, Chang EY, Cheng G. The type I IFN induction pathway constrains Th17-mediated autoimmune inflammation in mice. J Clin Invest 2008; 118:1680-90. [PMID: 18382764 DOI: 10.1172/jci33342] [Citation(s) in RCA: 297] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 02/13/2008] [Indexed: 01/17/2023] Open
Abstract
IFN-beta, a type I IFN, is widely used for the treatment of MS. However, the mechanisms behind its therapeutic efficacy are not well understood. Using a murine model of MS, EAE, we demonstrate that the Th17-mediated development of autoimmune disease is constrained by Toll-IL-1 receptor domain-containing adaptor inducing IFN-beta-dependent (TRIF-dependent) type I IFN production and its downstream signaling pathway. Mice with defects in TRIF or type I IFN receptor (IFNAR) developed more severe EAE. Notably, these mice exhibited marked CNS inflammation, as manifested by increased IL-17 production. In addition, IFNAR-dependent signaling events were essential for negatively regulating Th17 development. Finally, IFN-beta-mediated IL-27 production by innate immune cells was critical for the immunoregulatory role of IFN-beta in the CNS autoimmune disease. Together, our findings not only may provide a molecular mechanism for the clinical benefits of IFN-beta in MS but also demonstrate a regulatory role for type I IFN induction and its downstream signaling pathways in limiting Th17 development and autoimmune inflammation.
Collapse
Affiliation(s)
- Beichu Guo
- Department of Microbiology, Immunology, and Molecular Genetics, 8-240 Factor Building, University of California, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
131
|
|
132
|
Eskan MA, Rose BG, Benakanakere MR, Lee MJ, Kinane DF. Sphingosine 1-phosphate 1 and TLR4 mediate IFN-beta expression in human gingival epithelial cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:1818-25. [PMID: 18209079 DOI: 10.4049/jimmunol.180.3.1818] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IFN-beta production is a critical step in human innate immune responses and is primarily controlled at the transcription level by highly ordered mechanisms. IFN-beta can be induced by pattern-recognition receptors such as the TLR4. S1P1 is a G protein-coupled receptor, which has a high affinity for sphingosine 1-phosphate (S1P). Although many of the receptors and signaling pathways leading to the expression of IFN-beta have been identified and characterized, it is still unclear how IFN-beta is regulated in primary human gingival epithelial cells (HGECs). In this study, we demonstrate that S1P1 and TLR4, acting in unison, play an important role in IFN-beta expression at the protein and mRNA level in HGECs. We demonstrate that the expression of both IFN-beta and IFN-inducible protein-10 (CXCL-10) is significantly up-regulated by LPS and S1P or LPS and a specific S1P1 agonist. This enhanced innate immune response is attenuated in HGECs by small interfering RNA knockdown of either TLR4 or S1P1. Moreover, we show that triggering of TLR4 results in the increased expression of S1P1 receptors. Furthermore, we found that IFN-regulatory factor 3 activation was maximized by LPS and S1P through PI3K. Our data show that triggering TLR4 increases S1P1, such that both TLR4 and S1P1 acting through PI3K enhancement of IFN-regulatory factor 3 activation increase IFN-beta expression in epithelial cells. The functional association between TLR4 and the S1P1 receptor demonstrates a novel mechanism in the regulation of IFN-beta and CXCL-10 in human primary gingival epithelial cells.
Collapse
Affiliation(s)
- Mehmet A Eskan
- Center for Oral Health and Systemic Disease, Department of Periodontics, Endodontics, and Dental Hygiene, University of Louisville School of Dentistry, 501 South Preston Street, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
133
|
Sommereyns C, Paul S, Staeheli P, Michiels T. IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS Pathog 2008; 4:e1000017. [PMID: 18369468 PMCID: PMC2265414 DOI: 10.1371/journal.ppat.1000017] [Citation(s) in RCA: 643] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Accepted: 01/30/2008] [Indexed: 12/13/2022] Open
Abstract
Interferons (IFN) exert antiviral, immunomodulatory and cytostatic activities. IFN-alpha/beta (type I IFN) and IFN-lambda (type III IFN) bind distinct receptors, but regulate similar sets of genes and exhibit strikingly similar biological activities. We analyzed to what extent the IFN-alpha/beta and IFN-lambda systems overlap in vivo in terms of expression and response. We observed a certain degree of tissue specificity in the production of IFN-lambda. In the brain, IFN-alpha/beta was readily produced after infection with various RNA viruses, whereas expression of IFN-lambda was low in this organ. In the liver, virus infection induced the expression of both IFN-alpha/beta and IFN-lambda genes. Plasmid electrotransfer-mediated in vivo expression of individual IFN genes allowed the tissue and cell specificities of the responses to systemic IFN-alpha/beta and IFN-lambda to be compared. The response to IFN-lambda correlated with expression of the alpha subunit of the IFN-lambda receptor (IL-28R alpha). The IFN-lambda response was prominent in the stomach, intestine and lungs, but very low in the central nervous system and spleen. At the cellular level, the response to IFN-lambda in kidney and brain was restricted to epithelial cells. In contrast, the response to IFN-alpha/beta was observed in various cell types in these organs, and was most prominent in endothelial cells. Thus, the IFN-lambda system probably evolved to specifically protect epithelia. IFN-lambda might contribute to the prevention of viral invasion through skin and mucosal surfaces.
Collapse
Affiliation(s)
- Caroline Sommereyns
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| | - Sophie Paul
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| | - Peter Staeheli
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Thomas Michiels
- Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, Brussels, Belgium
| |
Collapse
|
134
|
Kristjansdottir G, Sandling JK, Bonetti A, Roos IM, Milani L, Wang C, Gustafsdottir SM, Sigurdsson S, Lundmark A, Tienari PJ, Koivisto K, Elovaara I, Pirttilä T, Reunanen M, Peltonen L, Saarela J, Hillert J, Olsson T, Landegren U, Alcina A, Fernández O, Leyva L, Guerrero M, Lucas M, Izquierdo G, Matesanz F, Syvänen AC. Interferon regulatory factor 5 (IRF5) gene variants are associated with multiple sclerosis in three distinct populations. J Med Genet 2008; 45:362-9. [PMID: 18285424 PMCID: PMC2564860 DOI: 10.1136/jmg.2007.055012] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: IRF5 is a transcription factor involved both in the type I interferon and the toll-like receptor signalling pathways. Previously, IRF5 has been found to be associated with systemic lupus erythematosus, rheumatoid arthritis and inflammatory bowel diseases. Here we investigated whether polymorphisms in the IRF5 gene would be associated with yet another disease with features of autoimmunity, multiple sclerosis (MS). Methods: We genotyped nine single nucleotide polymorphisms and one insertion-deletion polymorphism in the IRF5 gene in a collection of 2337 patients with MS and 2813 controls from three populations: two case–control cohorts from Spain and Sweden, and a set of MS trio families from Finland. Results: Two single nucleotide polymorphism (SNPs) (rs4728142, rs3807306), and a 5 bp insertion-deletion polymorphism located in the promoter and first intron of the IRF5 gene, showed association signals with values of p<0.001 when the data from all cohorts were combined. The predisposing alleles were present on the same common haplotype in all populations. Using electrophoretic mobility shift assays we observed allele specific differences in protein binding for the SNP rs4728142 and the 5 bp indel, and by a proximity ligation assay we demonstrated increased binding of the transcription factor SP1 to the risk allele of the 5 bp indel. Conclusion: These findings add IRF5 to the short list of genes shown to be associated with MS in more than one population. Our study adds to the evidence that there might be genes or pathways that are common in multiple autoimmune diseases, and that the type I interferon system is likely to be involved in the development of these diseases.
Collapse
Affiliation(s)
- G Kristjansdottir
- Molecular Medicine, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Benveniste EN, Qin H. Type I interferons as anti-inflammatory mediators. ACTA ACUST UNITED AC 2007; 2007:pe70. [PMID: 18073382 DOI: 10.1126/stke.4162007pe70] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The type I interferons (IFNs), IFN-alpha and IFN-beta, are cytokines that have antiviral, antiproliferative, and immunomodulatory activities. Data are now emerging that suggest that type I IFNs are also important mediators of anti-inflammatory responses. These findings, largely driven by studies to explain the beneficial effects of IFN-beta in the treatment of multiple sclerosis, an autoimmune disease of the central nervous system, offer a number of mechanisms for the anti-inflammatory properties of type I IFNs. Type I IFNs, through their ability to induce the immunosuppressive cytokine interleukin-10 (IL-10), mediate the inhibition of proinflammatory gene products. In addition, type I IFNs induce other immunosuppressive mediators such as suppressor of cytokine signaling-1 (SOCS-1) and tristetrapolin (TTP), which act by divergent mechanisms to restore homeostasis to the immune system. Furthermore, type I IFNs mediate anti-inflammatory and protective effects in a variety of autoimmune disease models such as experimental colitis, experimental allergic encephalomyelitis, experimental arthritis, and neonatal inflammation. Here, we discuss the molecular basis for the anti-inflammatory properties of type I IFNs and their therapeutic potential in autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Etty N Benveniste
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA.
| | | |
Collapse
|
136
|
Trebst C, Heine S, Lienenklaus S, Lindner M, Baumgärtner W, Weiss S, Stangel M. Lack of interferon-beta leads to accelerated remyelination in a toxic model of central nervous system demyelination. Acta Neuropathol 2007; 114:587-96. [PMID: 17940777 DOI: 10.1007/s00401-007-0300-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Revised: 09/18/2007] [Accepted: 09/18/2007] [Indexed: 11/26/2022]
Abstract
Interferon-beta (IFN-beta) is a pleiotropic cytokine that is known to modulate the immune response in multiple sclerosis (MS), an inflammatory demyelinating disease of the central nervous system (CNS). Spontaneous remyelination and repair mechanisms in MS are mostly insufficient and contribute to clinical disability. Here, we investigated whether IFN-beta has a potential in modifying the extent of de- and remyelination in a toxic model of CNS demyelination induced by the copper chelator cuprizone. IFN-beta deficient (k/o) mice showed an accelerated spontaneous remyelination. However, the amount of remyelination after 6 weeks did not differ between the two groups. Demyelination in IFN-beta k/o mice was paralleled by a diminished astrocytic and microglia response as compared with wildtype controls, whereas the accelerated remyelination was paralleled by an increased number of oligodendrocyte precursor cells (OPC) within the demyelinated lesion at the beginning of the remyelination phase. We hypothesize that the absence of IFN-beta leads to more efficient recruitment and proliferation of OPC already during demyelination, thus allowing early remyelination. These results demonstrate that IFN-beta is able to alter remyelination in the absence of an immune-mediated demyelination.
Collapse
Affiliation(s)
- Corinna Trebst
- Department of Neurology, Medical School Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
137
|
Kovarik P, Sauer I, Schaljo B. Molecular mechanisms of the anti-inflammatory functions of interferons. Immunobiology 2007; 212:895-901. [PMID: 18086388 PMCID: PMC3963707 DOI: 10.1016/j.imbio.2007.09.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 09/28/2007] [Indexed: 01/14/2023]
Abstract
Interferons are pleiotropic cytokines with important proinflammatory functions required in defence against infections with bacteria, viruses and multicellular parasites. In recent years, fundamental functions of interferons in other processes such as cancer immunosurveillance, immune homeostasis and immunosuppression have been established. In addition, anti-inflammatory roles of interferons are well-documented in several inflammatory disease models in the mouse, most importantly in experimental autoimmune encephalomyelitis that resembles multiple sclerosis in humans. While the beneficial effects of interferons in such disease models are known, the molecular mechanisms remain poorly understood. Only recently a few molecular principles for the anti-inflammatory properties of interferons at the cellular level have been revealed. They include the ability of interferons to reduce the expression of the receptors for the inflammation-related cytokines IL-1 and IL-4, or to increase the expression of the potent anti-inflammatory genes tristetraprolin and Twist. However, the individual contribution of these anti-inflammatory responses to the overall beneficial effects of interferons in inflammatory diseases is still an open question. Also, the reason for the apparently limited number of tissues that are susceptible to the anti-inflammatory functions of interferons remains enigmatic. This review summarizes the present knowledge of the anti-inflammatory effects of interferons, and describes the currently known molecular mechanisms that may help explain the benefits of interferon signalling in several inflammatory diseases.
Collapse
Affiliation(s)
- Pavel Kovarik
- Max F Perutz Laboratories, University of Vienna, Dr Bohr-Gasse 9, A-1030, Vienna, Austria.
| | | | | |
Collapse
|
138
|
Abstract
This article analyzes the conceptual and technological context in which, over a period of 50 years, exploration of the biological and clinical significance of type I interferon has evolved. The elaboration of techniques for production and purification of mouse and human interferons and the establishment of laboratory-size production units have been of crucial importance in this process. Animal experiments have been invaluable for elucidation of mechanisms underlying the in vivo antiviral, anti-tumour and immunomodulatory potential of interferon, but have been of limited help to define the areas of clinical applicability. Proof of principle for applications as they are established today has come from clinical trials performed quite independently of evidence from animal experiments.
Collapse
Affiliation(s)
- Alfons Billiau
- Rega Institute, University of Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium.
| |
Collapse
|
139
|
Bagnato F, Evangelou IE, Gallo A, Gaindh D, Yao K. The effect of interferon-β on black holes in patients with multiple sclerosis. Expert Opin Biol Ther 2007; 7:1079-91. [PMID: 17665995 DOI: 10.1517/14712598.7.7.1079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Multiple sclerosis (MS) is an immunological disorder of the CNS. Linked to an initial transient inflammation as the result of blood-brain barrier leakage, the disease progresses into a neurodegenerative phase. MRI is the most powerful paraclinical tool for diagnosing and monitoring MS. Although contrast enhancing lesions are the visible events of blood-brain barrier breakdown, accumulation of hypointense lesions, namely black holes, are recognised as irreversible axonal loss. IFN-beta is administered as a first-line drug in MS patients. However, whether the effect of IFN-beta extends beyond just prevention of blood-brain barrier leakage and further prevents the formation of black holes or promotes their recovery once formed, is not yet understood.
Collapse
Affiliation(s)
- Francesca Bagnato
- National Institute of Health, National Institute for Neurological Diseases and Stroke, Neuroimmunology Branch, Bethesda, MD 20892-1400, USA.
| | | | | | | | | |
Collapse
|
140
|
Molle C, Nguyen M, Flamand V, Renneson J, Trottein F, De Wit D, Willems F, Goldman M, Goriely S. IL-27 Synthesis Induced by TLR Ligation Critically Depends on IFN Regulatory Factor 3. THE JOURNAL OF IMMUNOLOGY 2007; 178:7607-15. [PMID: 17548596 DOI: 10.4049/jimmunol.178.12.7607] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
IL-27 is a heterodimeric cytokine composed of EBV-induced gene 3 and p28. Produced by dendritic cells (DCs) in response to TLR ligands, IL-27 recently emerged as a key regulator of inflammatory responses. In this study, we first demonstrate that Toll/IL-1R-containing adaptor inducing IFN-beta and its associated IFN regulatory factor (IRF) 3 transcription factor are critically involved in IL-27p28 expression in mouse DCs stimulated by TLR ligands. We then show that IL-27 serum levels are dramatically reduced in IRF3(-/-) upon LPS injection, indicating a critical role for IRF3 in TLR4-mediated IL-27 production in vivo. We identified an IRF3-binding site within the IL-27p28 promoter region which is required for IL-27p28 gene activation in reporter gene assays. In human DCs, IL-27p28 mRNA was preferentially induced by Toll/IL-1R-containing adaptor inducing IFN-beta-coupled TLR ligands and following CMV infection. Furthermore, chromatin immunoprecipitation studies demonstrate that IRF3 is recruited to the endogenous p28 promoter in TLR4-stimulated human DCs. We conclude that IRF3 activation is a master switch for IL-27 synthesis.
Collapse
Affiliation(s)
- Céline Molle
- Institute for Medical Immunology, Université Libre de Bruxelles, 8 rue Adrienne Bolland, B-6041 Charleroi-Gosselies, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Fabis MJ, Scott GS, Kean RB, Koprowski H, Hooper DC. Loss of blood-brain barrier integrity in the spinal cord is common to experimental allergic encephalomyelitis in knockout mouse models. Proc Natl Acad Sci U S A 2007; 104:5656-61. [PMID: 17372191 PMCID: PMC1838442 DOI: 10.1073/pnas.0701252104] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2007] [Indexed: 11/18/2022] Open
Abstract
Experimental allergic encephalomyelitis (EAE) is an inflammatory demyelinating disease of the CNS that is used to model certain parameters of multiple sclerosis. To establish the relative contributions of T cell reactivity, the loss of blood-brain barrier (BBB) integrity, CNS inflammation, and lesion formation toward the pathogenesis of EAE, we assessed the incidence of EAE and these parameters in mice lacking NF-kappaB, TNF-alpha, IFN-alphabeta receptors, IFN-gamma receptors, and inducible nitric oxide synthase. Although increased myelin oligodendrocyte glycoprotein-specific T cell reactivity was generally associated with a more rapid onset or increased disease severity, the loss of BBB integrity and cell accumulation in spinal cord tissues was invariably associated with the development of neurological disease signs. Histological and real-time RT-PCR analyses revealed differences in the nature of immune/inflammatory cell accumulation in the spinal cord tissues of the different mouse strains. On the other hand, disease severity during the acute phase of EAE directly correlated with the extent of BBB permeability. Thus, the loss of BBB integrity seems to be a requisite event in the development of EAE and can occur in the absence of important inflammatory mediators.
Collapse
Affiliation(s)
- Marzena J. Fabis
- *Center for Neurovirology, Department of Cancer Biology
- Biotechnology Foundation Laboratories, and
| | - Gwen S. Scott
- *Center for Neurovirology, Department of Cancer Biology
| | - Rhonda B. Kean
- *Center for Neurovirology, Department of Cancer Biology
- Biotechnology Foundation Laboratories, and
| | - Hilary Koprowski
- *Center for Neurovirology, Department of Cancer Biology
- Biotechnology Foundation Laboratories, and
| | - D. Craig Hooper
- *Center for Neurovirology, Department of Cancer Biology
- Biotechnology Foundation Laboratories, and
- Department of Neurological Surgery, Thomas Jefferson University, 1020 Locust Street, JAH 454, Philadelphia, PA 19107-6799
| |
Collapse
|
142
|
Burdeinick-Kerr R, Wind J, Griffin DE. Synergistic roles of antibody and interferon in noncytolytic clearance of Sindbis virus from different regions of the central nervous system. J Virol 2007; 81:5628-36. [PMID: 17376910 PMCID: PMC1900320 DOI: 10.1128/jvi.01152-06] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Sindbis virus (SINV) is an alphavirus that causes infection of neurons and encephalomyelitis in adult immunocompetent mice. Recovery can occur without apparent neurological damage. To better define the factors facilitating noncytolytic clearance of SINV in different regions of the central nervous system (CNS) and the roles of innate and adaptive immune responses at different times during infection, we have characterized SINV infection and clearance in the brain, brain stem, and spinal cords of severe combined immunodeficiency (SCID) and C57BL/6 (wild-type [WT]) mice and mice deficient in beta interferon (IFN-beta) (BKO), antibody (muMT), IFN-gamma (GKO), IFN-gamma receptor (GRKO), and both antibody and IFN-gamma (muMT/GKO). WT mice cleared infectious virus by day 8, while SCID mice had persistent virus replication at all sites. For 3 days after infection, BKO mice had higher titers at all sites than WT mice, despite similar IFN-alpha production, but cleared virus similarly. GKO and GRKO mice cleared infectious virus from all sites by days 8 to 10 and, like WT mice, displayed transient reactivation at 12 to 22 days. muMT mice did not clear virus from the brain, and clearance from the brain stem and lumbar spinal cord was delayed, followed by reactivation. Eighty-one days after infection, muMT/GKO mice had not cleared virus from any site, but titers were lower than for SCID mice. These studies show that IFN-beta is independently important for early control of CNS virus replication, that antiviral antibody is critical for clearance from the brain, and that both antibody and IFN-gamma contribute to prevention of reactivation after initial clearance.
Collapse
Affiliation(s)
- Rebeca Burdeinick-Kerr
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
143
|
Yarilina A, DiCarlo E, Ivashkiv LB. Suppression of the effector phase of inflammatory arthritis by double-stranded RNA is mediated by type I IFNs. THE JOURNAL OF IMMUNOLOGY 2007; 178:2204-11. [PMID: 17277125 DOI: 10.4049/jimmunol.178.4.2204] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Innate immune receptors that recognize nucleic acids, such as TLRs and RNA helicases, are potent activators of innate immunity that have been implicated in the induction and exacerbation of autoimmunity and inflammatory arthritis. Polyriboinosine-polyribocytidylic acid sodium salt (poly(IC)) is a mimic of dsRNA and viral infection that activates TLR3 and the RNA helicases retinoic acid-induced gene-1 and melanoma differentiation-associated gene-5, and strongly induces type I IFN production. We analyzed the effects of systemic delivery of poly(IC) on the inflammatory effector phase of arthritis using the collagen Ab-induced and KRN TCR-transgenic mouse serum-induced models of immune complex-mediated experimental arthritis. Surprisingly, poly(IC) suppressed arthritis, and suppression was dependent on type I IFNs that inhibited synovial cell proliferation and inflammatory cytokine production. Administration of exogenous type I IFNs was sufficient to suppress arthritis. These results suggest a regulatory role for innate immune receptors for dsRNA in modulating inflammatory arthritis and provide additional support for an anti-inflammatory function of type I IFNs in arthritis that directly contrasts with a pathogenic role in promoting autoimmunity in systemic lupus.
Collapse
MESH Headings
- Animals
- Antiviral Agents/immunology
- Antiviral Agents/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Autoimmunity/drug effects
- Immunity, Innate/drug effects
- Inflammation/immunology
- Inflammation/pathology
- Interferon Inducers/immunology
- Interferon Inducers/pharmacology
- Interferon Type I/immunology
- Interferon Type I/pharmacology
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Mice
- Mice, Inbred NOD
- Poly I-C/immunology
- Poly I-C/pharmacology
- RNA Helicases/immunology
- RNA, Double-Stranded/immunology
- RNA, Double-Stranded/pharmacology
- Toll-Like Receptor 3/immunology
- Virus Diseases/immunology
- Virus Diseases/pathology
Collapse
Affiliation(s)
- Anna Yarilina
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | |
Collapse
|
144
|
Wahl C, Bochtler P, Schirmbeck R, Reimann J. Type I IFN-producing CD4 Valpha14i NKT cells facilitate priming of IL-10-producing CD8 T cells by hepatocytes. THE JOURNAL OF IMMUNOLOGY 2007; 178:2083-93. [PMID: 17277112 DOI: 10.4049/jimmunol.178.4.2083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Upon entering the liver CD8 T cells encounter large numbers of NKT cells patrolling the hepatocyte (HC) surface facing the perisinusoidal space. We asked whether hepatic NKT cells modulate the priming of CD8 T cells by HC. Hepatic (alpha-galactosyl-ceramide-loaded CD1d dimer binding) NKT cells produce predominantly IL-4 when stimulated with glycolipid-presenting HC but predominantly IFN-gamma when stimulated with glycolipid-presenting dendritic cells. These NKT cells prime naive CD8 T cells to a (K(b)-presented) peptide ligand if they simultaneously recognize a CD1d-binding glycolipid presented to them on the surface of the responding CD8 T cells that they prime. No IL-10-producing CD8 T cells are detected if these T cells are primed by either HC or NKT cells. In contrast, IL-10 is produced by HC-primed CD8 T cells if IFN-beta-producing NKT cells are coactivated by the same HC presenting a glycolipid (in the context of CD1d) and an antigenic peptide (in the context of K(b)). Hence, IL-10-producing CD8 T cells are generated in a type I IFN-dependent manner if the three cell types (CD8 T cells, NKT cells, and ligand-presenting HC) specifically and closely interact. IL-10-producing CD8 T cells generated under these conditions down-modulate IL-2 (and proliferative) responses of naive CD4 or CD8 T cells primed by DC. If in close proximity, NKT cells can thus locally modulate the phenotype of CD8 T cells during their priming by HC thereby limiting the local activation of proinflammatory immune effector cells and protecting the liver against immune injury.
Collapse
Affiliation(s)
- Christian Wahl
- Department of Internal Medicine I, University of Ulm, Ulm, Germany
| | | | | | | |
Collapse
|
145
|
Paul S, Ricour C, Sommereyns C, Sorgeloos F, Michiels T. Type I interferon response in the central nervous system. Biochimie 2007; 89:770-8. [PMID: 17408841 DOI: 10.1016/j.biochi.2007.02.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Accepted: 02/16/2007] [Indexed: 12/25/2022]
Abstract
This review is dedicated to the influence of type I IFNs (also called IFN-alpha/beta) in the central nervous system (CNS). Studies in mice with type I IFN receptor or IFN-beta gene deficiency have highlighted the importance of the type I IFN system against CNS viral infections and non-viral autoimmune disorders. Direct antiviral effects of type I IFNs appear to be crucial in limiting early spread of a number of viruses in CNS tissues. Type I IFNs have also proved to be beneficial in autoimmune disorders like multiple sclerosis or experimental autoimmune encephalitis, probably through immunomodulatory effects. Increasing efforts are done to characterize IFN expression and response in the CNS: to identify type I IFN producing cells, to decipher pathways leading to type I IFN expression in those cells, and to identify responding cells. However, reversible and irreversible damages consecutive to chronic exposure of the CNS to type I IFNs underline the importance of a tightly regulated type I IFN homeostasis in this organ.
Collapse
Affiliation(s)
- Sophie Paul
- Université catholique de Louvain, Christian de Duve Institute of Cellular Pathology, Microbial Pathogenesis Unit, MIPA-VIRO 74-49, 74, avenue Hippocrate, B-1200, Brussels, Belgium
| | | | | | | | | |
Collapse
|
146
|
Mastronardi FG, Tsui H, Winer S, Wood DD, Selvanantham T, Galligan C, Fish EN, Dosch HM, Moscarello MA. Synergy between paclitaxel plus an exogenous methyl donor in the suppression of murine demyelinating diseases. Mult Scler 2007; 13:596-609. [PMID: 17548438 DOI: 10.1177/1352458506072167] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Progressive demyelination in multiple sclerosis (MS) reflects the negative balance between myelin damage and repair due to physical and molecular barriers, such as astrocytic glial scars, between oligodendrocytes and target neurons. In this paper, we show that combination therapy with paclitaxel (Taxol) plus the universal methyl-donor, vitamin B12CN (B12CN), dramatically limits progressive demyelination, and enhances remyelination in several independent, immune and nonimmune, in vivo and in vitro model systems. Combination therapy significantly reduced clinical signs of EAE in SJL mice, as well as the spontaneously demyelinating ND4 transgenic mouse. Astrocytosis was normalised in parallel to ultrastructural and biochemical evidence of remyelination. The combination therapy suppressed T cell expansion, reduced IFN-gamma, while enhancing IFN-beta and STAT-1 expression, STAT-1 phosphorylation and methylation of STAT-1 and MBP in the brain. Paclitaxel/B12CN has nearly identical effects to the previously described combination of IFN-beta/ B12CN, whose clinical usefulness is transient because of IFN-neutralising antibodies, not observed (or expected) with the present drug combination. This report provides a mechanistic foundation for the development of a new therapeutic strategy in humans with MS.
Collapse
Affiliation(s)
- F G Mastronardi
- Department of Structural Biology and Biochemistry, The Hospital for Sick Children, University Health Network, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Barca O, Seoane M, Ferré S, Prieto JM, Lema M, Señarís R, Arce VM. Mechanisms of interferon-beta-induced survival in fetal and neonatal primary astrocytes. Neuroimmunomodulation 2007; 14:39-45. [PMID: 17700039 DOI: 10.1159/000107287] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2006] [Accepted: 02/25/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We have previously shown that interferon-beta (IFN-beta) is a potent promoter of astrocyte survival. Although the mechanism(s) by which IFN-beta promotes astrocyte survival have not been completely elucidated, it has been shown that IFN-beta directly stimulates survival signaling pathways. In the present report, we took advantage of the differences in the susceptibility of fetal and neonatal astrocytes to apoptosis to further investigate the mechanism(s) underlying the antiapoptotic effect of IFN-beta. METHODS Primary monolayer cultures of cortical astrocytes were established from neonatal (3- to 6-day-old) or fetal (embryonic days: E15 or E17) Sprague-Dawley rat cerebral cortices. Apoptotic cell death was determined by fluorescent-microscopic analysis of staining patterns of cell DNA with Hoechst 33258, and determination of annexin V binding.Akt phosphorylation was detected by Western blottingusing a commercial kit that allows specific recognition of both non-phosphorylated and serine-phosphorylated Akt. RESULTS In the present work, we have found that primary astrocytes obtained from neonatal rats are resistant to apoptosis induced by serum starvation, though cell death may be induced by combining serum starvation with sodium butyrate treatment. This effect is counteracted by IFN-beta treatment through a mechanism that involves phosphatidylinositol 3-kinase stimulation. CONCLUSIONS IFN-beta can be considered as a neuroprotective agent and, therefore, part of its beneficial effects in multiple sclerosis (MS) treatment may depend on its capacity to protect astrocytes against the apoptotic cell death that occurs in the course of the MS lesions.
Collapse
Affiliation(s)
- Olga Barca
- Departamento de Fisioloxía, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
148
|
Teige I, Liu Y, Issazadeh-Navikas S. IFN-beta inhibits T cell activation capacity of central nervous system APCs. THE JOURNAL OF IMMUNOLOGY 2006; 177:3542-53. [PMID: 16951313 DOI: 10.4049/jimmunol.177.6.3542] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously investigated the physiological effects of IFN-beta on chronic CNS inflammation and shown that IFN-beta(-/-) mice develop a more severe experimental autoimmune encephalomyelitis than their IFN-beta(+/-) littermates. This result was shown to be associated with a higher activation state of the glial cells and a higher T cell cytokine production in the CNS. Because this state suggested a down-regulatory effect of IFN-beta on CNS-specific APCs, these results were investigated further. We report that IFN-beta pretreatment of astrocytes and microglia (glial cells) indeed down-modulate their capacity to activate autoreactive Th1 cells. First, we investigated the intrinsic ability of glial cells as APCs and report that glial cells prevent autoreactive Th1 cells expansion while maintaining Ag-specific T cell effector functions. However, when the glial cells are treated with IFN-beta before coculture with T cells, the effector functions of T cells are impaired as IFN-gamma, TNF-alpha, and NO productions are decreased. Induction of the T cell activation marker, CD25 is also reduced. This suppression of T cell response is cell-cell dependent, but it is not dependent on a decrease in glial expression of MHC class II or costimulatory molecules. We propose that IFN-beta might exert its beneficial effects mainly by reducing the Ag-presenting capacity of CNS-specific APCs, which in turn inhibits the effector functions of encephalitogenic T cells. This affect is of importance because activation of encephalitogenic T cells within the CNS is a prerequisite for the development of a chronic progressive CNS inflammation.
Collapse
Affiliation(s)
- Ingrid Teige
- Neuroinflammation Unit, Section for Immunology, Institute for Experimental Medical Science, Lund University, Lund, Sweden
| | | | | |
Collapse
|
149
|
Abstract
Nearly half a century has passed since the first published description of interferons (IFNs). This commentary introduces the four accompanying review articles on type I IFN research and attempts to relate how the field of IFN research has been changing during its history.
Collapse
Affiliation(s)
- Jan Vilcek
- Department of Microbiology, New York University School of Medicine, New York, New York 10016, USA.
| |
Collapse
|
150
|
Schaefer C, Hidalgo TR, Cashion L, Petry H, Brooks A, Szymanski P, Qian HS, Gross C, Wang P, Liu P, Goldman C, Rubanyi GM, Harkins RN. Gene-based delivery of IFN-beta is efficacious in a murine model of experimental allergic encephalomyelitis. J Interferon Cytokine Res 2006; 26:449-54. [PMID: 16800783 DOI: 10.1089/jir.2006.26.449] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Experimental allergic encephalomyelitis (EAE) is a model of central nervous system (CNS) inflammation that follows immunization with certain CNS antigens. The course and clinical manifestations of EAE are similar to those of multiple sclerosis (MS) in humans; therefore, EAE has become an accepted animal model to study MS. The purpose of this study was to demonstrate that systemic expression of murine interferon-beta (IFN-beta) (MuIFN-beta), following intramuscular (i.m.) delivery of plasmid DNA encoding MuIFN-beta to the hind limb of mice, is effective in reducing the clinical manifestations of disease in a model of EAE. The results of the study demonstrate that gene-based delivery of MuIFN-beta caused significantly decreased clinical scores compared with delivery of the null vector. A single injection of the MuIFN-beta plasmid was as effective in reducing the severity of the disease as an every other day injection of MuIFN-beta protein.
Collapse
Affiliation(s)
- Caralee Schaefer
- Department of Pharmacology, Berlex Biosciences, Richmond, CA 94804, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|