101
|
Jöhrens K, Dietel M, Anagnostopoulos I. Three different expression patterns of T-bet in angioimmunoblastic T-cell lymphoma. Leuk Lymphoma 2011; 53:152-5. [PMID: 21780995 DOI: 10.3109/10428194.2011.606941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) exhibits a multifaceted clinical picture and distinct architectural patterns that correlate with disease progression and the number of neoplastic cells. In this study we investigated the expression of the transcription factor T-bet and correlated it with the architectural patterns in 29 cases of AITL. Double immunolabelings for T-bet, CD20, CD3 or PD1 revealed the following patterns: predominant T-bet expression by neoplastic T-cells (A), by aggregates of small B-cells (B) or by B-immunoblasts (C). The majority of cases of AITL pattern II showed a T-bet expression pattern B (6/8 cases), while the majority of those with pattern III exhibited the T-bet pattern A (11/21 cases). We propose that T-bet expression by B-cells represents a T-cell independent immune response trying to cope with opportunistic infections, while T-bet expression by neoplastic T-cells is linked to the introduction of a Th17 response responsible for the immunologic derangements characteristic of AITL.
Collapse
Affiliation(s)
- Korinna Jöhrens
- Institute of Pathology, Charité University Medicine Berlin, Campus Mitte, Berlin, Germany.
| | | | | |
Collapse
|
102
|
Ganjalikhani Hakemi M, Ghaedi K, Andalib A, Hosseini M, Rezaei A. Optimization of human Th17 cell differentiation in vitro: evaluating different polarizing factors. In Vitro Cell Dev Biol Anim 2011; 47:581-592. [PMID: 21853398 DOI: 10.1007/s11626-011-9444-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 07/11/2011] [Indexed: 02/05/2023]
Abstract
Regarding discrepancies that exist among different studies which have tried to clarify critical factors in human Th17 cell differentiation, the aim of this study was to identify the best condition for human Th17 differentiation and to clarify the possible role of TGF-β in differentiation of these cells. Naïve CD4(+) T cells were isolated from cord blood samples and cultured either in X-VIVO 15 serum-free medium or RPMI 1640 containing 10% FBS. Purified cells were treated with different combinations of polarizing cytokines (TGF-β, IL-1β, IL-6, IL-23 and IL-21) followed by analysis of the expression of characteristic genes and their relevant cytokines by real-time quantitative RT-PCR and ELISA method, respectively. Our data indicate that a combination of TGF-β plus IL-6 and IL-23 cytokines in X-VIVO 15 serum-free medium could be applied as the best condition for developing human Th17 cells in compare with other studied cytokine treatments. It is shown that TGF-β could be considered as a positive regulator for human Th17 cell differentiation only if applied in average concentrations. Interestingly, polarizing treatments in absence of TGF-β, induced double-secreting Th17 cells which co-express IL-17 and IFN-γ whereas polarization in presence of TGF-β-induced single-secreting (only IL-17 expressing) Th17 cells.
Collapse
|
103
|
Huss DJ, Winger RC, Cox GM, Guerau-de-Arellano M, Yang Y, Racke MK, Lovett-Racke AE. TGF-β signaling via Smad4 drives IL-10 production in effector Th1 cells and reduces T-cell trafficking in EAE. Eur J Immunol 2011; 41:2987-96. [PMID: 21728174 DOI: 10.1002/eji.201141666] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 05/31/2011] [Accepted: 06/24/2011] [Indexed: 12/15/2022]
Abstract
Effector Th1 cells perpetuate inflammatory damage in a number of autoimmune diseases, including MS and its animal model EAE. Recently, a self-regulatory mechanism was described in which effector Th1 cells produce the immunomodulatory cytokine IL-10 to dampen the inflammatory response in both normal and autoimmune inflammation. While the presence of TGF-β has been suggested to enhance and stabilize an IFN-γ(+) IL-10(+) phenotype, the molecular mechanism is poorly understood. Additionally, in the context of adoptive transfer EAE, it is unclear whether IL-10 acts on the transferred Th1 cells or on endogenous host cells. In the present study, using myelin-specific TCR-Tg mice, we show that repetitive Ag stimulation of effector Th1 cells in the presence of TGF-β increases the population of IFN-γ(+) IL-10(+) cells, which correlates with a decrease in EAE severity. Additionally, TGF-β signaling causes binding of Smad4 to the IL-10 promoter, providing molecular evidence for TGF-β-mediated IL-10 production from Th1 effector cells. Finally, this study demonstrates that IL-10 not only reduces encephalitogenic markers such as IFN-γ and T-bet on Th1 effector cells expressing the IL-10R but also prevents recruitment of both transferred and host-derived inflammatory T cells. These data establish a regulatory mechanism by which highly activated Th1 effector cells modulate their pathogenicity through the induction of IL-10.
Collapse
Affiliation(s)
- David J Huss
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University Medical Center, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
104
|
Jadidi-Niaragh F, Mirshafiey A. Th17 cell, the new player of neuroinflammatory process in multiple sclerosis. Scand J Immunol 2011; 74:1-13. [PMID: 21338381 DOI: 10.1111/j.1365-3083.2011.02536.x] [Citation(s) in RCA: 294] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by recurrent episodes of demyelination and axonal lesion mediated by CD4(+) T cells with a proinflammatory Th1 and Th17 phenotype, macrophages, and soluble inflammatory mediators. Identification of Th17 cells led to breaking the dichotomy of Th1/Th2 axis in immunopathogenesis of autoimmune diseases such as MS, and its experimental model, experimental autoimmune encephalomyelitis (EAE). Th17 cells are characterized by expression of retinoic acid-related orphan receptor (ROR)γt and signal transducer and activator of transcription 3 (STAT3) factors. Th17-produced cytokine profile including interleukin (IL)-17, IL-6, IL-21, IL-22, IL-23 and tumour necrosis factor (TNF)-α, which have proinflammatory functions, suggests it as an important factor in immunopathogenesis of MS, because the main feature of MS pathophysiology is the neuroinflammatory reaction. The blood brain barrier (BBB) disruption is an early and central event in MS pathogenesis. Autoreactive Th17 cells can migrate through the BBB by the production of cytokines such as IL-17 and IL-22, which disrupt tight junction proteins in the central nervous system (CNS) endothelial cells. Consistent with this observation and regarding the wide range production of proinflammatory cytokines and chemokines by Th17 cells, it is expected that Th17 cell to be as a potent pathogenic factor in disease immunopathophysiology. Th17-mediated inflammation is characterized by neutrophil recruitment into the CNS and neurons killing. However, the majority of our knowledge about the role of Th17 in MS pathogenesis is resulted in investigation into EAE animal models. In this review, we intend to focus on the newest information regarding the precise role of Th17 cells in immunopathogenesis of MS, and its animal model, EAE.
Collapse
Affiliation(s)
- F Jadidi-Niaragh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
105
|
Abstract
CD4+ T effector lymphocytes are distinguished in different subsets on the basis of their patterns of cytokine secretion. Th1 cells, thank to IFN-γ production, are responsible for cell-mediated immunity against intracellular pathogens, Th2 cells, through the production of IL-4, provide some degree of protection against helminthes, and Th17 cells, via IL-17, promote neutrophils recruitment for the clearance of bacteria and fungi. However, beyond their protective role, these T-helper subsets can also be involved in the pathogenesis of several inflammatory diseases. Asthma is an inflammatory disease characterized by different clinical phenotypes. Allergic asthma is the result of an inflammatory process driven by allergen-specific Th2 lymphocytes, whereas Th17 cells are mainly involved in those forms of asthma, where neutrophils more than eosinophils, contribute to the inflammation. The identification in allergic asthma of Th17/Th2 cells, able to produce both IL-4 and IL-17, is in keeping with the observation that different clinical phenotypes can coexist in the same patient. In conclusion, a picture in which different T-cell subpopulations are active in different phase of bronchial asthma is emerging, and the wide spectrum of clinical phenotypes is probably the expression of different cellular characters playing a role in lung inflammation.
Collapse
Affiliation(s)
- L Cosmi
- Center of Excellence for Research, Transfer of Research and High Education for the Development of Novel therapies, University of Florence, Florence, Italy
| | | | | | | | | |
Collapse
|
106
|
PPAR Regulation of Inflammatory Signaling in CNS Diseases. PPAR Res 2011; 2008:658520. [PMID: 18670616 PMCID: PMC2490815 DOI: 10.1155/2008/658520] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2007] [Revised: 04/22/2008] [Accepted: 05/12/2008] [Indexed: 12/25/2022] Open
Abstract
Central nervous system (CNS) is an immune privileged site, nevertheless inflammation associates with many CNS diseases. Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors that regulate immune and inflammatory responses. Specific ligands for PPARα, γ, and δ isoforms have proven effective in the animal models of multiple sclerosis (MS), Alzheimer's disease, Parkinson's disease, and trauma/stroke, suggesting their use in the treatment of neuroinflammatory diseases. The activation of NF-κB and Jak-Stat signaling pathways and secretion of inflammatory cytokines are critical in the pathogenesis of CNS diseases. Interestingly, PPAR agonists mitigate CNS disease by modulating inflammatory signaling network in immune cells. In this manuscript, we review the current knowledge on how PPARs regulate neuroinflammatory signaling networks in CNS diseases.
Collapse
|
107
|
Racke MK. Immunopathogenesis of multiple sclerosis. Ann Indian Acad Neurol 2011; 12:215-20. [PMID: 20182567 PMCID: PMC2824947 DOI: 10.4103/0972-2327.58274] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 04/10/2009] [Accepted: 07/06/2009] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a suspected autoimmune disease in which myelin-specific CD4+ and CD8+ T cells enter the central nervous system (CNS) and initiate an inflammatory response directed against myelin and other components of the CNS. Acute MS exacerbations are believed be the result of active inflammation, and progression of disability is generally believed to reflect accumulation of damage to the CNS, particularly axonal damage. Over the last several years, the pathophysiology of MS is being appreciated to be much more complex, and it appears that the development of the MS plaque involves a large number of cell populations, including CD8+ T lymphocytes, B cells, and Th17 cells (a population of helper T cells that secrete the inflammatory cytokine IL-17). The axonal transection and degeneration that is thought to represent the basis for progressive MS is now recognized to begin early in the disease process and to continue in the progressive forms of the disease. Molecules important for limiting aberrant neural connections in the CNS have been identified, which suppress axonal sprouting and regeneration of transected axons within the CNS. Pathways have also been identified that prevent remyelination of the MS lesion by oligodendrocyte precursors. Novel neuroimaging methodologies and potential biomarkers are being developed to monitor various aspects of the disease process in MS. As we identify the pathways responsible for the clinical phenomena of MS, we will be able to develop new therapeutic strategies for this disabling illness of young adults.
Collapse
Affiliation(s)
- Michael K Racke
- The Helen C. Kurtz Chair of Neurology, The Ohio State University Medical Center, 395 West 12 Avenue, Columbus, OH 43210 USA
| |
Collapse
|
108
|
Sinha S, Miller LM, Subramanian S, Burrows GG, Vandenbark AA, Offner H. RTL551 treatment of EAE reduces CD226 and T-bet+ CD4 T cells in periphery and prevents infiltration of T-bet+ IL-17, IFN-γ producing T cells into CNS. PLoS One 2011; 6:e21868. [PMID: 21750737 PMCID: PMC3130056 DOI: 10.1371/journal.pone.0021868] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 06/08/2011] [Indexed: 01/12/2023] Open
Abstract
Recombinant T cell receptor ligands (RTLs) that target encephalitogenic T-cells can reverse clinical and histological signs of EAE, and are currently in clinical trials for treatment of multiple sclerosis. To evaluate possible regulatory mechanisms, we tested effects of RTL therapy on expression of pathogenic and effector T-cell maturation markers, CD226, T-bet and CD44, by CD4+ Th1 cells early after treatment of MOG-35-55 peptide-induced EAE in C57BL/6 mice. We showed that 1-5 daily injections of RTL551 (two-domain I-A(b) covalently linked to MOG-35-55 peptide), but not the control RTL550 ("empty" two-domain I-A(b) without a bound peptide) or Vehicle, reduced clinical signs of EAE, prevented trafficking of cells outside the spleen, significantly reduced the frequency of CD226 and T-bet expressing CD4+ T-cells in blood and inhibited expansion of CD44 expressing CD4+ T-cells in blood and spleen. Concomitantly, RTL551 selectively reduced CNS inflammatory lesions, absolute numbers of CNS infiltrating T-bet expressing CD4+ T-cells and IL-17 and IFN-γ secretion by CNS derived MOG-35-55 reactive cells cultured ex vivo. These novel results demonstrate that a major effect of RTL therapy is to attenuate Th1 specific changes in CD4+ T-cells during EAE and prevent expansion of effector T-cells that mediate clinical signs and CNS inflammation in EAE.
Collapse
Affiliation(s)
- Sushmita Sinha
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lisa M. Miller
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Sandhya Subramanian
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, Oregon, United States of America
| | - Gregory G. Burrows
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Arthur A. Vandenbark
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- Research Service, Department of Veterans Affairs Medical Center, Portland, Oregon, United States of America
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Halina Offner
- Neuroimmunology Research, Veterans Affairs Medical Center, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
109
|
Kroenke MA, Segal BM. IL-23 modulated myelin-specific T cells induce EAE via an IFNγ driven, IL-17 independent pathway. Brain Behav Immun 2011; 25:932-7. [PMID: 20951792 PMCID: PMC3064959 DOI: 10.1016/j.bbi.2010.10.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 09/17/2010] [Accepted: 10/01/2010] [Indexed: 11/25/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an inflammatory demyelinating disease of the central nervous system (CNS) mediated by myelin-reactive CD4(+) T cells. An unresolved issue that has important clinical implications concerns the cytokines produced by myelin-reactive T cells that determine their pathogenicity. Initially, IL-12 polarized, IFNγ producing Th1 cells were thought to be essential for the development of EAE. More recently, IL-23 polarized, IL-17 producing Th17 cells have been highlighted as critical encephalitogenic effectors. There is growing evidence that parallel autoimmune pathways can result in common clinical and histopathological endpoints. In the current study, we describe a form of EAE induced by the transfer of IL-23 modulated CD4(+) T cells into IL-17 receptor (IL-17R) deficient hosts. We found that IL-23 stimulates myelin-reactive T cells to produce both IFNγ and IL-17. Surprisingly, in this model the development of EAE is IFNγ dependent. Our findings illustrate a novel mechanism by which IL-23 promotes encephalitogenicity and they further expand the spectrum of autoreactive T cells capable of mediating inflammatory demyelinating disease of the CNS.
Collapse
Affiliation(s)
- Mark A. Kroenke
- Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Department of Neurology, University of Michigan, Ann Arbor, MI 48109
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Benjamin M. Segal
- Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Department of Neurology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
110
|
Cravens PD, Hussain RZ, Zacharias TE, Ben LH, Herndon E, Vinnakota R, Lambracht-Washington D, Nessler S, Zamvil SS, Eagar TN, Stüve O. Lymph node-derived donor encephalitogenic CD4+ T cells in C57BL/6 mice adoptive transfer experimental autoimmune encephalomyelitis highly express GM-CSF and T-bet. J Neuroinflammation 2011; 8:73. [PMID: 21702922 PMCID: PMC3161869 DOI: 10.1186/1742-2094-8-73] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 06/24/2011] [Indexed: 01/20/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a relevant animal model for the human demyelinating inflammatory disorder of the central nervous system (CNS), multiple sclerosis (MS). Induction of EAE by adoptive transfer allows studying the role of the donor T lymphocyte in disease pathogenesis. It has been challenging to reliably induce adoptive transfer EAE in C57BL/6 (H-2b) mice. The goal of this study was to develop a reproducible and high yield protocol for adoptive transfer EAE in C57BL/6 mice. A step-wise experimental approach permitted us to develop a protocol that resulted in a consistent relatively high disease incidence of ~70% in recipient mice. Donor mice were immunized with myelin oligodendrocyte glycoprotein (MOG)p35-55 in complete Freund's adjuvant (CFA) followed by pertussis toxin (PT). Only lymph node cells (LNC) isolated at day 12 post immunization, and restimulated in vitro for 72 hours with 10 μg/mL of MOGp35-55 and 0.5 ng/mL of interleukin-12 (IL-12) were able to transfer disease. The ability of LNC to transfer disease was associated with the presence of inflammatory infiltrates in the CNS at day 12. Interferon gamma (IFNγ) was produced at comparable levels in cell cultures prepared from mice at both day 6 and day 12 post immunization. By contrast, there was a trend towards a negative association between IL-17 and disease susceptibility in our EAE model. The amount of GM-CSF secreted was significantly increased in the culture supernatants from cells collected at day 12 post immunization versus those collected at day 6 post-immunization. Activated CD4+ T cells present in the day 12 LNC cultures maintained expression of the transcription factor T-bet, which has been shown to regulate the expression of the IL-23 receptor. Also, there was an increased prevalence of MOGp35-55-specific CD4+ T cells in day 12 LNC after in vitro re-stimulation. In summary, encephalitogenic LNC that adoptively transfer EAE in C57BL/6 mice were not characterized by a single biomarker in our study, but by a composite of inflammatory markers. Our data further suggest that GM-CSF expression by CD4+ T cells regulated by IL-23 contributes to their encephalitogenicity in our EAE model.
Collapse
Affiliation(s)
- Petra D Cravens
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Abdelsamed HA, Desai M, Nance SC, Fitzpatrick EA. T-bet controls severity of hypersensitivity pneumonitis. JOURNAL OF INFLAMMATION-LONDON 2011; 8:15. [PMID: 21699708 PMCID: PMC3131238 DOI: 10.1186/1476-9255-8-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 06/23/2011] [Indexed: 05/26/2023]
Abstract
Hypersensitivity Pneumonitis (HP) is an interstitial lung disease that develops following repeated exposure to inhaled environmental antigens. The disease is characterized by alveolitis, granuloma formation and in some patients' fibrosis. IFNγ plays a critical role in HP; in the absence of IFNγ granuloma formation does not occur. However, recent studies using animal models of HP have suggested that HP is a Th17 disease calling into question the role of IFNγ. In this study, we report that initially IFNγ production is dependent on IL-18 and the transcription factor T-bet, however as the disease continues IFNγ production is IL-18-independent and partially T-bet dependent. Although IFNγ production is required for granuloma formation its role is distinct from that of T-bet. Mice that are deficient in T-bet and exposed to S. rectivirgula develop more severe disease characterized by an exacerbated Th17 cell response, decreased Th1 cell response, and increased collagen production in the lung. T-bet-mediated protection does not appear to be due to the development of a protective Th1 response; shifting the balance from a Th17 predominant response to a Th1 response by inhibition of IL-6 also results in lung pathology. The results from this study suggest that both Th1 and Th17 cells can be pathogenic in this model and that IFNγ and T-bet play divergent roles in the disease process.
Collapse
Affiliation(s)
- Hossam Aly Abdelsamed
- University of Tennessee Health Science Center, Dept, of Microbiology, Immunology and Biochemistry, Memphis, TN 38163, USA.
| | | | | | | |
Collapse
|
112
|
El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, Rostami A. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol 2011; 12:568-75. [PMID: 21516111 PMCID: PMC3116521 DOI: 10.1038/ni.2031] [Citation(s) in RCA: 878] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 04/04/2011] [Indexed: 12/12/2022]
Abstract
Interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) require exposure to IL-23 to become encephalitogenic, but the mechanism by which IL-23 promotes their pathogenicity is not known. Here we found that IL-23 induced production of the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) in T(H)17 cells and that GM-CSF had an essential role in their encephalitogenicity. Our findings identify a chief mechanism that underlies the important role of IL-23 in autoimmune diseases. IL-23 induced a positive feedback loop whereby GM-CSF secreted by T(H)17 cells stimulated the production of IL-23 by antigen-presenting cells. Such cross-regulation of IL-23 and GM-CSF explains the similar pattern of resistance to autoimmunity when either of the two cytokines is absent and identifies T(H)17 cells as a crucial source of GM-CSF in autoimmune inflammation.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibodies/pharmacology
- CD11c Antigen/immunology
- CD11c Antigen/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Coculture Techniques
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Flow Cytometry
- Glycoproteins
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Interleukin-1/pharmacology
- Interleukin-1beta/pharmacology
- Interleukin-23/immunology
- Interleukin-23/metabolism
- Interleukin-23/pharmacology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Myelin-Oligodendrocyte Glycoprotein
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Peptide Fragments
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Mohamed El-Behi
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Transcription factor Smad-independent T helper 17 cell induction by transforming-growth factor-β is mediated by suppression of eomesodermin. Immunity 2011; 34:741-54. [PMID: 21600798 DOI: 10.1016/j.immuni.2011.02.021] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2010] [Revised: 01/05/2011] [Accepted: 02/04/2011] [Indexed: 12/29/2022]
Abstract
Transforming growth factor-β (TGF-β) has been shown to be required for Th17 cell differentiation via Smad-independent mechanisms. The molecular mechanism underlying this pathway remains to be clarified, however. We searched for genes regulated by TGF-β through the Smad-independent pathway by using Smad2 and Smad3 double-deficient T cells and identified the transcription factor Eomesodermin (Eomes), whose expression was suppressed by TGF-β via the c-Jun N-terminal kinase (JNK)-c-Jun signaling pathway. Inhibition of JNK strongly suppressed disease in an in vivo EAE model as well as in vitro Th17 cell induction. Overexpression of Eomes substantially suppressed Th17 cell differentiation, whereas ablation of Eomes expression could substitute for TGF-β in Th17 cell induction in primary T cells. Eomes suppressed Rorc and Il17a promoters by directly binding to the proximal region of these promoters. In conclusion, the suppression of Eomes by TGF-β via the JNK pathway is an important mechanism for Smad-independent Th17 cell differentiation.
Collapse
|
114
|
Lewis W, Simanyi E, Li H, Thompson CA, Nasti TH, Jaleel T, Xu H, Yusuf N. Regulation of ultraviolet radiation induced cutaneous photoimmunosuppression by toll-like receptor-4. Arch Biochem Biophys 2011; 508:171-7. [PMID: 21236239 PMCID: PMC3115632 DOI: 10.1016/j.abb.2011.01.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/06/2011] [Accepted: 01/06/2011] [Indexed: 10/18/2022]
Abstract
UVB radiation is a potent immunosuppressive agent that inhibits cell-mediated immune responses. The mechanisms by which UVB radiation influences cell-mediated immune responses have been the subject of extensive investigation. However, the role of innate immunity on photoimmunological processes has received little attention. The purpose of this study was to determine whether Toll-like receptor-4 (TLR4) contributed to UV-induced suppression of contact hypersensitivity (CHS) responses. TLR4⁻/⁻ and wild type C57BL/6 (TLR4+/+) mice were subjected to a local UVB immunosuppression regimen consisting of 100 mJ/cm² UVB radiation followed by sensitization with the hapten DNFB. Wild type TLR4+/+ mice exhibited significant suppression of contact hypersensitivity response, whereas TLR4⁻/⁻ developed significantly less suppression. The suppression in wild type TLR4+/+ mice could be adoptively transferred to naïve syngeneic recipients. Moreover, there were significantly fewer Foxp3 expressing CD4+CD25+ regulatory T-cells in the draining lymph nodes of UV-irradiated TLR4⁻/⁻ mice than TLR4+/+ mice. When cytokine levels were compared in these two strains after UVB exposure, T-cells from TLR4+/+ mice produced higher levels of IL-10 and TGF-β and lower levels of IFN-γ and IL-17. Strategies to inhibit TLR4 may allow us to develop immunopreventive and immunotherapeutic approaches for management of UVB induced cutaneous immunosuppression.
Collapse
Affiliation(s)
- Wesley Lewis
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | - Eva Simanyi
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | - Hui Li
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | - Camilla A Thompson
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | - Tahseen H Nasti
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | - Tarannum Jaleel
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
| | | | - Nabiha Yusuf
- Department of Dermatology and Skin Diseases Research Center, Birmingham, AL
- Veteran Affairs Medical Center, Birmingham, AL
- Comprehensive Cancer Center, University of Alabama at Birmingham, AL
| |
Collapse
|
115
|
Itk: the rheostat of the T cell response. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:297868. [PMID: 21747996 PMCID: PMC3116522 DOI: 10.1155/2011/297868] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Accepted: 01/19/2011] [Indexed: 12/28/2022]
Abstract
The nonreceptor tyrosine kinase Itk plays a key role in TCR-initiated signaling that directly and significantly affects the regulation of PLCγ1 and the consequent mobilization of Ca2+. Itk also participates in the regulation of cytoskeletal reorganization as well as cellular adhesion, which is necessary for a productive T cell response. The functional cellular outcome of these molecular regulations by Itk renders it an important mediator of T cell development and differentiation. This paper encompasses the structure of Itk, the signaling parameters leading to Itk activation, and Itk effects on molecular pathways resulting in functional cellular outcomes. The incorporation of these factors persuades one to believe that Itk serves as a modulator, or rheostat, critically fine-tuning the T cell response.
Collapse
|
116
|
Lambracht-Washington D, Qu BX, Fu M, Anderson LD, Stüve O, Eagar TN, Rosenberg RN. DNA immunization against amyloid beta 42 has high potential as safe therapy for Alzheimer's disease as it diminishes antigen-specific Th1 and Th17 cell proliferation. Cell Mol Neurobiol 2011; 31:867-74. [PMID: 21625960 DOI: 10.1007/s10571-011-9680-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 02/23/2011] [Indexed: 10/18/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) has been strongly associated with the accumulation of amyloid beta (Aβ) peptides in brain, and immunotherapy targeting Aβ provides potential for AD prevention. A clinical trial in which AD patients were immunized with Aβ42 peptide was stopped when 6% of participants showed meningoencephalitis, apparently due to an inflammatory Th1 immune response. Previously, we and other have shown that Aβ42 DNA vaccination via gene gun generates a Th2 cellular immune response, which was shown by analyses of the respective antibody isotype profiles. We also determined that in vitro T cell proliferation in response to Aβ42 peptide re-stimulation was absent in DNA Aβ42 trimer-immunized mice when compared to Aβ42 peptide-immunized mice. To further characterize this observation prospectively and longitudinally, we analyzed the immune response in wild-type mice after vaccination with Aβ42 trimer DNA and Aβ42 peptide with Quil A adjuvant. Wild-type mice were immunized with short-term (1-3× vaccinations) or long-term (6× vacinations) immunization strategies. Antibody titers and isotype profiles of the Aβ42 specific antibodies, as well as cytokine profiles and cell proliferation studies from this longitudinal study were determined. Sufficient antibody titers to effectively reduce Aβ42, but an absent T cell proliferative response and no IFNγ or IL-17 secretion after Aβ42 DNA trimer immunization minimizes the risk of inflammatory activities of the immune system towards the self antigen Aβ42 in brain. Therefore, Aβ42 DNA trimer immunization has a high probability to be effective and safe to treat patients with early AD.
Collapse
|
117
|
Li C, Zhao Y, Zhang W, Zhang W. Increased prevalence of T(H)17 cells in the peripheral blood of patients with head and neck squamous cell carcinoma. ACTA ACUST UNITED AC 2011; 112:81-9. [PMID: 21458322 DOI: 10.1016/j.tripleo.2010.11.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 11/12/2010] [Accepted: 11/19/2010] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study was performed to determine whether T(H)17 cells are involved in the development and metastasis of head and neck squamous cell carcinomas (HNSCCs). STUDY DESIGN T(H)17 cells frequencies in 67 HNSCC patients and 21 healthy volunteers were examined by flow cytometric analysis. T(H)17 cell-related cytokines in serum (interleukin (IL) 17, transforming growth factor (TGF) β, and IL-6) were evaluated by using enzyme-linked immunosorbent assay. RESULTS It was discovered that the higher frequency of T(H)17 cells was in HNSCCs patients (1.0 ± 0.4%). The cell proportions and related cytokine concentrations were consistent with the tumor TNM stage. The IL-6 concentration showed positive correlation with the frequency of T(H)17 cells (r = 0.661) and IL-17 levels (r = 0.597). The TGF-β concentration showed a positive correlation with IL-17 (r = 0.626) but no relationship with T(H)17 cells (r = 0.431). CONCLUSIONS The present data suggested that T(H)17 cells may be involved in tumor growth and metastasis of HNSCCs. IL-6 may play an important role in T(H)17 cell differentiation and functions, and TGF-β may be related to IL-17 secretion but not to the differentiation of T(H)17 cells.
Collapse
Affiliation(s)
- Chen Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Ministry of Education, Wuhan University, Wuhan, China
| | | | | | | |
Collapse
|
118
|
Boppana S, Huang H, Ito K, Dhib-Jalbut S. Immunologic Aspects of Multiple Sclerosis. ACTA ACUST UNITED AC 2011; 78:207-20. [DOI: 10.1002/msj.20249] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
119
|
Comabella M, Khoury SJ. Immunopathogenesis of multiple sclerosis. Clin Immunol 2011; 142:2-8. [PMID: 21458377 DOI: 10.1016/j.clim.2011.03.004] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 03/02/2011] [Accepted: 03/03/2011] [Indexed: 01/10/2023]
Abstract
Multiple sclerosis (MS) is a chronic disorder of the central nervous system characterized by autoimmune inflammation, demyelination, and axonal damage. MS etiology remains unknown, but disease phenotype is most likely the result of an interaction between complex genetic factors and environmental influences. The better understanding of the mechanisms involved in the immunopathogenesis of MS has led to the development of promising new therapeutic strategies for the disease. This review will discuss the key pathogenic steps implicated in the disease and the role of the main cellular populations that drive the immune responses in MS.
Collapse
Affiliation(s)
- Manuel Comabella
- Centre d'Esclerosi Múltiple de Catalunya, CEM-Cat, Unitat de Neuroimmunologia Clínica, Hospital Universitari Vall d'Hebron (HUVH), Barcelona, Spain.
| | | |
Collapse
|
120
|
Ye J, Su X, Hsueh EC, Zhang Y, Koenig JM, Hoft DF, Peng G. Human tumor-infiltrating Th17 cells have the capacity to differentiate into IFN-γ+ and FOXP3+ T cells with potent suppressive function. Eur J Immunol 2011; 41:936-51. [PMID: 21381020 DOI: 10.1002/eji.201040682] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 11/29/2010] [Accepted: 01/05/2011] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that Th17 cells and Tregs may exhibit development plasticity and that CD4(+) Tregs can differentiate into IL-17-producing T cells; however, whether Th17 cells can reciprocally convert into Tregs has not been described. In this study, we generated Th17 clones from tumor-infiltrating T lymphocytes (TILs). We showed that Th17 clones generated from TILs can differentiate into IFN-γ-producing and FOXP3(+) cells after in vitro stimulation with OKT3 and allogeneic peripheral blood mononuclear cells. We further demonstrated that T-cell receptor (TCR) engagement was responsible for this conversion, and that this differentiation was due to the epigenetic modification and reprogramming of gene expression profiles, including lineage-specific transcriptional factor and cytokine genes. In addition to expressing IFN-γ and FOXP3, we showed that these differentiated Th17 clones mediated potent suppressive function after repetitive stimulation with OKT3, suggesting that these Th17 clones had differentiated into functional Tregs. We further demonstrated that the Th17-derived Tregs, unlike naturally occurring CD4(+) CD25(+) Tregs, did not reconvert back into Th17 cells even under Th17-biasing cytokine conditions. These results provide the critical evidence that human tumor-infiltrating Th17 cells can differentiate into Tregs and indicate a substantial developmental plasticity of Th17 cells.
Collapse
Affiliation(s)
- Jian Ye
- Division of Immunobiology, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, MO 63104, USA
| | | | | | | | | | | | | |
Collapse
|
121
|
Ji N, Sosa RA, Forsthuber TG. More than just a T-box: the role of T-bet as a possible biomarker and therapeutic target in autoimmune diseases. Immunotherapy 2011; 3:435-41. [PMID: 21395384 PMCID: PMC3160828 DOI: 10.2217/imt.10.111] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
T-bet was initially described as a T-box transcription factor with an essential role in orchestrating Th1 cell differentiation. Subsequently, it was determined that T-bet controls the expression of numerous cytokines and their receptors, adhesion molecules and chemokine receptors, and therefore determines the differentiation and development status of many types of immune cells. The critical role of T-bet in autoimmune diseases, particularly multiple sclerosis and its animal model experimental autoimmune encephalomyelitis, implicates it as a potential biomarker for pathogenic T cells as well as a therapeutic drug target.
Collapse
Affiliation(s)
- Niannian Ji
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | - Rebecca A Sosa
- Department of Biology, University of Texas at San Antonio, TX 78249, USA
| | | |
Collapse
|
122
|
Nasti TH, Iqbal O, Tamimi IA, Geise JT, Katiyar SK, Yusuf N. Differential roles of T-cell subsets in regulation of ultraviolet radiation induced cutaneous photocarcinogenesis. Photochem Photobiol 2011; 87:387-98. [PMID: 21143237 PMCID: PMC3049951 DOI: 10.1111/j.1751-1097.2010.00859.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ultraviolet (UV) radiation, in particular the midwavelength range (UVB; 290-320 nm), is one of the most significant risk factors for the development of nonmelanoma skin cancer. UVB radiation-induced immunosuppression, which occurs in both humans and laboratory animals, contributes to their pathogenesis. However, there are conflicting reports on the relative role of CD4(+) and CD8(+) T cells in UVB induced skin cancer. The purpose of this study was to delineate the contribution of these two cell subpopulations to UVB induced immunosuppression and tumor development using C3H/HeN (WT), CD4 knockout (CD4(-/-) ) and CD8 knockout (CD8(-/-) ) mice. We observed that UVB induced skin carcinogenesis was retarded in terms of number of tumors per group, tumor volume and percentage of mice with tumors, in mice deficient in CD4(+) T cells compared with wild-type mice, whereas significantly greater (P < 0.05) numbers of tumors occurred in CD8(-/-) mice. These results indicate that, CD4(+) T cells promote tumor development while CD8(+) T cells have the opposite effect. Further, we found that CD4(+) T cells from tumor-bearing mice produced interleukin (IL)-4, IL-10, and IL-17 whereas CD8(+) T cells produced interferon-γ. Manipulation of T-cell subpopulations that are induced by UVB radiation could be a means of preventing skin cancers caused by this agent.
Collapse
Affiliation(s)
- Tahseen H Nasti
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
| | - Omer Iqbal
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
| | - Iman A Tamimi
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
| | - James T Geise
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
| | - Santosh K Katiyar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
- Veteran Affairs Medical Center, University of Alabama at Birmingham, AL
- Comprehensive Cancer Center, University of Alabama at Birmingham, AL
| | - Nabiha Yusuf
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, AL
- Veteran Affairs Medical Center, University of Alabama at Birmingham, AL
- Comprehensive Cancer Center, University of Alabama at Birmingham, AL
| |
Collapse
|
123
|
Sharabi A, Mozes E. Harnessing regulatory T cells for the therapy of lupus and other autoimmune diseases. Immunotherapy 2011; 1:385-401. [PMID: 20635958 DOI: 10.2217/imt.09.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Regulatory T cells (Tregs) maintain immunological homeostasis and prevent autoimmunity. The depletion or functional alteration of Tregs may lead to the development of autoimmune diseases. Tregs consist of different subpopulations of cells, of which CD4(+)CD25(+)Foxp3(+) cells are the most well characterized. However, CD8 Tregs also constitute a major cell population that has been shown to play an important role in autoimmune diseases. This review will discuss the role of Tregs in autoimmune diseases in general and specifically in systemic lupus erythematosus (SLE). SLE is a multisystem autoimmune disease characterized by the production of autoantibodies against nuclear components and by the deposition of immune complexes in the kidneys as well as in other organs. Abnormalities in Tregs were reported in SLE patients and in animal models of the disease. Current treatment of SLE is based on immunosuppressive drugs that are nonspecific and may cause adverse effects. Therefore, the development of novel, specific, side effect-free therapeutic means that will induce functional Tregs is a most desirable goal. Our group and others have designed and utilized tolerogenic peptides that ameliorate SLE manifestations in murine models. Here, we demonstrate the role of CD4 and CD8 Tregs, as well as the interaction between the two subsets of cells and the mechanism of action of the tolerogenic peptides. We also discuss their therapeutic potential for the treatment of SLE.
Collapse
Affiliation(s)
- Amir Sharabi
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
124
|
Lovett-Racke AE, Yang Y, Racke MK. Th1 versus Th17: are T cell cytokines relevant in multiple sclerosis? BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1812:246-51. [PMID: 20600875 PMCID: PMC3004998 DOI: 10.1016/j.bbadis.2010.05.012] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 05/17/2010] [Indexed: 12/01/2022]
Abstract
Our understanding of the pathophysiology of multiple sclerosis (MS) has evolved significantly over the past two decades as the fields of immunology and neurobiology provide new avenues of exploration into the cause and mechanism of the disease. It has been known for decades that T cells have different cytokine phenotypes, yet the cytokine phenotype of pathogenic T cells in MS is still an area of debate. In EAE, it appears that IFNγ and IL-17, produced by Th1 and Th17 cells respectively, are not the critical factor that determines T cell encephalitogenicity. However, there are molecules such as IL-23, T-bet and STAT4, that appear to be critical, yet it is unclear whether all these molecules contribute to a common, yet undefined pathway, or act in a synergistic manner which culminates in encephalitogenicity has still to be determined. Therefore, the focus of research on effector T cells in MS should focus on pathways upstream of the cytokines that define Th1 and Th17 cells, since downstream products, such as IFNγ and IL-17, probably are not critical determinants of whether an effector T cells is capable of trafficking to the CNS and inducing inflammatory demyelination.
Collapse
Affiliation(s)
- Amy E Lovett-Racke
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Medical Center, Columbus, OH 43210, USA.
| | | | | |
Collapse
|
125
|
Yu B, Shao Y, Yue X, Zhang J, Guan M, Wan J, Zhang W. Copy number variations of Interleukin-12B and T-bet are associated with systemic lupus erythematosus. Rheumatology (Oxford) 2011; 50:1201-5. [DOI: 10.1093/rheumatology/keq439] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
126
|
Lazarevic V, Chen X, Shim JH, Hwang ES, Jang E, Bolm AN, Oukka M, Kuchroo VK, Glimcher LH. T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt. Nat Immunol 2011; 12:96-104. [PMID: 21151104 PMCID: PMC3077962 DOI: 10.1038/ni.1969] [Citation(s) in RCA: 298] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 11/12/2010] [Indexed: 12/12/2022]
Abstract
Overactive responses by interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) are tightly linked to the development of autoimmunity, yet the factors that negatively regulate the differentiation of this lineage remain unknown. Here we report that the transcription factor T-bet suppressed development of the T(H)17 cell lineage by inhibiting transcription of Rorc (which encodes the transcription factor RORγt). T-bet interacted with the transcription factor Runx1, and this interaction blocked Runx1-mediated transactivation of Rorc. T-bet Tyr304 was required for formation of the T-bet-Runx1 complex, for blockade of Runx1 activity and for inhibition of the T(H)17 differentiation program. Our data reinforce the idea of master regulators that shape immune responses by simultaneously activating one genetic program while silencing the activity of competing regulators in a common progenitor cell.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Lineage/genetics
- Cells, Cultured
- Core Binding Factor Alpha 2 Subunit/immunology
- Core Binding Factor Alpha 2 Subunit/metabolism
- Cytokines/genetics
- Cytokines/immunology
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation
- Gene Regulatory Networks
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Protein Binding/genetics
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- T-Box Domain Proteins/metabolism
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Th17 Cells/pathology
- T-bet Transcription Factor
Collapse
Affiliation(s)
- Vanja Lazarevic
- Harvard School of Public Health, Department of Immunology and Infectious Diseases, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
127
|
PPARδ deficient mice develop elevated Th1/Th17 responses and prolonged experimental autoimmune encephalomyelitis. Brain Res 2010; 1376:101-12. [PMID: 21192919 DOI: 10.1016/j.brainres.2010.12.059] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 12/17/2010] [Accepted: 12/19/2010] [Indexed: 02/04/2023]
Abstract
Multiple sclerosis (MS) is a neurological disorder that affects more than a million people worldwide. The etiology of MS is not known and there is no medical treatment that can cure MS. Earlier studies have shown that peroxisome proliferator-activated receptor (PPARs) agonists ameliorate MS-like disease in experimental allergic encephalomyelitis (EAE). In this study we have used PPARδ deficient mice to determine its physiological role in the regulation of CNS EAE and MS. We found that PPARδ(-/-) mice develop EAE with similar day of onset and disease incidence compared to C57BL/6 wild type mice. Interestingly, both male and female PPARδ(-/-) mice showed prolonged EAE with resistance to remission and recovery. PPARδ(-/-) mice with EAE expressed elevated levels of IFNγ and IL-17 along with IL-12p35 and IL-12p40 in the brain and spleen. PPARδ(-/-) mice also developed augmented neural antigen-specific Th1/Th17 responses and impaired Th2/Treg responses compared to wild type mice. These findings indicate that PPARδ(-/-) mice develop prolonged EAE in association with augmented Th1/Th17 responses, suggesting a critical physiological role for PPARδ in the remission and recovery of EAE.
Collapse
|
128
|
Frisullo G, Iorio R, Plantone D, Marti A, Nociti V, Patanella AK, Batocchi AP. CD4+T-bet+, CD4+pSTAT3+ and CD8+T-bet+ T cells accumulate in peripheral blood during NZB treatment. Mult Scler 2010; 17:556-66. [PMID: 21177324 DOI: 10.1177/1352458510392263] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Circulating T cells and monocytes expressing T-bet, pSTAT1 and pSTAT3 increase in relapsing-remitting multiple sclerosis (RRMS) during relapse. Natalizumab (NZB) is an effective drug in RRMS, but exacerbation of the disease after its discontinuation has been described in some patients. The aim of this research was to study the effect of NZB treatment on circulating lymphomonocyte subpopulations expressing T-bet, pSTAT1, pSTAT3 and CD4+CD25+Foxp3+ regulatory T cells. Flow cytometry was used to evaluate the percentages of circulating CD4+ and CD8+ T cells, CD14+ monocytes and B cells expressing T-bet, pSTAT1, and pSTAT3, and CD4+CD25+Foxp3+ regulatory T cells from RRMS patients before and after 6-12 NZB infusions. In NZB-treated RRMS patients, the percentages of CD4+pSTAT1+ and CD8+pSTAT1+ T cells, CD14+pSTAT1+ monocytes, CD4+T-bet+, CD8+T-bet+ and CD4+pSTAT3+ T cells and CD14+pSTAT3+ monocytes increased after 12 drug infusions and were similar to those observed in untreated relapsing RRMS patients. Otherwise in vitro NZB exposure of peripheral blood mononuclear cells from untreated RRMS patients and controls had no effect. It was concluded that NZB treatment determines an accumulation of CD4+pSTAT1+, CD8+pSTAT1+, CD4+T-bet+, CD8+T-bet+ and CD4+STAT3+ T cells in peripheral blood that may account for the exacerbation of the disease observed in some patients after the discontinuation of the drug.
Collapse
Affiliation(s)
- Giovanni Frisullo
- Institute of Neurology, Department of Neurosciences, Catholic University, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
129
|
Powell N, Canavan JB, MacDonald TT, Lord GM. Transcriptional regulation of the mucosal immune system mediated by T-bet. Mucosal Immunol 2010; 3:567-77. [PMID: 20844482 DOI: 10.1038/mi.2010.53] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The immune system faces the arduous task of defending the mucosal surfaces from invading pathogens, but must simultaneously repress responses against commensal organisms and other inert antigens that are abundant in the external environment, as inappropriate immune activation might expose the host to increased risk of autoimmunity. The behavior of individual immune cells is governed by the expression of transcription factors that are responsible for switching immune response genes on and off. T-bet (T-box expressed in T cells) has emerged as one of the key transcription factors responsible for controlling the fate of both innate and adaptive immune cells, and its expression in different immune cells found at mucosal surfaces is capable of dictating the critical balance between permitting robust host immunity and limiting susceptibility to autoimmunity and allergy.
Collapse
Affiliation(s)
- N Powell
- National Institute for Health Research (NIHR) Comprehensive Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, King's College London, London, UK
| | | | | | | |
Collapse
|
130
|
Fletcher JM, Lalor SJ, Sweeney CM, Tubridy N, Mills KHG. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol 2010; 162:1-11. [PMID: 20682002 DOI: 10.1111/j.1365-2249.2010.04143.x] [Citation(s) in RCA: 721] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS), which involves autoimmune responses to myelin antigens. Studies in experimental autoimmune encephalomyelitis (EAE), an animal model for MS, have provided convincing evidence that T cells specific for self-antigens mediate pathology in these diseases. Until recently, T helper type 1 (Th1) cells were thought to be the main effector T cells responsible for the autoimmune inflammation. However more recent studies have highlighted an important pathogenic role for CD4(+) T cells that secrete interleukin (IL)-17, termed Th17, but also IL-17-secreting γδ T cells in EAE as well as other autoimmune and chronic inflammatory conditions. This has prompted intensive study of the induction, function and regulation of IL-17-producing T cells in MS and EAE. In this paper, we review the contribution of Th1, Th17, γδ, CD8(+) and regulatory T cells as well as the possible development of new therapeutic approaches for MS based on manipulating these T cell subtypes.
Collapse
Affiliation(s)
- J M Fletcher
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity College, St Vincent's University Hospital, Dublin, Ireland
| | | | | | | | | |
Collapse
|
131
|
Wainwright DA, Sengupta S, Han Y, Ulasov IV, Lesniak MS. The presence of IL-17A and T helper 17 cells in experimental mouse brain tumors and human glioma. PLoS One 2010; 5:e15390. [PMID: 21060663 PMCID: PMC2963644 DOI: 10.1371/journal.pone.0015390] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Accepted: 08/20/2010] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Recently, CD4(+)IL-17A(+) T helper 17 (Th17) cells were identified and reported in several diseased states, including autoimmunity, infection and various peripheral nervous system tumors. However, the presence of Th17 in glia-derived tumors of the central nervous system has not been studied. METHODOLOGY/PRINCIPAL FINDINGS In this report, we demonstrate that mRNA expression for the Th17 cell cytokine IL-17A, as well as Th17 cells, are present in human glioma. The mRNA expression for IL-17A in glioma was recapitulated in an immunocompetent mouse model of malignant glioma. Furthermore, the presence of Th17 cells was confirmed in both human and mouse glioma. Interestingly, some Th17 cells present in mouse glioma co-expressed the Th1 and Th2 lineage markers, IFN-γ and IL-4, respectively, but predominantly co-expressed the Treg lineage marker FoxP3. CONCLUSIONS These data confirm the presence of Th17 cells in glia-derived CNS tumors and provide the rationale for further investigation into the role of Th17 cells in malignant glioma.
Collapse
Affiliation(s)
- Derek A. Wainwright
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, United States of America
| | - Sadhak Sengupta
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, United States of America
| | - Yu Han
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, United States of America
| | - Ilya V. Ulasov
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, United States of America
| | - Maciej S. Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
132
|
Ye ZJ, Zhou Q, Gu YY, Qin SM, Ma WL, Xin JB, Tao XN, Shi HZ. Generation and differentiation of IL-17-producing CD4+ T cells in malignant pleural effusion. THE JOURNAL OF IMMUNOLOGY 2010; 185:6348-54. [PMID: 20952674 DOI: 10.4049/jimmunol.1001728] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IL-17-producing CD4(+) T (Th17) cells have been found to be increased in some human cancers; however, the possible implication of Th17 cells in regulating antitumor responses in malignant pleural effusion (MPE) remains to be elucidated. In the current study, distribution and phenotypic features of Th17 cells in both MPE and peripheral blood from patients with lung cancer were determined by flow cytometry or double immunofluorescence staining. The impacts of cytokines on Th17 cell generation and differentiation were explored. The chemoattractant activity of chemokines CCL20 and CCL22 for Th17 cells in vitro was also observed. It was found that the increased Th17 cells could be found in MPE compared with blood. The in vitro experiments showed that IL-1β, IL-6, IL-23, or their various combinations could promote Th17 cell generation and differentiation from naive CD4(+) T cells. MPE was chemotactic for Th17 cells, and this activity was partly blocked by anti-CCL20 and/or CCL22 Abs. Our data also showed that the accumulation of Th17 cells in MPE predicted improved patient survival. It could be concluded that the overrepresentation of Th17 cells in MPE might be due to Th17 cell differentiation and expansion stimulated by pleural proinflammatory cytokines and to recruitment of Th17 cells from peripheral blood induced by pleural chemokines CCL20 and CCL22. Furthermore, the accumulation of Th17 cells in MPE predicted improved patient survival. These data provide the basis for developing immune-boosting strategies based on ridding the cancer patient of this cell population.
Collapse
Affiliation(s)
- Zhi-Jian Ye
- Department of Respiratory Diseases, Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Abstract
Gut inflammation occurring in patients with Crohn's disease and patients with ulcerative colitis has been traditionally associated with an exaggerated Th1 or Th2 cell response, respectively. However, recent studies have shown that in both inflammatory bowel diseases (IBD) there is also enhanced synthesis of cytokines made by a distinct subset of T helper cells, termed Th17 cells. The discovery that this new T-cell subset drives immune-mediated pathology in the gut, and that interleukin (IL)-23 amplifies Th17 cell responses and gut inflammation, has contributed to elucidate new pathways of tissue damage as well as to open new avenues for development of therapeutic strategies in IBD. Nonetheless, it has been recently shown that Th17-related cytokines, such as IL-17A and IL-22, can exert protective rather than detrimental effects in the gut. We here review the available data regarding the role of Th17 cells and IL-23 in chronic intestinal inflammation.
Collapse
Affiliation(s)
- Massimiliano Sarra
- Department of Internal Medicine, University Tor Vergata of Rome, Rome, Italy.
| | | | | | | |
Collapse
|
134
|
Lee K, Min HJ, Jang EJ, Hong JH, Hwang ES. In vivo tumor suppression activity by T cell-specific T-bet restoration. Int J Cancer 2010; 127:2129-37. [PMID: 20143391 DOI: 10.1002/ijc.25238] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
T-box-containing protein expressed in T cells (T-bet) is a master transcription factor for the development of interferon (IFN) gamma-producing T helper 1 (Th1) cells and also functions in other immune cells including natural killer (NK), cytotoxic T lymphocytes and dendritic cells. T-bet-deficient mice increased susceptibility to viral infection and tumor development due to the defective functions of immune cells. T-bet is known to play a key role in NK-mediated antimetastatic response; however, it remains to be characterized whether T-bet is essential for in vivo tumor suppression mediated by T cells. Here, we have investigated in vivo tumor suppression effect of T-bet-restored T cells using T cell-specific and inducible T-bet transgenic mice generated in a T-bet-deficient background. T-bet-null mice increased susceptibility to tumor development, whereas induction of T cell-specific T-bet expression upon melanoma cell injection substantially suppressed tumor development by inducing IFNgamma production in T cells and tumor cell apoptosis. Late induction of T-bet expression in tumor-bearing mice produced comparable amounts of IFNgamma with control and significantly decreased tumor volume. In addition, increased melanoma lung metastasis in T-bet-deficient mice was strikingly inhibited by T-bet restoration in T cells. Intravenous injection of activated Th1 cells, not T-bet-null Th1 cells, attenuated metastatic melanoma progression, in addition, restoration of T-bet in T-bet-null Th1 cells certainly retrieved antimetastatic activity. These results suggest that T-bet expression in T cells is crucial for the control of tumor development and antimetastatic activity.
Collapse
Affiliation(s)
- Kihyun Lee
- College of Pharmacy, Division of Life and Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, Ewha Womans University, Seoul, Korea
| | | | | | | | | |
Collapse
|
135
|
Abstract
Naive CD4 cells are capable of integrating signals from antigen-activated cells of the innate immune system and differentiating into effector CD4 cells, also termed T helper (Th) cells. According to the traditional paradigm explaining adaptive CD4 cell responses, there are two subsets of Th cells: the Th-1 and Th-2 subset. Each of these subsets undergoes a distinct differentiation pathway (a pathway that is characterized by a unique profile of cytokine production and has specific immunoregulatory functions). However, recent studies in mouse models have forwarded evidence of a third subset of Th cells: the Th-17 subset. As indicated predominantly in studies on mice, the Th-17 subset is characterized by an ability to produce the neutrophil-mobilizing cytokine IL-17 in response to stimulation with the cytokine IL-23, an IL-12-related cytokine released from antigen-presenting cells. There is now a growing body of evidence from animal models that the Th-17 subset plays an important role in host defence in the lungs and other organs. Altered IL-17 levels have also been demonstrated in human patients with asthma, exacerbations of cystic fibrosis or following lung transplantation. There is now also evidence that the Th-17 subset is functionally distinct from the Th-2 subset but little is known of the functional inter-relationship between the Th-1 and Th-17 cell subsets; this is particularly true in human lungs. It has been proposed that the Th-17 subset plays a unique role by linking the arms of innate and adaptive immunity. Thus, an improved understanding of the human correlate to the Th-17 subset may reveal new targets for pharmacotherapy against lung disorders that are characterized by aberrant innate responses in host defense.
Collapse
Affiliation(s)
- Stefan Ivanov
- Lung Immunology & Pharmacology Groups, Department of Internal Medicine/Respiratory Medicine and Allergology, Institute of Medicine Sahlgrenska Academy at Göteborg University, Guldhedsgatan 10 A, S-413 46 Gothenburg, Sweden.
| | | |
Collapse
|
136
|
Abstract
The third lineage of T helper subsets, Th17, has recently been identified as an IL- 17-producing CD4+ Th cell, and its functions and regulatory mechanisms have been extensively characterized in immune responses. Functional studies have provided evidence that Th17 cells are important for the modulation of autoimmune responses, such as chronic asthma, rheumatoid arthritis, inflammatory bowel diseases, and multiple sclerosis. Murine Th17 cell differentiation is enhanced by the coordinated functions of distinct cytokines including TGFbeta, IL-6, IL-21, and IL-23, whereas IL-2, IL-4, IFNgamma, and IL-27 inhibit its differentiation. In addition, Th17 cells are controlled by several transcription factors such as RORgammat, IRF4, BATF, FoxP3, T-bet, PPARgamma, E-FABP, and SOCSs. This review focuses on the functions and regulatory mechanisms of several transcription factors in the control of Th17 cell differentiation.
Collapse
Affiliation(s)
- Eun Sook Hwang
- Division of Life and Pharmaceutical Sciences, College of Phamacy, Ewha Womans University, Center for Cell Signaling and Drug Discovery Research, 11-1 Daehyeon-dong, Seodaemun-gu, Seoul 120-750, Korea.
| |
Collapse
|
137
|
Obaculactone suppresses Th1 effector cell function through down-regulation of T-bet and prolongs skin graft survival in mice. Biochem Pharmacol 2010; 80:218-25. [DOI: 10.1016/j.bcp.2010.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 03/22/2010] [Accepted: 03/24/2010] [Indexed: 11/20/2022]
|
138
|
Ingram G, Hakobyan S, Hirst CL, Harris CL, Pickersgill TP, Cossburn MD, Loveless S, Robertson NP, Morgan BP. Complement regulator factor H as a serum biomarker of multiple sclerosis disease state. Brain 2010; 133:1602-11. [PMID: 20421219 DOI: 10.1093/brain/awq085] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025] Open
Abstract
Multiple sclerosis has a variable phenotypic presentation and subsequent disease course that, although unpredictable at disease onset, is of crucial importance in guiding interventions. Effective and accessible biomarkers are required in order to stratify patients and inform treatment. We examined whether the complement regulator factor H and its Tyr402His polymorphism, recently implicated as biomarkers in other chronic inflammatory central nervous system conditions, might identify or predict specific pathological processes and outcomes in multiple sclerosis. Employing novel assays, we measured factor H and its His402 variant in serum from 350 patients with multiple sclerosis classified according to disease course and relapse status. Serum factor H levels were significantly higher in progressive disease (P < 0.001) compared to controls and relapsing patients, after controlling for variables including disease duration, age, gender, disability and treatment. Serum factor H levels were capable of distinguishing secondary progressive from relapsing remitting disease (excluding patients in clinical relapse) with a sensitivity of 89.41%, specificity of 69.47% and a positive predictive value of 72.38%. Acute relapse was also associated with transiently increased factor H levels (P = 0.009) compared to stable relapsing disease. In clinically stable patients, factor H levels remained constant over 1 year (coefficient of variation percentage = 6.8), however, in patients in transition from relapsing to progressive disease, factor H levels significantly increased over a period of 2 years (P = 0.007). Concentration of the His402 variant in heterozytgotes was significantly higher in secondary progressive (P < 0.01) and primary progressive (P < 0.05) disease, suggesting altered expression or consumption of variants when factor H is upregulated. Serum factor H may be an effective indicator of progression and a practical and accessible biomarker and stratifying tool in determining disease course, providing objective evidence to help guide therapeutic decisions.
Collapse
Affiliation(s)
- Gillian Ingram
- Department of Neurosciences, Cardiff University, Cardiff CF14 4XN, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
139
|
El-behi M, Rostami A, Ciric B. Current views on the roles of Th1 and Th17 cells in experimental autoimmune encephalomyelitis. J Neuroimmune Pharmacol 2010; 5:189-97. [PMID: 20107924 PMCID: PMC2866798 DOI: 10.1007/s11481-009-9188-9] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 12/16/2009] [Indexed: 01/09/2023]
Abstract
Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are autoimmune demyelinating diseases of the central nervous system (CNS). Interferon-gamma-producing Th1 and interleukin-17-producing Th17 CD4(+) T helper (Th) cells mediate disease pathogenesis in EAE and likely in MS as well. However, the relative contribution of each Th subset to autoimmune processes in the CNS remains unclear. Emerging data suggest that both Th1 and Th17 cells contribute to CNS autoimmunity, albeit through different mechanisms. A better understanding of the roles that Th1 and Th17 cells play in autoimmune inflammation will be helpful in developing new therapeutic approaches. In this review, we discuss recent findings on the roles of Th1 and Th17 cells in the pathogenesis of EAE.
Collapse
Affiliation(s)
- Mohamed El-behi
- Department of Neurology, Thomas Jefferson University, Ste. 300 JHN, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Ste. 300 JHN, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Ste. 300 JHN, 900 Walnut Street, Philadelphia, PA 19107, USA
| |
Collapse
|
140
|
Boniface K, Blumenschein WM, Brovont-Porth K, McGeachy MJ, Basham B, Desai B, Pierce R, McClanahan TK, Sadekova S, de Waal Malefyt R. Human Th17 cells comprise heterogeneous subsets including IFN-gamma-producing cells with distinct properties from the Th1 lineage. THE JOURNAL OF IMMUNOLOGY 2010; 185:679-87. [PMID: 20511558 DOI: 10.4049/jimmunol.1000366] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Th17 cells have been named after their signature cytokine IL-17 and accumulating evidence indicates their involvement in the induction and progression of inflammatory diseases. In addition to IL-17 single-producing T cells, IL-17/IFN-gamma double-positive T cells are found in significantly elevated numbers in inflamed tissues or blood from patients with chronic inflammatory disorders. Because IFN-gamma is the classical Th1-associated cytokine, the origin and roles of these subsets remain elusive. In this paper, we show that not only IL-17(+)/IFN-gamma(+) but also IFN-gamma(+) (IL-17(-)) cells arise under Th17-inducing condition and have distinct properties from the Th1 lineage. In fact, these populations displayed characteristics reminiscent to IL-17 single-producing cells, including production of IL-22, CCL20, and induction of antimicrobial gene expression from epithelial cells. Live sorted IL-17(+) and Th17-IFN-gamma(+) cells retained expression of IL-17 or IFN-gamma after culture, respectively, whereas the IL-17(+)/IFN-gamma(+) population was less stable and could also become IL-17 or IFN-gamma single-producing cells. Interestingly, these Th17 subsets became "Th1-like" cells in the presence of IL-12. These results provide novel insights into the relationship and functionality of the Th17 and Th1 subsets and have direct implications for the analysis and relevance of IL-17 and/or IFN-gamma-producing T cells present in patients' peripheral blood and inflamed tissues.
Collapse
Affiliation(s)
- Katia Boniface
- Department of Immunology, Schering-Plough Biopharma, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Huss DJ, Winger RC, Peng H, Yang Y, Racke MK, Lovett-Racke AE. TGF-beta enhances effector Th1 cell activation but promotes self-regulation via IL-10. THE JOURNAL OF IMMUNOLOGY 2010; 184:5628-36. [PMID: 20393141 DOI: 10.4049/jimmunol.1000288] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myelin-specific effector Th1 cells are able to perpetuate CNS inflammation in experimental autoimmune encephalomyelitis, an animal model representative of multiple sclerosis. Although the effects of cytokines in the CNS microenvironment on naive CD4(+) T cells have been well described, much less is known about their ability to influence Ag-experienced effector cells. TGF-beta is a multifunctioning cytokine present in the healthy and inflamed CNS with well-characterized suppressive effects on naive T cell functions. However, the effects of TGF-beta on effector Th1 cells are not well defined. Using myelin-specific TCR transgenic mice, we demonstrate that TGF-beta elicits differential effects on naive versus effector Th1 cells. TGF-beta enhances cellular activation, proliferation, and cytokine production of effector Th1 cells; however, adoptive transfer of these cells into naive mice showed a reduction in encephalitogenicity. We subsequently demonstrate that the reduced encephalitogenic capacity is due to the ability of TGF-beta to promote the self-regulation of Th1 effector cells via IL-10 production. These data demonstrate a mechanism by which TGF-beta is able to suppress the encephalitogenicity of myelin-specific Th1 effector cells that is unique from its suppression of naive T cells.
Collapse
Affiliation(s)
- David J Huss
- Department of Molecular Virology, Immunology, and Medical Genetics, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
142
|
Yang Y, Liu Y, Wei P, Peng H, Winger R, Hussain RZ, Ben LH, Cravens PD, Gocke AR, Puttaparthi K, Racke MK, McTigue DM, Lovett-Racke AE. Silencing Nogo-A promotes functional recovery in demyelinating disease. Ann Neurol 2010; 67:498-507. [PMID: 20437585 PMCID: PMC2929680 DOI: 10.1002/ana.21935] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To determine if suppressing Nogo-A, an axonal inhibitory protein, will promote functional recovery in a murine model of multiple sclerosis (MS). METHODS A small interfering RNA was developed to specifically suppress Nogo-A (siRNA-NogoA). The siRNA-NogoA silencing effect was evaluated in vitro and in vivo via immunohistochemistry. The siRNA was administered intravenously in 2 models of experimental autoimmune encephalomyelitis (EAE). Axonal repair was measured by upregulation of GAP43. Enzyme-linked immunosorbent assay, flow cytometry, and (3)H-thymidine incorporation were used to determine immunological changes in myelin-specific T cells in mice with EAE. RESULTS The siRNA-NogoA suppressed Nogo-A expression in vitro and in vivo. Systemic administration of siRNA-NogoA ameliorated EAE and promoted axonal repair, as demonstrated by enhanced GAP43+ axons in the lesions. Myelin-specific T-cell proliferation and cytokine production were unchanged in the siRNA-NogoA-treated mice. INTERPRETATION Silencing Nogo-A in EAE promotes functional recovery. The therapeutic benefit appears to be mediated by axonal growth and repair, and is not attributable to changes in the encephalitogenic capacity of the myelin-specific T cells. Silencing Nogo-A may be a therapeutic option for MS patients to prevent permanent functional deficits caused by immune-mediated axonal damage.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Enzyme-Linked Immunosorbent Assay/methods
- Flow Cytometry/methods
- GAP-43 Protein/genetics
- GAP-43 Protein/metabolism
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/genetics
- Glycoproteins/adverse effects
- Interferon-gamma/metabolism
- Interleukin-10/metabolism
- Lymphocytes/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Myelin Basic Protein/genetics
- Myelin Basic Protein/pharmacology
- Myelin Proteins/genetics
- Myelin Proteins/metabolism
- Myelin-Oligodendrocyte Glycoprotein
- Neuroblastoma
- Nogo Proteins
- Peptide Fragments/adverse effects
- Peptide Fragments/genetics
- Peptide Fragments/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/therapeutic use
- Spinal Cord/metabolism
- Tetradecanoylphorbol Acetate/analogs & derivatives
- Tetradecanoylphorbol Acetate/pharmacology
- Transfection/methods
Collapse
Affiliation(s)
- Yuhong Yang
- Department of Neurology, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Leng RX, Pan HF, Chen GM, Wang C, Qin WZ, Chen LL, Tao JH, Ye DQ. IL-23: A Promising Therapeutic Target for Systemic Lupus Erythematosus. Arch Med Res 2010; 41:221-5. [DOI: 10.1016/j.arcmed.2010.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 02/05/2010] [Indexed: 12/18/2022]
|
144
|
|
145
|
Reynolds AD, Stone DK, Hutter JAL, Benner EJ, Mosley RL, Gendelman HE. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson's disease. THE JOURNAL OF IMMUNOLOGY 2010; 184:2261-71. [PMID: 20118279 DOI: 10.4049/jimmunol.0901852] [Citation(s) in RCA: 307] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nitrated alpha-synuclein (N-alpha-syn) immunization elicits adaptive immune responses to novel antigenic epitopes that exacerbate neuroinflammation and nigrostriatal degeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease. We show that such neuroimmune degenerative activities, in significant measure, are Th17 cell-mediated, with CD4(+)CD25(+) regulatory T cell (Treg) dysfunction seen among populations of N-alpha-syn-induced T cells. In contrast, purified vasoactive intestinal peptide induced and natural Tregs reversed N-alpha-syn T cell nigrostriatal degeneration. Combinations of adoptively transferred N-alpha-syn and vasoactive intestinal peptide immunocytes or natural Tregs administered to MPTP mice attenuated microglial inflammatory responses and led to robust nigrostriatal protection. Taken together, these results demonstrate Treg control of N-alpha-syn-induced neurodestructive immunity and, as such, provide a sound rationale for future Parkinson's disease immunization strategies.
Collapse
Affiliation(s)
- Ashley D Reynolds
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | |
Collapse
|
146
|
Tang Y, Desierto MJ, Chen J, Young NS. The role of the Th1 transcription factor T-bet in a mouse model of immune-mediated bone-marrow failure. Blood 2010; 115:541-8. [PMID: 19903901 PMCID: PMC2810980 DOI: 10.1182/blood-2009-03-211383] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 10/15/2009] [Indexed: 12/25/2022] Open
Abstract
The transcription factor T-bet is a key regulator of type 1 immune responses. We examined the role of T-bet in an animal model of immune-mediated bone marrow (BM) failure using mice carrying a germline T-bet gene deletion (T-bet(-/-)). In comparison with normal C57BL6 (B6) control mice, T-bet(-/-) mice had normal cellular composition in lymphohematopoietic tissues, but T-bet(-/-) lymphocytes were functionally defective. Infusion of 5 x 10(6) T-bet(-/-) lymph node (LN) cells into sublethally irradiated, major histocompatibility complex-mismatched CByB6F1 (F1) recipients failed to induce the severe marrow hypoplasia and fatal pancytopenia that is produced by injection of similar numbers of B6 LN cells. Increasing T-bet(-/-) LN-cell dose to 10 to 23 x 10(6) per recipient led to only mild hematopoietic deficiency. Recipients of T-bet(-/-) LN cells had no expansion in T cells or interferon-gamma-producing T cells but showed a significant increase in Lin(-)Sca1(+)CD117(+)CD34(-) BM cells. Plasma transforming growth factor-beta and interleukin-17 concentrations were increased in T-bet(-/-) LN-cell recipients, possibly a compensatory up-regulation of the Th17 immune response. Continuous infusion of interferon-gamma resulted in hematopoietic suppression but did not cause T-bet(-/-) LN-cell expansion or BM destruction. Our data provided fresh evidence demonstrating a critical role of T-bet in immune-mediated BM failure.
Collapse
Affiliation(s)
- Yong Tang
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
147
|
Fong DJ, Hogaboam CM, Matsuno Y, Akira S, Uematsu S, Joshi AD. Toll-like receptor 6 drives interleukin-17A expression during experimental hypersensitivity pneumonitis. Immunology 2010; 130:125-36. [PMID: 20070409 DOI: 10.1111/j.1365-2567.2009.03219.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hypersensitivity pneumonitis (HP) is a T-cell-driven disease that is histologically characterized by diffuse mononuclear cell infiltrates and loosely formed granulomas in the lungs. We have previously reported that interleukin-17A (IL-17A) contributes to the development of experimental HP, and that the pattern recognition receptor Toll-like receptor 6 (TLR6) might be a factor in the initiation of this response. Using a well-established murine model of Saccharopolyspora rectivirgula-induced HP, we investigated the role of TLR6 in the immunopathogenesis of this disease. In the absence of TLR6 signalling, mice that received multiple challenges with S. rectivirgula-antigen (SR-Ag) had significantly less lung inflammation compared with C57BL/6 mice (wild-type; WT) similarly challenged with SR-Ag. Flow cytometric analysis of whole lung samples from SR-Ag-challenged mice showed that TLR6(-/-) mice had a decreased CD4(+) : CD8(+) T-cell ratio compared with WT mice. Cytokine analysis at various days after the final SR-Ag challenge revealed that whole lungs from TLR6(-/-) mice contained significantly less IL-17A than lungs from WT mice with HP. The IL-17A-driving cytokines IL-21 and IL-23 were also expressed at lower levels in SR-Ag-challenged TLR6(-/-) mice, when compared with SR-Ag-challenged WT mice. Other pro-inflammatory cytokines, namely interferon-gamma and RANTES, were also found to be regulated by TLR6 signalling. Anti-TLR6 neutralizing antibody treatment of dispersed lung cells significantly impaired SR-Ag-induced IL-17A and IL-6 generation. Together, these results indicate that TLR6 plays a pivotal role in the development and severity of HP via its role in IL-17A production.
Collapse
Affiliation(s)
- Daniel J Fong
- Immunology Program, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0602, USA
| | | | | | | | | | | |
Collapse
|
148
|
Abstract
Autoreactive effector CD4+ T cells have been associated with the pathogenesis of autoimmune disorders. Early studies implicated the interferon (IFN)-gamma-producing T helper (Th)1 subset of CD4+ cells as the causal agents in the pathogenesis of autoimmunity. However, further studies have suggested a more complex story. In models thought to be driven by Th1 cells, mice lacking the hallmark Th1 cytokine IFN-gamma were not protected but tended to have enhanced susceptibility to disease. Identification of the IL-17-producing CD4+ effector cell lineage (Th17) has helped shed light on this issue. Th17 effector cells are induced in parallel to Th1, and, like Th1, polarized Th17 cells have the capacity to cause inflammation and autoimmune disease. This, together with the finding that deficiency of the Th17-related cytokine IL-23 but not the Th1-related cytokine IL-12 causes resistance, led to the notion that Th17 cells are the chief contributors to autoimmune tissue inflammation. Nevertheless, mice lacking IL-17 are not protected from disease and display elevated numbers of IFN-gamma-producing CD4+ T cells, and, in some cases, lack of IFN-gamma does confer resistance. Recent studies report overlapping as well as differential roles of these cells in tissue inflammation, which suggests the existence of a more complex relationship between these two effector T-cell subsets than has hitherto been suspected. This review will attempt to bring together current information regarding interaction, balance, and collaborative potential between the Th1 and Th17 effector lineages.
Collapse
Affiliation(s)
- Jesse M Damsker
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892-1857, USA
| | | | | |
Collapse
|
149
|
Doodes PD, Cao Y, Hamel KM, Wang Y, Rodeghero RL, Mikecz K, Glant TT, Iwakura Y, Finnegan A. IFN-gamma regulates the requirement for IL-17 in proteoglycan-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2009; 184:1552-9. [PMID: 20028652 DOI: 10.4049/jimmunol.0902907] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The contribution of the proinflammatory cytokines IFN-gamma and IL-17 to the pathogenesis of experimental arthritis is controversial. In proteoglycan (PG)-induced arthritis (PGIA), severe arthritis is dependent on the production of IFN-gamma, whereas IL-17 is dispensable. In collagen-induced arthritis and Ag-induced arthritis, although high levels of IFN-gamma are secreted, disease is exacerbated in IFN-gamma or IFN-gamma receptor-deficient mice due to the ability of IFN-gamma to suppress IL-17 expression. In the current study, we investigated the effect of IFN-gamma on the IL-17 response and its consequences in PGIA. In PG-immunized IFN-gamma(-/-) mice, despite reduction in arthritis, the PG-specific CD4(+) T cell IL-17 response was significantly increased. Elevated IL-17 contributed to development of arthritis, as disease in IFN-gamma/IL-17(-/-) was significantly reduced in comparison with either IFN-gamma(-/-) or IL-17(-/-) mice. A contribution of IFN-gamma and IL-17 to the development of arthritis was also identified in T-bet(-/-) mice. PG-specific CD4(+) T cells from T-bet(-/-) mice produced reduced IFN-gamma and elevated concentrations of IL-17. Both IFN-gamma and IL-17 contribute to arthritis, as T-bet(-/-) mice lacking IL-17 (T-bet/IL-17(-/-)) were resistant, whereas wild-type, T-bet(-/-), and IL-17(-/-) mice were susceptible to PGIA. T cell proliferation and autoantibody production did not correlate with development of disease; however, expression of cytokines and chemokines in joint tissues demonstrate that IFN-gamma and IL-17 cooperatively contribute to inflammation. These results demonstrate that both IFN-gamma and IL-17 have the potential to induce PGIA, but it is the strength of the IFN-gamma response that regulates the contribution of each of these Th effector cytokines to disease.
Collapse
Affiliation(s)
- Paul D Doodes
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Michael S, Warstat C, Michel F, Yan L, Müller CE, Nieber K. Adenosine A(2A) agonist and A(2B) antagonist mediate an inhibition of inflammation-induced contractile disturbance of a rat gastrointestinal preparation. Purinergic Signal 2009; 6:117-24. [PMID: 20020217 DOI: 10.1007/s11302-009-9174-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Accepted: 12/01/2009] [Indexed: 12/22/2022] Open
Abstract
Adenosine can show anti-inflammatory as well as pro-inflammatory activities. The contribution of the specific adenosine receptor subtypes in various cells, tissues and organs is complex. In this study, we examined the effect of the adenosine A(2A) receptor agonist CGS 21680 and the A(2B)R antagonist PSB-1115 on acute inflammation induced experimentally by 2,4,6-trinitrobenzenesulfonic acid (TNBS) on rat ileum/jejunum preparations. Pre-incubation of the ileum/jejunum segments with TNBS for 30 min resulted in a concentration-dependent inhibition of acetylcholine (ACh)-induced contractions. Pharmacological activation of the A(2A)R with CGS 21680 (0.1-10 microM) pre-incubated simultaneously with TNBS (10 mM) prevented concentration-dependently the TNBS-induced inhibition of the ACh contractions. Stimulation of A(2B)R with the selective agonist BAY 60-6583 (10 microM) did neither result in an increase nor in a further decrease of ACh-induced contractions compared to the TNBS-induced inhibition. The simultaneous pre-incubation of the ileum/jejunum segments with TNBS (10 mM) and the selective A(2B)R antagonist PSB-1115 (100 microM) inhibited the contraction-decreasing effect of TNBS. The effects of the A(2A)R agonist and the A(2B)R antagonist were in the same range as the effect induced by 1 microM methotrexate. The combination of the A(2A)R agonist CGS 21680 and the A(2B)R antagonist PSB-1115 at subthreshold concentrations of both agents found a significant amelioration of the TNBS-diminished contractility. Our results demonstrate that the activation of A(2A) receptors or the blockade of the A(2B) receptors can prevent the inflammation-induced disturbance of the ACh-induced contraction in TNBS pre-treated small intestinal preparations. The combination of both may be useful for the treatment of inflammatory bowel diseases.
Collapse
|