101
|
Abstract
Some immunologists have characterized T helper (Th)17 T cells as the master mediators of tissue damage in a variety of pathological conditions. New data now demonstrate that Th1 and Th17 T cells are independently capable of inducing disease in two established models of autoimmunity. Thus, the role of Th17 cytokines as the central mediators of pathological tissue damage seems to require clarification.
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences and the Interdepartmental Program in Immunology, Beckman Center for Molecular Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
102
|
Shi M, Lin TH, Appell KC, Berg LJ. Janus-kinase-3-dependent signals induce chromatin remodeling at the Ifng locus during T helper 1 cell differentiation. Immunity 2008; 28:763-73. [PMID: 18549798 PMCID: PMC2587400 DOI: 10.1016/j.immuni.2008.04.016] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 04/10/2008] [Accepted: 04/11/2008] [Indexed: 01/06/2023]
Abstract
Differentiation of naive CD4+ T cells into T helper type 1 (Th1) effector cells requires both T cell receptor (TCR) signaling and cytokines such as interleukin-12 and interferon gamma (IFN-gamma). Here, we report that a third cytokine signal, mediated by the Janus family tyrosine kinase 3 (Jak3) and signal transducer and activator of transcription 5 (STAT5) pathway, is also required for Th1 cell differentiation. In the absence of Jak3-dependent signals, naive CD4+ T cells proliferate robustly but produce little IFN-gamma after Th1 cell polarization in vitro. This defect is not due to reduced activation of STAT1 or STAT4 or to impaired upregulation of the transcription factor T-bet. Instead, we find that T-bet binding to the Ifng promoter is greatly diminished in the absence of Jak3-dependent signals, correlating with a decrease in Ifng promoter accessibility and histone acetylation. These data indicate that Jak3 regulates epigenetic modification and chromatin remodeling of the Ifng locus during Th1 cell differentiation.
Collapse
Affiliation(s)
- Min Shi
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
103
|
Abstract
CD4+ T cells can differentiate into multiple effector subsets, but the potential roles of these subsets in anti-tumor immunity have not been fully explored. Seeking to study the impact of CD4+ T cell polarization on tumor rejection in a model mimicking human disease, we generated a new MHC class II-restricted, T-cell receptor (TCR) transgenic mouse model in which CD4+ T cells recognize a novel epitope in tyrosinase-related protein 1 (TRP-1), an antigen expressed by normal melanocytes and B16 murine melanoma. Cells could be robustly polarized into Th0, Th1, and Th17 subtypes in vitro, as evidenced by cytokine, chemokine, and adhesion molecule profiles and by surface markers, suggesting the potential for differential effector function in vivo. Contrary to the current view that Th1 cells are most important in tumor rejection, we found that Th17-polarized cells better mediated destruction of advanced B16 melanoma. Their therapeutic effect was critically dependent on interferon-gamma (IFN-gamma) production, whereas depletion of interleukin (IL)-17A and IL-23 had little impact. Taken together, these data indicate that the appropriate in vitro polarization of effector CD4+ T cells is decisive for successful tumor eradication. This principle should be considered in designing clinical trials involving adoptive transfer-based immunotherapy of human malignancies.
Collapse
|
104
|
Nakamura M, Jo JI, Tabata Y, Ishikawa O. Controlled delivery of T-box21 small interfering RNA ameliorates autoimmune alopecia (Alopecia Areata) in a C3H/HeJ mouse model. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:650-8. [PMID: 18245811 DOI: 10.2353/ajpath.2008.061249] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Autoimmune alopecia (alopecia areata) is considered to be triggered by a collapse of immune privilege in hair follicles. Here we confirmed that infiltrating CD4 T lymphocytes around hair follicles of patients with alopecia areata were primarily CCR5-positive with few CCR4-positive cells, suggesting a dominant role of Th1 cells in the alopecic lesion. Given this finding, we sought to elucidate the effect of cytokine therapy in C3H/HeJ mice, a mouse model of alopecia areata, by applying recombinant interleukin-4 and neutralizing anti-interferon-gamma antibody. We found that local injections of both interleukin-4 and neutralizing anti-interferon-gamma antibody effectively treated alopecia in C3H/HeJ mice. Results from immunohistochemistry and semiquantitative reverse transcription-polymerase chain reaction demonstrated that intralesional injection of interleukin-4 suppressed CD8 T cell infiltrates around the hair follicles and repressed enhanced interferon-gamma mRNA expression in the affected alopecic skin. Furthermore, Th1 transcription factor T-box21 small interfering RNAs conjugated to cationized gelatin showed mitigating effects on alopecia in C3H/HeJ mice, resulting in the restoration of hair shaft elongation. Taken together, the use of gelatin-small interfering RNA conjugates promises to be a novel, efficient, and safe tool as an alternative gene therapy for the treatment of various human diseases. To our knowledge, this is the first report of effective controlled delivery of small interfering RNA using biodegradable cationized gelatin microspheres in an animal model of disease.
Collapse
|
105
|
Abstract
The immune system has evolved to protect the host from microbial infection; nevertheless, a breakdown in the immune system often results in infection, cancer, and autoimmune diseases. Multiple sclerosis, rheumatoid arthritis, type 1 diabetes, inflammatory bowel disease, myocarditis, thyroiditis, uveitis, systemic lupus erythromatosis, and myasthenia gravis are organ-specific autoimmune diseases that afflict more than 5% of the population worldwide. Although the etiology is not known and a cure is still wanting, the use of herbal and dietary supplements is on the rise in patients with autoimmune diseases, mainly because they are effective, inexpensive, and relatively safe. Curcumin is a polyphenolic compound isolated from the rhizome of the plant Curcuma longa that has traditionally been used for pain and wound-healing. Recent studies have shown that curcumin ameliorates multiple sclerosis, rheumatoid arthritis, psoriasis, and inflammatory bowel disease in human or animal models. Curcumin inhibits these autoimmune diseases by regulating inflammatory cytokines such as IL-1beta, IL-6, IL-12, TNF-alpha and IFN-gamma and associated JAK-STAT, AP-1, and NF-kappaB signaling pathways in immune cells. Although the beneficial effects of nutraceuticals are traditionally achieved through dietary consumption at low levels for long periods of time, the use of purified active compounds such as curcumin at higher doses for therapeutic purposes needs extreme caution. A precise understanding of effective dose, safe regiment, and mechanism of action is required for the use of curcumin in the treatment of human autoimmune diseases.
Collapse
Affiliation(s)
- John J Bright
- Neuroscience Research Laboratory, Methodist Research Institute, Clarian Health, Indianapolis, IN 46202, USA.
| |
Collapse
|
106
|
McCracken SA, Hadfield K, Rahimi Z, Gallery ED, Morris JM. NF-kappaB-regulated suppression of T-bet in T cells represses Th1 immune responses in pregnancy. Eur J Immunol 2007; 37:1386-96. [PMID: 17407192 DOI: 10.1002/eji.200636322] [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] [Indexed: 01/17/2023]
Abstract
The molecular mechanisms that suppress Th1 immune responses in pregnancy are unknown. We assessed the expression of the Th1 cytokine transcription factor T-bet. We isolated PBMC and T cells from non-pregnant and pregnant women and demonstrated that T-bet is specifically down-regulated in pregnancy under basal and stimulated conditions. Low levels of T-bet protein were detected in the nuclear fraction of unstimulated PBMC from non-pregnant, but not pregnant women. Nuclear levels of T-bet increased in response to PMA/ionomycin in PBMC from non-pregnant, but not pregnant women. T-bet expression was greater in whole cell lysates of stimulated CD3(+) T cells from non-pregnant relative to pregnant women. NF-kappaB is specifically down-regulated in T cells in pregnant women, resulting in suppressed expression of Th1 cytokines IL-2, IFN-gamma and TNF-alpha. In this study, down-regulation of NF-kappaB also resulted in diminished expression of T-bet. PMA induces NF-kappaB translocation, T-bet expression and IL-2, IFN-gamma and TNF-alpha production. Conversely, pre-incubation with SN50, and NF-kappaB oligodeoxyribonucleotide decoys suppressed PMA-induced NF-kappaB translocation and gene transcription, respectively, resulting in diminished T-bet expression and Th1 cytokine production. Therefore, maintenance of the cytokine environment for pregnancy success is mediated via strict regulation of Th1 immune responses, more specifically through control of NF-kappaB and T-bet transcription.
Collapse
Affiliation(s)
- Sharon A McCracken
- Perinatal Research Group, Kolling Institute of Medical Research, University of Sydney, St Leonards, Australia.
| | | | | | | | | |
Collapse
|
107
|
Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 2007; 25:821-52. [PMID: 17201677 DOI: 10.1146/annurev.immunol.25.022106.141557] [Citation(s) in RCA: 1404] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Since its conception two decades ago, the Th1-Th2 paradigm has provided a framework for understanding T cell biology and the interplay of innate and adaptive immunity. Naive T cells differentiate into effector T cells with enhanced functional potential for orchestrating pathogen clearance largely under the guidance of cytokines produced by cells of the innate immune system that have been activated by recognition of those pathogens. This secondary education of post-thymic T cells provides a mechanism for appropriately matching adaptive immunity to frontline cues of the innate immune system. Owing in part to the rapid identification of novel cytokines of the IL-17 and IL-12 families using database searches, the factors that specify differentiation of a new effector T cell lineage-Th17-have now been identified, providing a new arm of adaptive immunity and presenting a unifying model that can explain many heretofore confusing aspects of immune regulation, immune pathogenesis, and host defense.
Collapse
Affiliation(s)
- Casey T Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | |
Collapse
|
108
|
Kleiter I, Pedré X, Mueller AM, Poeschl P, Couillard-Despres S, Spruss T, Bogdahn U, Giegerich G, Steinbrecher A. Inhibition of Smad7, a negative regulator of TGF-beta signaling, suppresses autoimmune encephalomyelitis. J Neuroimmunol 2007; 187:61-73. [PMID: 17553571 DOI: 10.1016/j.jneuroim.2007.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 04/12/2007] [Indexed: 02/07/2023]
Abstract
We studied the role of the Transforming growth factor (TGF)-beta signaling antagonist Smad7 in autoimmune central nervous system (CNS) inflammation by using specific antisense oligonucleotides (Smad7-as). Elevated Smad7 protein expression was found in the spinal cord of SJL/J mice and DA rats with experimental autoimmune encephalomyelitis (EAE) and in effector T cells upon antigen stimulation. Smad7-as specifically decreased Smad7 mRNA and protein in cell lines and in ex-vivo-treated primary mouse lymph node cells (LNC). LNC exposed to Smad7-as during secondary activation showed reduced proliferation and encephalitogenicity. After systemic administration, Smad7-as ameliorated clinical signs of active and adoptively transferred EAE, diminished CNS inflammation, and reduced Smad7 protein levels in the brain. Smad7-as was found to be incorporated by peritoneal macrophages as well as by cells of the liver, kidneys, and peripheral lymph nodes. Importantly, Smad7-as treatment was not toxic and did not increase extracellular matrix formation. Smad7 inhibition thus represents a novel systemic treatment strategy for autoimmune CNS inflammation, targeting TGF-beta signaling without TGF-beta-associated toxicity.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Female
- Gene Expression Regulation/drug effects
- Lymphocytes/drug effects
- Lymphocytes/metabolism
- Mice
- Mice, Inbred Strains
- Oligonucleotides, Antisense/pharmacology
- Oligonucleotides, Antisense/therapeutic use
- RNA, Messenger/biosynthesis
- Rats
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Smad7 Protein/chemistry
- Smad7 Protein/metabolism
- Thionucleotides/pharmacology
- Thionucleotides/therapeutic use
- Time Factors
- Transforming Growth Factor beta/physiology
Collapse
Affiliation(s)
- Ingo Kleiter
- Department of Neurology, University of Regensburg, Universitaetsstr. 84, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Abstract
CD4(+) effector T cells have been categorized into two subsets: T helper type 1 (T(H)1) and T(H)2. Another subset of T cells that produce interleukin 17 (IL-17; 'T(H)-17 cells') has been identified that is highly proinflammatory and induces severe autoimmunity. Whereas IL-23 serves to expand previously differentiated T(H)-17 cell populations, IL-6 and transforming growth factor-beta (TGF-beta) induce the differentiation of T(H)-17 cells from naive precursors. These data suggest a dichotomy between CD4(+) regulatory T cells positive for the transcription factor Foxp3 and T(H)-17 cells: TGF-beta induces Foxp3 and generates induced regulatory T cells, whereas IL-6 inhibits TGF-beta-driven Foxp3 expression and together with TGF-beta induces T(H)-17 cells. Emerging data regarding T(H)-17 cells suggest a very important function for this T cell subset in immunity and disease.
Collapse
Affiliation(s)
- Estelle Bettelli
- Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
110
|
Ni J, Shu YY, Zhu YN, Fu YF, Tang W, Zhong XG, Wang H, Yang YF, Ren J, Wang MW, Zuo JP. COX-2 inhibitors ameliorate experimental autoimmune encephalomyelitis through modulating IFN-γ and IL-10 production by inhibiting T-bet expression. J Neuroimmunol 2007; 186:94-103. [PMID: 17442406 DOI: 10.1016/j.jneuroim.2007.03.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 03/06/2007] [Accepted: 03/07/2007] [Indexed: 11/22/2022]
Abstract
The COX-2 inhibitors Rofecoxib (Rof) and Lumiracoxib (Lum) were evaluated in experimental autoimmune encephalomyelitis (EAE), the model of multiple sclerosis (MS). Administration of Rof and Lum significantly reduced the incidence and severity of EAE, which was associated with the inhibition of MOG 35-55 lymphocyte recall response, anti-MOG 35-55 T cell responses, and modulation of cytokines production. In vitro Rof and Lum inhibited primary T cells proliferation and modulated cytokine production. These findings highlight the fact that Rof and Lum likely prevents EAE by modulating Th1/Th2 response, and suggest its utility in the treatment of MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Jia Ni
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Abstract
RNA interference (RNAi) is a potent method of gene silencing that has developed rapidly over the past few years as a result of its extensive importance in the study of genetics, molecular biology and physiology. RNAi technology has also recently yielded significant insight into the innate and adaptive immune systems by helping to elucidate numerous mechanisms that regulate the development, activation and function of cells that mediate immunity. In addition, because of its ability to suppress gene expression effectively, this technique may be used to regulate the immune response for clinical purposes. Nonetheless, before RNAi can be successfully administered into human patients as a medical treatment, it is necessary to overcome several major limitations of this technology, such as inefficient in vivo delivery, incomplete silencing of target genes, non-specific immune responses, and off-target effects. As novel developments and discoveries in molecular biology swiftly continue to unfold, it is likely that RNAi may soon translate into a potent form of in vivo gene silencing with profound applications to vaccination and immunotherapy. In the present review, we examine the current progress of immunological studies employing RNAi and discuss the prospects for the implementation of this technique in the clinical arena.
Collapse
Affiliation(s)
- Chih-Ping Mao
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Yen-Yu Lin
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - T-C Wu
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Oncology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Obstetrics and Gynecology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
112
|
Gocke AR, Cravens PD, Ben LH, Hussain RZ, Northrop SC, Racke MK, Lovett-Racke AE. T-bet regulates the fate of Th1 and Th17 lymphocytes in autoimmunity. THE JOURNAL OF IMMUNOLOGY 2007; 178:1341-8. [PMID: 17237380 DOI: 10.4049/jimmunol.178.3.1341] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-17-producing T cells (Th17) have recently been implicated in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model for the human disease multiple sclerosis. However, little is known about the transcription factors that regulate these cells. Although it is clear that the transcription factor T-bet plays an essential role in the differentiation of IFN-gamma-producing CD4(+) Th1 lymphocytes, the potential role of T-bet in the differentiation of Th17 cells is not completely understood. In this study, therapeutic administration of a small interfering RNA specific for T-bet significantly improved the clinical course of established EAE. The improved clinical course was associated with suppression of newly differentiated T cells that express IL-17 in the CNS as well as suppression of myelin basic protein-specific Th1 autoreactive T cells. Moreover, T-bet was found to directly regulate transcription of the IL-23R, and, in doing so, influenced the fate of Th17 cells, which depend on optimal IL-23 production for survival. We now show for the first time that suppression of T-bet ameliorates EAE by limiting the differentiation of autoreactive Th1 cells, as well as inhibiting pathogenic Th17 cells via regulation of IL-23R.
Collapse
MESH Headings
- Animals
- Autoimmunity
- Cell Differentiation/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Gene Expression Regulation
- Humans
- Interleukin-17
- Interleukin-23/genetics
- Mice
- Mice, Transgenic
- Myelin Basic Protein
- Nerve Tissue Proteins/immunology
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/therapeutic use
- T-Box Domain Proteins/immunology
- T-Box Domain Proteins/physiology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Th1 Cells/immunology
- Transcription Factors/immunology
Collapse
Affiliation(s)
- Anne R Gocke
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | |
Collapse
|
113
|
Fu YF, Zhu YN, Ni J, Zhong XG, Tang W, Re YD, Shi LP, Wan J, Yang YF, Yuan C, Nan FJ, Lawson BR, Zuo JP. A reversible S-adenosyl-L-homocysteine hydrolase inhibitor ameliorates experimental autoimmune encephalomyelitis by inhibiting T cell activation. J Pharmacol Exp Ther 2006; 319:799-808. [PMID: 16914558 DOI: 10.1124/jpet.106.107185] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The reversible S-adenosyl-l-homocysteine hydrolase inhibitor DZ2002 [methyl 4-(adenin-9-yl)-2-hydroxybutanoate] suppresses antigen-induced-specific immune responses, particularly type 1 helper T cell (Th1)-type responses. Experimental autoimmune encephalomyelitis (EAE) is thought to be a Th1 cell-mediated inflammatory demyelinating autoimmune disease model of human multiple sclerosis (MS). In this study, we examined the effects of DZ2002 on active EAE induced by myelin oligodendrocyte glycoprotein (MOG) 35-55 in female C57BL/6 mice. Administration of DZ2002 (50 mg/kg/day i.p.) significantly reduced the incidence and severity of EAE, which was associated with the inhibition of MOG35-55-specific T cell proliferation and Th1-type cytokine production. In vitro studies also demonstrated that DZ2002 inhibited anti-CD3/28-induced naive T cell activation concomitant with the down-regulation of cyclin-dependent kinase (CDK) 4, CDK6, cyclin D3, and the up-regulation or protection of the CDK inhibitor p27. These findings highlight the fact that DZ2002 likely prevents EAE by suppressing T cell activation and suggest its utility in the treatment of MS and other Th1-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yun-Feng Fu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Nath N, Prasad R, Giri S, Singh AK, Singh I. T-bet is essential for the progression of experimental autoimmune encephalomyelitis. Immunology 2006; 118:384-91. [PMID: 16827899 PMCID: PMC1782298 DOI: 10.1111/j.1365-2567.2006.02385.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is mediated by myelin-specific CD4+ T helper 1 (Th1) cells, while recovery from the disease is associated with the presence of Th2 cells. Here we used animals with targeted deletion of the T-bet gene to determine its role in the progression of EAE. T-bet regulates the production of interferon-gamma (IFN-gamma) in CD4+ and natural killer cells, and CD4+ T cells from T-bet-deficient mice were unable to differentiate into a Th1 phenotype. Moreover BALB/c mice deficient in T-bet were resistant to the induction of EAE disease, with minimal inflammatory infiltrates in the central nervous system. These mice were resistant to EAE induction even when PLP(180-199) peptide specific effector T cells from BALB/c wild type were transferred to BALB/c T-bet-deficient mice. This resistance to EAE is may be caused by the production of the anti-inflammatory cytokine interleukin-10 (IL-10) from the spleen cells upon ex vivo stimulation with PLP(180-199) peptide and in vivo presence in the central nervous system. There was no difference in the recall responses in spleen cells from T-bet-deficient and wild type mice; however, less secretion of IFN-gamma was observed from primed splenocytes. The expression of IFN-gamma was less in the central nervous system of T-bet-deficient mice whereas IL-10 was significantly higher in T-bet-deficient as compared to wild type mice. These data indicate that T-bet genes play a critical role in maintaining the encephalitogenic nature of CD4+ T cells in autoimmune responses during EAE disease progression.
Collapse
Affiliation(s)
- Narender Nath
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
115
|
Butz NV, Gronostajski RM, Campbell CE. T-box proteins differentially activate the expression of the endogenous interferon γ gene versus transfected reporter genes in non-immune cells. Gene 2006; 377:130-9. [PMID: 16737784 DOI: 10.1016/j.gene.2006.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Revised: 03/17/2006] [Accepted: 03/25/2006] [Indexed: 02/02/2023]
Abstract
The T-box transcription factor T-bet is expressed in a number of hematopoetic cell types and plays an essential role in the lineage determination of Th1 T-helper cells. In the absence of T-bet, CD4(+) T-cells fail to induce IFNgamma, the cytokine whose expression characterizes Th1 cells. Here we show that, surprisingly, T-bet induces the expression of endogenous IFNgamma in non-immune human cells, including 293 and other cell lines. Thus T-bet can induce IFNgamma expression independently of its role in T-cell lineage determination. In addition, mutations in T-bet, and chimeras of T-bet with other transcription factors including the T-box transcription factor, TBX2, differentially affect the ability of T-bet to activate expression of endogenous IFNgamma versus a T-site regulated reporter gene. A truncated T-betVp16 fusion protein strongly activates the T-site reporter but fails to activate endogenous IFNgamma. Conversely, native T-bet strongly activates endogenous IFNgamma expression but only weakly activates the reporter gene. Fusion of the Vp16 activation domain to full-length T-bet greatly increases its activation of both endogenous IFNgamma and transfected T-site reporter gene expression. In contrast, TBX2Vp16 potently activates the T-site reporter but has a negligible effect on endogenous IFNgamma expression. Butyrate treatment of T-bet expressing cells potentiates the expression of endogenous IFNgamma but weakly represses expression of the T-site reporter gene. These data indicate that induction of endogenous IFNgamma can be uncoupled from differentiation into the Th1 lineage and that the expression of endogenous IFNgamma versus a T-site reporter gene is differentially regulated by T-bet and other T-box proteins.
Collapse
Affiliation(s)
- Nataliya V Butz
- Department of Biochemistry, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, NY 14214, USA
| | | | | |
Collapse
|
116
|
Rangachari M, Mauermann N, Marty RR, Dirnhofer S, Kurrer MO, Komnenovic V, Penninger JM, Eriksson U. T-bet negatively regulates autoimmune myocarditis by suppressing local production of interleukin 17. ACTA ACUST UNITED AC 2006; 203:2009-19. [PMID: 16880257 PMCID: PMC2118365 DOI: 10.1084/jem.20052222] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Experimental autoimmune myocarditis (EAM) appears after infectious heart disease, the most common cause of dilated cardiomyopathy in humans. Here we report that mice lacking T-bet, a T-box transcription factor required for T helper (Th)1 cell differentiation and interferon (IFN)-γ production, develop severe autoimmune heart disease compared to T-bet−/− control mice. Experiments in T-bet−/−IL-4−/− and T-bet−/− IL-4Rα−/− mice, as well as transfer of heart-specific Th1 and Th2 cell lines, showed that autoimmune heart disease develops independently of Th1 or Th2 polarization. Analysis of T-bet−/−IL-12Rβ1−/− and T-bet−/− IL-12p35−/− mice then identified interleukin (IL)-23 as critical for EAM pathogenesis. In addition, T-bet−/− mice showed a marked increase in production of the IL-23–dependent cytokine IL-17 by heart-infiltrating lymphocytes, and in vivo IL-17 depletion markedly reduced EAM severity in T-bet−/− mice. Heart-infiltrating T-bet−/− CD8+ but not CD8− T cells secrete IFN-γ, which inhibits IL-17 production and protects against severe EAM. In contrast, T-bet−/− CD8+ lymphocytes completely lost their capacity to release IFN-γ within the heart. Collectively, these data show that severe IL-17–mediated EAM can develop in the absence of T-bet, and that T-bet can regulate autoimmunity via the control of nonspecific CD8+ T cell bystander functions in the inflamed target organ.
Collapse
Affiliation(s)
- Manu Rangachari
- Institute for Molecular Biotechnology of the Austrian Academy of Sciences, A-1030 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
117
|
Muthian G, Raikwar HP, Rajasingh J, Bright JJ. 1,25 Dihydroxyvitamin-D3 modulates JAK-STAT pathway in IL-12/IFNgamma axis leading to Th1 response in experimental allergic encephalomyelitis. J Neurosci Res 2006; 83:1299-309. [PMID: 16547967 DOI: 10.1002/jnr.20826] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Experimental allergic encephalomyelitis (EAE) is a Th1 cell-mediated autoimmune disease model of multiple sclerosis (MS). Vitamin D deficiency is commonly observed in MS patients and vitamin D supplements reduce the clinical symptoms of EAE and MS. Earlier studies have shown that in vivo treatment with vitamin D analogs ameliorates EAE in association with the inhibition of IL-12 production and Th1 differentiation. The mechanisms in the regulation of Th1 response by vitamin D in EAE/MS are, however, not known. We show that in vivo treatment of C57BL/6 and SJL/J mice (i.p.) with 100 ng of 1,25 dihydroxyvitamin D3, on every other day from Day 0-30, ameliorates EAE in association with the inhibition of IL-12 production and neural antigen-specific Th1 response. In vitro treatment with 1,25(OH)2D3 inhibited IFNgamma-induced tyrosine phosphorylation of STAT1, without affecting JAK2, in EOC-20 microglial cells. Treatment of activated T cells with 1,25(OH)2D3 also inhibited the IL-12-induced tyrosine phosphorylation of JAK2, TYK2, STAT3, and STAT4 in association with a decrease in T cell proliferation in vitro. These findings highlight the fact that vitamin D modulates JAK-STAT signaling pathway in IL-12/IFNgamma axis leading to Th1 differentiation and further suggest its use in the treatment of MS and other Th1 cell-mediated autoimmune diseases.
Collapse
MESH Headings
- Animals
- Calcitriol/metabolism
- Calcitriol/pharmacology
- Cells, Cultured
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-12/immunology
- Interleukin-12/metabolism
- Janus Kinase 2
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Phosphorylation/drug effects
- Protein-Tyrosine Kinases/immunology
- Protein-Tyrosine Kinases/metabolism
- Proto-Oncogene Proteins/immunology
- Proto-Oncogene Proteins/metabolism
- STAT1 Transcription Factor/immunology
- STAT1 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Gladson Muthian
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | |
Collapse
|
118
|
Page TJ, Sikder D, Yang L, Pluta L, Wolfinger RD, Kodadek T, Thomas RS. Genome-wide analysis of human HSF1 signaling reveals a transcriptional program linked to cellular adaptation and survival. MOLECULAR BIOSYSTEMS 2006; 2:627-39. [PMID: 17216044 DOI: 10.1039/b606129j] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Although HSF1 plays an important role in the cellular response to proteotoxic stressors, little is known about the structure and function of the human HSF1 signaling network under both stressed and unstressed conditions. In this study, we used a combination of chromatin immunoprecipitation microarray analysis and time course gene expression microarray analysis with and without siRNA-mediated inhibition of HSF1 to comprehensively identify genes regulated directly and indirectly by HSF1. The correlation between promoter binding and gene expression was not significant for all genes bound by HSF1, suggesting that HSF1 binding per se is not sufficient for expression. However, the correlation with promoter binding was significant for genes identified as HSF1-regulated following siRNA knockdown. Among promoters bound by HSF1 following heat shock, a gene ontology analysis showed significant enrichment only in categories related to protein folding. In contrast, analysis of the extended HSF1 signaling network following siRNA knockdown showed enrichment in a variety of categories related to protein folding, anti-apoptosis, RNA splicing, ubiquitinylation and others, highlighting a complex transcriptional program regulated directly and indirectly by HSF1.
Collapse
Affiliation(s)
- Todd J Page
- CIIT Centers for Health Research, 6 Davis Drive, Research Triangle Park, NC 27709-2137, USA
| | | | | | | | | | | | | |
Collapse
|
119
|
Chen Y, Langrish CL, Mckenzie B, Joyce-Shaikh B, Stumhofer JS, McClanahan T, Blumenschein W, Churakovsa T, Low J, Presta L, Hunter CA, Kastelein RA, Cua DJ. Anti-IL-23 therapy inhibits multiple inflammatory pathways and ameliorates autoimmune encephalomyelitis. J Clin Invest 2006; 116:1317-26. [PMID: 16670771 PMCID: PMC1450386 DOI: 10.1172/jci25308] [Citation(s) in RCA: 447] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 01/17/2006] [Indexed: 12/18/2022] Open
Abstract
IL-23 is a member of the IL-12 cytokine family that drives a highly pathogenic T cell population involved in the initiation of autoimmune diseases. We have shown that IL-23-dependent, pathogenic T cells produced IL-17 A, IL-17 F, IL-6, and TNF but not IFN-gamma or IL-4. We now show that T-bet and STAT1 transcription factors are not required for the initial production of IL-17. However, optimal IL-17 production in response to IL-23 stimulation appears to require the presence of T-bet. To explore the clinical efficacy of targeting the IL-23 immune pathway, we generated anti-IL-23p19-specific antibodies and tested to determine whether blocking IL-23 function can inhibit EAE, a preclinical animal model of human multiple sclerosis. Anti-IL-23p19 treatment reduced the serum level of IL-17 as well as CNS expression of IFN-gamma, IP-10, IL-17, IL-6, and TNF mRNA. In addition, therapeutic treatment with anti-IL-23p19 during active disease inhibited proteolipid protein (PLP) epitope spreading and prevented subsequent disease relapse. Thus, therapeutic targeting of IL-23 effectively inhibited multiple inflammatory pathways that are critical for driving CNS autoimmune inflammation.
Collapse
Affiliation(s)
- Yi Chen
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Claire L. Langrish
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Brent Mckenzie
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Barbara Joyce-Shaikh
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Jason S. Stumhofer
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Terrill McClanahan
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Wendy Blumenschein
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Tatyana Churakovsa
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Justin Low
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Leonard Presta
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Christopher A. Hunter
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Robert A. Kastelein
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| | - Daniel J. Cua
- Discovery Research, Schering-Plough Biopharma, Palo Alto, California, USA.
Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Experimental Pathology and Pharmacology and
Protein Engineering, Schering-Plough Biopharma, Palo Alto, California, USA
| |
Collapse
|
120
|
Morris K, Castanotto D, Al-Kadhimi Z, Jensen M, Rossi J, Cooper LJN. Enhancing siRNA effects in T cells for adoptive immunotherapy. ACTA ACUST UNITED AC 2006; 10:461-7. [PMID: 16321811 DOI: 10.1080/10245330500233569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetically manipulated T cells can be endowed with novel functions to obtain desired in vivo effects after adoptive transfer. This genetic approach is being used to introduce genes such as chimeric immunoreceptors and tumor-specific T cells are being evaluated in early phase clinic trials. However, the ability to alter the genetic programming of T cells also presents opportunities to remove unwanted T-cell functions in order to augment an anti-tumor effect or endow resistance such as to HIV infection. Specifically, the use of RNA interference (RNAi) to disrupt gene expression by targeting either the mRNA or the promoter, provides investigators with many new opportunities to genetically modify T cells that should prove useful in future applications of adoptive immunotherapy.
Collapse
Affiliation(s)
- Kevin Morris
- Division of Molecular Biology, Beckman Research Institute and City of Hope National Medical Center, Duarte, CA 90010-3000, USA
| | | | | | | | | | | |
Collapse
|
121
|
Fu YF, Zhu YN, Ni J, Zhong XG, Tang W, Zhou R, Zhou Y, Dong JR, He PL, Wan H, Li YC, Yang YF, Zuo JP. (5R)-5-Hydroxytriptolide (LLDT-8), a novel triptolide derivative, prevents experimental autoimmune encephalomyelitis via inhibiting T cell activation. J Neuroimmunol 2006; 175:142-51. [PMID: 16712960 DOI: 10.1016/j.jneuroim.2006.03.011] [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] [Received: 01/18/2006] [Revised: 03/14/2006] [Accepted: 03/20/2006] [Indexed: 11/16/2022]
Abstract
A novel triptolide derivative (5R)-5-hydroxytriptolide (LLDT-8) has been shown to have potent immunosuppressive activities. Here LLDT-8 was evaluated in experimental autoimmune encephalomyelitis (EAE), the model of multiple sclerosis (MS). LLDT-8 reduced the incidence and severity of EAE, which was associated with the inhibition of the MOG 35-55 lymphocyte recall response, anti-MOG 35-55 T cell responses, interleukin (IL)-2 and interferon (IFN)-gamma production. In vitro, LLDT-8 inhibited primary T cells proliferation, division, IL-2 and IFN-gamma production stimulated with anti-CD3/28. These findings highlight the fact that LLDT-8 prevents EAE by suppressing T cell proliferation and activation, with a potential for treatment of MS.
Collapse
Affiliation(s)
- Yun-Feng Fu
- Laboratories of Immunopharmacology and Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Skundric DS, Cai J, Cruikshank WW, Gveric D. Production of IL-16 correlates with CD4+ Th1 inflammation and phosphorylation of axonal cytoskeleton in multiple sclerosis lesions. J Neuroinflammation 2006; 3:13. [PMID: 16729885 PMCID: PMC1488832 DOI: 10.1186/1742-2094-3-13] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2006] [Accepted: 05/26/2006] [Indexed: 12/15/2022] Open
Abstract
Background Multiple sclerosis (MS) is a central nervous system-specific autoimmune, demyelinating and neurodegenerative disease. Infiltration of lesions by autoaggressive, myelin-specific CD4+Th1 cells correlates with clinical manifestations of disease. The cytokine IL-16 is a CD4+ T cell-specific chemoattractant that is biased towards CD4+ Th1 cells. IL-16 precursor is constitutively expressed in lymphocytes and during CD4+ T cell activation; active caspase-3 cleaves and releases C-terminal bioactive IL-16. Previously, we used an animal model of MS to demonstrate an important role for IL-16 in regulation of autoimmune inflammation and subsequent axonal damage. This role of IL-16 in MS is largely unexplored. Here we examine the regulation of IL-16 in relation to CD4+ Th1 infiltration and inflammation-related changes of axonal cytoskeleton in MS lesions. Methods We measured relative levels of IL-16, active caspase-3, T-bet, Stat-1 (Tyr 701), and phosphorylated NF(M+H), in brain and spinal cord lesions from MS autopsies, using western blot analysis. We examined samples from 39 MS cases, which included acute, subacute and chronic lesions, as well as adjacent, normal-appearing white and grey matter. All samples were taken from patients with relapsing remitting clinical disease. We employed two-color immunostaining and confocal microscopy to identify phenotypes of IL-16-containing cells in frozen tissue sections from MS lesions. Results We found markedly increased levels of pro- and secreted IL-16 (80 kD and 22 kD, respectively) in MS lesions compared to controls. Levels of IL-16 peaked in acute, diminished in subacute, and were elevated again in chronic active lesions. Compared to lesions, lower but still appreciable IL-6 levels were measured in normal-appearing white matter adjacent to active lesions. Levels of IL-16 corresponded to increases in active-caspase-3, T-bet and phosphorylated Stat-1. In MS lesions, we readily observed IL-16 immunoreactivity confined to infiltrating CD3+, T-bet+ and active caspase-3+ mononuclear cells. Conclusion We present evidence suggesting that IL-16 production occurs in MS lesions. We show correlations between increased levels of secreted IL-16, CD4+ Th1 cell inflammation, and phosphorylation of axonal cytoskeleton in MS lesions. Overall, the data suggest a possible role for IL-16 in regulation of inflammation and of subsequent changes in the axonal cytoskeleton in MS.
Collapse
Affiliation(s)
- Dusanka S Skundric
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Juan Cai
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | - Djordje Gveric
- Department of Neuroinflammation, Institute of Neurology, University College London WC1N 1PJ, UK
| |
Collapse
|
123
|
Wang G, Liu CT, Wang ZL, Jiang LL, Yan CL, Luo FM. Antisense oligonucleotides-induced local blockade of T-bet expression leads to airway inflammation in rats. Acta Pharmacol Sin 2006; 27:561-7. [PMID: 16626511 DOI: 10.1111/j.1745-7254.2006.00323.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIM To explore whether local blockade of T-box expressed in T cells (T-bet) expression in the lungs could lead to airway inflammation. METHODS Twenty-four rats were randomly divided into 4 groups: saline group, ovalbumin (OVA)-sensitized group, nonsense group, and the antisense group. The OVA-sensitized rats were sensitized and challenged with OVA, and the rats in the nonsense and antisense groups were subjected to an aerosol delivery of the nonsense and antisense oligonucleotides (AS-ODN) of T-bet (0.1%, w/v). The levels of interferon-gamma (IFN-gamma), interleukin-4 (IL-4), and IL-5 in the bronchoalveolar lavage fluid (BALF) were detected by ELISA, and the mRNA and the protein expression of T-bet and GATA-3 genes were examined by in situ hybridization and Western blot analysis, respectively. RESULTS The expression of T-bet mRNA and protein in the lungs of the rats in the antisense group were inhibited effectively. The lungs of the rats in the antisense and OVA-sensitized groups showed eosinophil and lymphocyte inflammatory infiltration, and eosinophilia located predominantly around the airways. The number of GATA-3 mRNA-positive cells and the level of GATA-3 protein in the lungs of the rats in the antisense and the OVA-sensitized groups significantly increased. The level of IL-4 and IL-5 in the BALF in the antisense and OVA-sensitized groups were elevated, but the level of IFN-gamma decreased markedly. CONCLUSION Antisense ODN-induced local blockade of T-bet expression leads to airway inflammation with a selective alteration in patterns of cytokine expression and recruitment of eosinophil cells similar to that in the OVA-sensitized animals.
Collapse
Affiliation(s)
- Gang Wang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | |
Collapse
|
124
|
Harrington LE, Mangan PR, Weaver CT. Expanding the effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol 2006; 18:349-56. [PMID: 16616472 DOI: 10.1016/j.coi.2006.03.017] [Citation(s) in RCA: 429] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Accepted: 03/28/2006] [Indexed: 12/13/2022]
Abstract
The Th1/Th2 paradigm has provided the framework for understanding CD4 T-cell biology and the interplay between innate and adaptive immunity for almost two decades. Recent studies have defined a previously unknown arm of the CD4 T-cell effector response--the Th17 lineage--that promises to change our understanding of immune regulation, immune pathogenesis and host defense. The factors that specify differentiation of IL-17-producing effector T-cells from naïve T-cell precursors are being rapidly discovered and are providing insights into mechanisms by which signals from cells of the innate immune system guide alternative pathways of Th1, Th2 or Th17 development.
Collapse
Affiliation(s)
- Laurie E Harrington
- Department of Pathology, University of Alabama at Birmingham, 845 19th Street South, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
125
|
Behlke MA. Progress towards in vivo use of siRNAs. Mol Ther 2006; 13:644-70. [PMID: 16481219 PMCID: PMC7106286 DOI: 10.1016/j.ymthe.2006.01.001] [Citation(s) in RCA: 375] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 01/11/2006] [Accepted: 01/11/2006] [Indexed: 01/28/2023] Open
Abstract
RNA interference (RNAi) has become the method of choice to suppress gene expression in vitro. It is also emerging as a powerful tool for in vivo research with over 90 studies published using synthetic small interfering RNAs in mammals. These reports demonstrate the potential for use of synthetic small interfering RNAs (siRNAs) as therapeutic agents, especially in the areas of cancer and viral infection. The number of reports using siRNAs for functional genomics applications, for validation of targets for small-molecule drug development programs, and to address questions of basic biology will rapidly grow as methods and protocols for use in animals become more established. This review will first discuss aspects of RNAi biochemistry and biology that impact in vivo use, especially as relates to experimental design, and will then provide an overview of published work with a focus on methodology.
Collapse
Affiliation(s)
- Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA 52241, USA.
| |
Collapse
|
126
|
Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006; 24:179-89. [PMID: 16473830 DOI: 10.1016/j.immuni.2006.01.001] [Citation(s) in RCA: 2947] [Impact Index Per Article: 155.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2005] [Revised: 11/28/2005] [Accepted: 01/04/2006] [Indexed: 11/27/2022]
Abstract
We describe de novo generation of IL-17-producing T cells from naive CD4 T cells, induced in cocultures of naive CD4 T cells and naturally occurring CD4+ CD25+ T cells (Treg) in the presence of TLR3, TLR4, or TLR9 stimuli. Treg can be substituted by TGFbeta1, which, together with the proinflammatory cytokine IL-6, supports the differentiation of IL-17-producing T cells, a process that is amplified by IL-1beta and TNFalpha. We could not detect a role for IL-23 in the differentiation of IL-17-producing T cells but confirmed its importance for their survival and expansion. Transcription factors GATA-3 and T-bet, as well as its target Hlx, are absent in IL-17-producing T cells, and they do not express the negative regulator for TGFbeta signaling, Smad7. Our data indicate that, in the presence of IL-6, TGFbeta1 subverts Th1 and Th2 differentiation for the generation of IL-17-producing T cells.
Collapse
Affiliation(s)
- Marc Veldhoen
- Division of Molecular Immunology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, United Kingdom
| | | | | | | | | |
Collapse
|
127
|
Affiliation(s)
- Elliot M Frohman
- Department of Neurology and Ophthalmology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| | | | | |
Collapse
|
128
|
Racke MK, Gocke AR, Muir M, Diab A, Drew PD, Lovett-Racke AE. Nuclear receptors and autoimmune disease: the potential of PPAR agonists to treat multiple sclerosis. J Nutr 2006; 136:700-3. [PMID: 16484546 PMCID: PMC2819754 DOI: 10.1093/jn/136.3.700] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a T-cell-mediated, autoimmune disorder characterized by central nervous system inflammation and demyelination, features reminiscent of the human disease, multiple sclerosis (MS). Prior work in the EAE model has suggested that encephalitogenic T cells are of the T helper (Th)-1 phenotype. Our group has performed several studies in the EAE model that suggest that a strategy for treating autoimmune disorders is to convert the pathogenic cells from the Th1 to Th2 phenotype. Peroxisome proliferator-activated receptors (PPARs) are members of a nuclear hormone receptor superfamily that include receptors for steroids, retinoids, and thyroid hormone, all of which are known to affect the immune response. Recently, we examined the role of PPARgamma in EAE and observed that administration of the PPARgamma agonist 15-deoxy-Delta(12,14) prostaglandin J2 exerted a significant therapeutic effect predominantly by inhibiting the activation and expansion of encephalitogenic T cells. One potential advantage in studying PPARalpha agonists is that they have been very well tolerated when used in humans to treat conditions such as elevated triglycerides. Building on prior work in immune deviation and with PPAR agonists, we have demonstrated that PPARalpha agonists can alter the cytokine phenotype of myelin-reactive T cells, alter their encephalitogenicity, and be useful in the treatment of EAE. The fact that PPARalpha agonists have been used as therapeutic agents in humans to treat metabolic conditions for over 25 years with little toxicity makes them attractive candidates for use as adjunctive therapies in MS.
Collapse
Affiliation(s)
- Michael K Racke
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, 75390, USA.
| | | | | | | | | | | |
Collapse
|
129
|
Beima KM, Miazgowicz MM, Lewis MD, Yan PS, Huang THM, Weinmann AS. T-bet binding to newly identified target gene promoters is cell type-independent but results in variable context-dependent functional effects. J Biol Chem 2006; 281:11992-2000. [PMID: 16473879 DOI: 10.1074/jbc.m513613200] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recently developed target gene identification strategies based upon the chromatin immunoprecipitation assay provide a powerful method to determine the localization of transcription factor binding within mammalian genomes. However, in many cases, it is unclear if the binding capacity of a transcription factor correlates with an obligate role in gene regulation in diverse contexts. It is therefore important to carefully examine the relationship between transcription factor binding and its ability to functionally regulate gene expression. T-bet is a T-box transcription factor expressed in several hematopoietic cell types. By utilizing a chromatin immunoprecipitation assay coupled to genomic microarray technology approach, we identified numerous promoters, including CXCR3, IL2Rbeta, and CCL3, that are bound by T-bet in B cells. Most surprisingly, the ability of T-bet to associate with the target promoters is not dependent upon the cell type background. Several of the promoters appear to be functionally regulated by T-bet. However, we could not detect a functional consequence for T-bet association with many of the identified promoters in overexpression studies or an examination of wild type and T-bet-/- primary B, CD4+, and CD8+ T cells. Thus, there is a high variability in the functional consequences, if any, that result from the association of T-bet with individual target promoters.
Collapse
Affiliation(s)
- Kristin M Beima
- Department of Immunology, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | |
Collapse
|
130
|
Broderick L, Brooks SP, Takita H, Baer AN, Bernstein JM, Bankert RB. IL-12 reverses anergy to T cell receptor triggering in human lung tumor-associated memory T cells. Clin Immunol 2006; 118:159-69. [PMID: 16271513 DOI: 10.1016/j.clim.2005.09.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Revised: 09/08/2005] [Accepted: 09/14/2005] [Indexed: 02/02/2023]
Abstract
Memory T cells in human non-small cell lung cancer are unresponsive to progressing tumors. T cells were evaluated at the single cell level by imaging the nuclear translocation of NF-kappaB and NFAT via immunofluorescence confocal microscopy as an early measure of responsiveness to T cell receptor triggering. Little translocation of NF-kappaB or NFAT occurred in tumor-associated T cells in response to CD3+CD28 cross-linking under conditions which led to maximal translocation in normal donor peripheral blood T cells. TNF-alpha induced maximal NF-kappaB translocation in these T cells, indicating that they remain receptive to alternative signaling pathways, and pulsing with IL-12 prior to TCR triggering reversed their apparent anergy. T cells from additional chronic inflammatory microenvironments demonstrated a similar refractoriness to TCR activation, suggesting either that a common regulatory mechanism present within the microenvironment controls these cells or that with continuous antigen exposure, they remain refractory to activation via the TCR.
Collapse
Affiliation(s)
- Lori Broderick
- Department of Microbiology and Immunology, 138 Farber Hall, State University of New York at Buffalo, 3435 Main Street, Buffalo, NY 14214, USA
| | | | | | | | | | | |
Collapse
|
131
|
Long M, Slaiby AM, Hagymasi AT, Mihalyo MA, Lichtler AC, Reiner SL, Adler AJ. T-bet down-modulation in tolerized Th1 effector CD4 cells confers a TCR-distal signaling defect that selectively impairs IFN-gamma expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 176:1036-45. [PMID: 16393991 PMCID: PMC2846362 DOI: 10.4049/jimmunol.176.2.1036] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
When Th1 effector CD4 cells encounter tolerizing Ag in vivo, their capacity to express the effector cytokines IFN-gamma and TNF-alpha is lost more rapidly than noneffector functions such as IL-2 production and proliferation. To localize the relevant intracellular signaling defects, cytokine expression was compared following restimulation with Ag vs agents that bypass TCR-proximal signaling. IFN-gamma and TNF-alpha expression were both partially rescued when TCR-proximal signaling was bypassed, indicating that both TCR-proximal and -distal signaling defects impair the expression of these two effector cytokines. In contrast, bypassing TCR-proximal signaling fully rescued IL-2 expression. T-bet, a transcription and chromatin remodeling factor that is required to direct the differentiation of naive CD4 cells into IFN-gamma-expressing Th1 effectors, was partially down-modulated in tolerized Th1 effectors. Enforcing T-bet expression during tolerization selectively rescued the ability to express IFN-gamma, but not TNF-alpha. Conversely, expression of a dominant-negative T-bet in Th1 effectors selectively impaired the ability to express IFN-gamma, but not TNF-alpha. Analysis of histone acetylation at the IFN-gamma promoter further suggested that down-modulation of T-bet expression during Th1 effector CD4 cell tolerization does not impair IFN-gamma expression potential through alterations in chromatin structure.
Collapse
Affiliation(s)
- Meixiao Long
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Aaron M. Slaiby
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Adam T. Hagymasi
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Marianne A. Mihalyo
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Alexander C. Lichtler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Steven L. Reiner
- Abramson Family Cancer Research Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam J. Adler
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
132
|
Ichim TE, Popov IA, Riordan NH, Izadi H, Zhong Z, Yijian L, Sher S, Oleinik EK. A novel method of modifying immune responses by vaccination with lipiodol-siRNA mixtures. J Transl Med 2006; 4:2. [PMID: 16390542 PMCID: PMC1343589 DOI: 10.1186/1479-5876-4-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 01/03/2006] [Indexed: 01/15/2023] Open
Abstract
The dendritic cell (DC) possesses the ability to stimulate both T helper 1 (Th1) and Th2 responses depending on activation stimuli. Although it is known that chemically or genetically modified DC can be used therapeutically to steer immune responses towards either Th1 or Th2, cellular therapy with ex vivo manipulated DC is clinically difficult. Here we demonstrate a novel method of switching immune responses from Th1 to Th2 through in vivo immune modulation by administration of siRNA. We demonstrate that siRNA targeting of the IL-12p35 gene leads to a Th2 bias in vitro through an IL-10 dependent mechanism. In vivo administration of siRNA admixed with the oil-based contrast agent lipiodol in the presence of antigen and adjuvant induced a deviation in recall response to reduced production of IFN-gamma and augmented IL-4 response using either KLH or ovalbumin. This simple method of in vivo modification of immune response possesses therapeutic potential in Th1-mediated diseases such as multiple sclerosis and autoimmune diabetes.
Collapse
Affiliation(s)
| | - Igor A Popov
- Department of Surgery, University of Western Ontario, London, Ontario, Canada
| | | | - Hamid Izadi
- Medistem Laboratories Inc, Tempe Arizona, USA
| | - Zaohui Zhong
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Yijian
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Salman Sher
- Division of Cardiology, Emory University, Atlanta, USA
| | - Eugenia K Oleinik
- Institute of Biology, Karelian Research Center, Russian Academy of Sciences, Petrozavodsk, Russia
| |
Collapse
|
133
|
Lin YY, Hung CF, Wu TC. Functional Studies of Lymphocytes Using RNAi Technology. Transfus Med Hemother 2006. [DOI: 10.1159/000090204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
134
|
Frisullo G, Angelucci F, Caggiula M, Nociti V, Iorio R, Patanella AK, Sancricca C, Mirabella M, Tonali PA, Batocchi AP. pSTAT1, pSTAT3, and T-bet expression in peripheral blood mononuclear cells from relapsing-remitting multiple sclerosis patients correlates with disease activity. J Neurosci Res 2006; 84:1027-36. [PMID: 16865709 DOI: 10.1002/jnr.20995] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, and it is considered to be a T helper 1 (Th1) cell-mediated autoimmune disease. T-bet has been identified as a key transcription factor for the development of Th1 cells and the induction of interferon (IFN)-gamma production. T-bet is induced during T-cell activation by the IFN-gamma signal transducer and activator of transcription (STAT)-1 signalling pathway. In this study we found an up-regulation of T-bet and pSTAT1 in peripheral blood CD4+ and CD8+ T cells and monocytes from relapsing-remitting MS patients in relapse compared with patients in remission and with healthy subjects. The increased expression of pSTAT1 strongly correlated with T-bet expression in CD4+ and CD8+ cells and monocytes from patients in relapse and was associated with an increased production of IFN-gamma by peripheral blood mononuclear cells (PBMCs). pSTAT3 was also up-regulated in CD4+ and CD8+ cells and monocytes from patients in relapse and was associated with an increased production of interleukin (IL)-10 but not of IL-6. pSTAT1, pSTAT3, and T-bet expression strongly correlated with Gd-DTPA-enhanced lesions on brain and spinal cord magnetic resonance imaging. Our data show for the first time that there is an up-regulation of type 1 immunity-correlated transcription factors such as STAT1 and T-bet in peripheral blood subpopulations of MS patients in the active phase of disease. The evaluation of T-bet and pSTAT1 expression in peripheral blood CD4+, CD8+ T cells and monocytes could be used as a marker of disease activity in relapsing-remitting MS.
Collapse
Affiliation(s)
- Giovanni Frisullo
- Institute of Neurology, Department of Neuroscience, Catholic University, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Ylikoski E, Lund R, Kyläniemi M, Filén S, Kilpeläinen M, Savolainen J, Lahesmaa R. IL-12 up-regulates T-bet independently of IFN-gamma in human CD4+ T cells. Eur J Immunol 2005; 35:3297-306. [PMID: 16220539 DOI: 10.1002/eji.200526101] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
T-bet is an important Th1 driving transcription factor regulated by IFN-gamma/STAT1 pathway. T-bet turns on IFN-gamma transcription in CD4+ T cells and T-bet-deficient cells fail to differentiate to Th1 direction. Previous reports have characterized function of T-bet mainly in murine cells and very little is known about its functions in human cells. Here, we studied T-bet expression kinetics in parallel with GATA3 during Th1/Th2 polarization. We demonstrate that in addition to CD3/CD28 activation, cytokines IL-12 and IFN-alpha in the presence of neutralizing anti-IFN-gamma enhanced T-bet mRNA and protein expression in human CD4+ cells. T-bet is known to be a potent inducer of IFN-gamma. Even though IFN-gamma and IL-12 stimulation induced similar levels of T-bet protein in human CD4+ cells, IFN-gamma-treated cells produced considerably less IFN-gamma than cells treated with IL-12. Therefore, high T-bet protein expression does not necessarily correlate with IFN-gamma production. In addition, we show that the immunosuppressive cytokine TGF-beta inhibits T-bet and GATA3 protein expression only if it is present prior to primary T cell activation and is maintained in the cultures during the early polarization of Th1/Th2 cells. In conclusion, we report new insights into the cytokine regulation of T-bet in human CD4+ T cells.
Collapse
Affiliation(s)
- Emmi Ylikoski
- Centre for Biotechnology, University of Turku and Abo Akademi University, Turku, Finland
| | | | | | | | | | | | | |
Collapse
|
136
|
McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23-IL-17 immune pathway. Trends Immunol 2005; 27:17-23. [PMID: 16290228 DOI: 10.1016/j.it.2005.10.003] [Citation(s) in RCA: 568] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 10/06/2005] [Accepted: 10/06/2005] [Indexed: 12/17/2022]
Abstract
Interleukin (IL)-23 is a heterodimeric cytokine closely related to IL-12. Yet, despite a strong structural relationship that includes a shared p40 subunit, this does not translate into functional similarity. In fact, the opposite is true, in that these two cytokines appear to have profoundly different roles in regulating host immune responses. It is now clear that IL-23 has key roles in autoimmune destruction in experimental allergic encephalomyelitis, collagen-induced arthritis and inflammatory bowel disease. IL-23 drives the development of autoreactive IL-17-producing T cells and promotes chronic inflammation dominated by IL-17, IL-6, IL-8 and tumor necrosis factor as well as neutrophils and monocytes. It is unlikely that IL-23 and its downstream effects evolved just to cause autoimmunity, but its real benefit to the host and the lineage relationship between IL-17-producing cells and T helper 1 cells remain unclear. By comparing the pathophysiological function of IL-12 and IL-23 in the context of host defense and autoimmune inflammation, we are beginning to understand the novel IL-23-IL-17 immune pathway.
Collapse
Affiliation(s)
- Brent S McKenzie
- Discovery Research, DNAX Research Inc., 901 California Ave, Palo Alto, CA 94304-1104, USA
| | | | | |
Collapse
|
137
|
Matsui M, Moriya O, Yoshimoto T, Akatsuka T. T-bet is required for protection against vaccinia virus infection. J Virol 2005; 79:12798-806. [PMID: 16188982 PMCID: PMC1235859 DOI: 10.1128/jvi.79.20.12798-12806.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The transcription factor T-bet regulates the differentiation of CD4(+) T-helper type 1 (Th1) cells and represses Th2 lineage commitment. Since Th1 cells are crucial in the defense against pathogens, several studies addressed the role of T-bet in immunity to infection using T-bet knockout (T-bet(-/-)) mice. Nevertheless, it is still unclear whether T-bet is required for defense. Although vaccinia virus (VV) has extensively been used as an expression vector and the smallpox vaccine, there is only limited knowledge about immunity to VV infection. The urgency to understand the immune responses has been increased because of concerns about bioterrorism. Here, we show that T-bet is critical in the defense against VV infection as follows: (i) the survival rate of T-bet(-/-) mice was lower than that of control littermates postinfection; (ii) T-bet(-/-) mice lost more weight postinfection; and (iii) control mice cleared VV faster than T-bet(-/-) mice. As expected, a significant Th2 shift was observed in CD4(+) T cells of T-bet(-/-) mice. Furthermore, absence of T-bet impaired VV-specific CD8(+) cytotoxic T-lymphocyte (CTL) function, including cytolytic activity, antiviral cytokine production, and proliferation. Cytolytic capacity of natural killer (NK) cells was also diminished in T-bet(-/-) mice, whereas anti-VV antibody production was not impaired. These data reveal that the enhanced susceptibility to VV infection in T-bet(-/-) mice was at least partially due to the Th2 shift of CD4(+) T cells and the diminished function of VV-specific CTLs and NK cells but not due to downregulation of antibody production.
Collapse
Affiliation(s)
- Masanori Matsui
- Department of Microbiology, Saitama Medical School, Moroyama-Cho, Iruma-Gun, Japan.
| | | | | | | |
Collapse
|
138
|
Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver CT. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005; 6:1123-32. [PMID: 16200070 DOI: 10.1038/ni1254] [Citation(s) in RCA: 3583] [Impact Index Per Article: 179.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Accepted: 08/09/2005] [Indexed: 11/09/2022]
Abstract
CD4(+) T cells producing interleukin 17 (IL-17) are associated with autoimmunity, although the precise mechanisms that control their development are undefined. Here we present data that challenge the idea of a shared developmental pathway with T helper type 1 (T(H)1) or T(H)2 lineages and instead favor the idea of a distinct effector lineage we call 'T(H)-17'. The development of T(H)-17 cells from naive precursor cells was potently inhibited by interferon-gamma (IFN-gamma) and IL-4, whereas committed T(H)-17 cells were resistant to suppression by T(H)1 or T(H)2 cytokines. In the absence of IFN-gamma and IL-4, IL-23 induced naive precursor cells to differentiate into T(H)-17 cells independently of the transcription factors STAT1, T-bet, STAT4 and STAT6. These findings provide a basis for understanding how inhibition of IFN-gamma signaling enhances development of pathogenic T(H)-17 effector cells that can exacerbate autoimmunity.
Collapse
Affiliation(s)
- Laurie E Harrington
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | |
Collapse
|
139
|
Yamashita M, Shinnakasu R, Asou H, Kimura M, Hasegawa A, Hashimoto K, Hatano N, Ogata M, Nakayama T. Ras-ERK MAPK cascade regulates GATA3 stability and Th2 differentiation through ubiquitin-proteasome pathway. J Biol Chem 2005; 280:29409-19. [PMID: 15975924 DOI: 10.1074/jbc.m502333200] [Citation(s) in RCA: 130] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Differentiation of naive CD4 T cells into Th2 cells requires protein expression of GATA3. Interleukin-4 induces STAT6 activation and subsequent GATA3 transcription. Little is known, however, on how T cell receptor-mediated signaling regulates GATA3 and Th2 cell differentiation. Here we demonstrated that T cell receptor-mediated activation of the Ras-ERK MAPK cascade stabilizes GATA3 protein in developing Th2 cells through the inhibition of the ubiquitin-proteasome pathway. Mdm2 was associated with GATA3 and induced ubiquitination on GATA3, suggesting its role as a ubiquitin-protein isopeptide ligase for GATA3 ubiquitination. Thus, the Ras-ERK MAPK cascade controls GATA3 protein stability by a post-transcriptional mechanism and facilitates GATA3-mediated chromatin remodeling at Th2 cytokine gene loci leading to successful Th2 cell differentiation.
Collapse
Affiliation(s)
- Masakatsu Yamashita
- Department of Immunology, Graduate School of Medicine, Chiba University, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
PURPOSE OF REVIEW The aim of this article is to describe recent observations regarding the basis for the initiation and disease evolution of multiple sclerosis. RECENT FINDINGS A current debate is where and what initiates the neuroinflammatory reaction that characterizes the acute multiple sclerosis lesion. Immune sensitization to neural antigens could develop within the systemic compartment consequent to exposure to cross-reacting, possibly viral derived, peptides (molecular mimicry). Although CD4 T cells are considered central to initiating central nervous system inflammation, the actual extent and specificity of tissue injury reflects the array of adaptive (CD8 T cells and antibody) and innate (microglia/macrophages) immune constituents present in the lesions. Neuropathologic studies indicate that lethal changes in neural cells (oligodendrocytes) could also be the initiating event, reflecting as yet unidentified acquired insults (e.g. exogenous virus or reactivated endogenous retrovirus) or intrinsic abnormalities ('neurodegenerative' hypothesis). Recurrence or persistence of the disease process can reflect events occurring at multiple sites including expansion of the immune repertoire in response to neural antigens transported to regional lymph nodes (determinant spreading), especially if immune regulatory mechanisms are defective; alterations in blood-brain barrier properties consequent to initial cellular transmigration; and participation of endogenous (microglia, astrocytes) or long lived infiltrating cells (macrophages, B cells in ectopic germinal centers) in regulating and effecting immune functions within the central nervous system. Accumulating neurologic deficit reflects the balance between injury and repair; the latter also being negatively or positively (trophic support and clearance of tissue debris) impacted by inflammatory processes. SUMMARY Understanding the full spectrum of multiple sclerosis presents a continuing challenge for both immunology and neurobiology.
Collapse
Affiliation(s)
- Alexandre Prat
- Neuroimmunology Laboratory and Multiple Sclerosis Clinic, CHUM Notre-Dame Hospital, Montreal, Quebec, Canada
| | | |
Collapse
|
141
|
Minter LM, Turley DM, Das P, Shin HM, Joshi I, Lawlor RG, Cho OH, Palaga T, Gottipati S, Telfer JC, Kostura L, Fauq AH, Simpson K, Such KA, Miele L, Golde TE, Miller SD, Osborne BA. Inhibitors of γ-secretase block in vivo and in vitro T helper type 1 polarization by preventing Notch upregulation of Tbx21. Nat Immunol 2005. [DOI: 10.1038/ni1209x] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
142
|
Abstract
The size of the peripheral T cell pool is remarkably stable throughout life, reflecting precise regulation of cellular survival, proliferation, and apoptosis. Homeostatic proliferation refers to the process by which T cells spontaneously proliferate in a lymphopenic host. The critical signals driving this expansion are "space," contact with self-major histocompatibility complex (MHC)/peptide complexes, and cytokine stimulation. A number of studies have delineated an association between T cell lymphopenia, compensatory homeostatic expansion, and the development of diverse autoimmune syndromes. In the nonobese diabetic mouse model of type 1 diabetes, lymphopenia-induced homeostatic expansion fuels the generation of islet-specific T cells. Excess interleukin-21 facilitates T cell cycling but limited survival, resulting in recurrent stimulation of T cells specific for self-peptide/MHC complexes. Indeed, data from several experimental models of autoimmunity indicate that a full T cell compartment restrains homeostatic expansion of self-reactive cells that could otherwise dominate the repertoire. This review describes the mechanisms that govern T cell homeostatic expansion and outlines the evidence that lymphopenia presents a risk for development of autoimmune disease.
Collapse
Affiliation(s)
- Annette M Marleau
- The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
143
|
Bettelli E, Kuchroo VK. IL-12- and IL-23-induced T helper cell subsets: birds of the same feather flock together. ACTA ACUST UNITED AC 2005; 201:169-71. [PMID: 15657286 PMCID: PMC2212800 DOI: 10.1084/jem.20042279] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Traditionally, CD4(+) T cells have been separated into two different subsets named T helper (Th)1 and Th2. A new IL-23-driven subset of Th cells called Th(IL-17) has now been described. The data suggest that IL-23 plays an important role in the differentiation of autoreactive pathogenic T cells. Whether these IL-23-induced Th(IL-17) cells are a unique subset or are related to other Th subsets is discussed.
Collapse
Affiliation(s)
- Estelle Bettelli
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|