201
|
Newcomb DC, Zhou W, Moore ML, Goleniewska K, Hershey GKK, Kolls JK, Peebles RS. A functional IL-13 receptor is expressed on polarized murine CD4+ Th17 cells and IL-13 signaling attenuates Th17 cytokine production. THE JOURNAL OF IMMUNOLOGY 2009; 182:5317-21. [PMID: 19380778 DOI: 10.4049/jimmunol.0803868] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-17A is produced from Th17 cells, and is involved in many autoimmune and inflammatory diseases. IL-13R has not previously been reported to be functionally expressed on T cells; however, we found that purified BALB/c CD4(+) cells polarized to Th17 with TGF-beta, IL-6, and IL-23 have increased mRNA and protein expression of IL-13R alpha1 and mRNA expression of IL-4R alpha compared with Th0, Th1, or Th2 polarized cells. The addition of IL-13 at Th17 polarization negatively regulated IL-17A and IL-21 expression, and reduced the number of CD4(+) T cells producing IL-17A. Further, adding IL-13 at the time of Th17 cell restimulation attenuated IL-17A expression. CD4(+) Th17 polarized cells from IL-4 knockout (KO) mice also had IL-13-induced inhibition of IL-17A production, but this was not observed in IL-4R KO and STAT6 KO mice. Addition of IL-13 at polarization increased IL-13R expression in wild-type Th17 cells. Further, IL-13 administration during Th17 polarization down-regulated retinoic acid-related-gammaT, the transcription required for Th17 development; increased STAT6 phosphorylation, and up-regulated GATA3, the transcription factor activated during the development of Th2 cells. This IL-13-mediated effect was specific to Th17 cells as IL-13 neither decreased IFN-gamma expression by Th1 cells nor affected Th2 cell production of IL-4. Collectively, we have shown that Th17 cells express a functional IL-13R and that IL-13 negatively regulates IL-17A and IL-21 production by decreasing retinoic acid-related-gammaT expression and while increasing phosphorylation of STAT6 and GATA3 expression. Therefore, therapeutic intervention inhibiting IL-13 production could have adverse consequences by up-regulating Th17 inflammation in certain disease states.
Collapse
Affiliation(s)
- Dawn C Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
202
|
Yoo J, Jang SI, Kim S, Cho JH, Lee HJ, Rhee MH, Lillehoj HS, Min W. Molecular characterization of duck interleukin-17. Vet Immunol Immunopathol 2009; 132:318-22. [PMID: 19573930 DOI: 10.1016/j.vetimm.2009.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Revised: 05/28/2009] [Accepted: 06/03/2009] [Indexed: 11/30/2022]
Abstract
Interleukin-17 (IL17), belonging to the Th17 family, is a proinflammatory cytokine produced by activated T cells. A 1034bp cDNA encoding duck IL17 (duIL17) was cloned from Con A-activated splenic lymphocytes of ducks. The encoded protein, which is predicted to consist of 169 amino acids, has a molecular weight of 18.8kDa and includes a 29 residue NH(2)-terminal signal peptide, a single potential N-linked glycosylation site, and six cysteine residues that are conserved in mammalian IL17. The duIL17 shared 84% amino acid sequence identity with the previously described chicken IL17 (chIL17), 36-47% to mammalian homologues, and open reading frame 13 of Herpesvirus saimiri (HVS13). The genomic structure of duIL17 was quite similar to its chicken and mammalian counterparts. The duIL17 mRNA expression was detected only in Con A-activated splenic lymphocytes by RT-PCR, although its expression was undetectable in a variety of normal tissues. Two mAbs against chIL17 showed cross-reactivity with duIL17 as detected by indirect ELISA and Western blot analysis. These findings indicate that the structure of IL17 is highly conserved among poultry, and two mAbs detecting common epitopes of IL17 are available for molecular and immunological studies of IL17 in birds.
Collapse
Affiliation(s)
- Jeongmi Yoo
- College of Veterinary Medicine & Research Institute of Life Science, Gyeongsang National University, 900 Gajwa-dong, Jinju, Gyeongnam 660-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Abstract
IL-23 belongs to IL-12 family and is composed of IL-23 P19 and IL-12 P40 subunits. IL-23 functions through a receptor complex consisting of an IL-12Rβ1 and a specific IL-23R chain in T cells, induces activation of Stat1, Stat3 and Stat4, and triggers T cells to produce IL-10 and INF-γ. Additionally, IL-23 induces the differentiation of native CD4+ T cells into highly pathogenic Th17 cells that produce IL-17, IL-6 and TNF-α, and subsequently causes chronic colitis. Recent studies have demonstrated that the IL-23/IL-17 axis plays a critical role in the pathogenesis of inflammatory bowel disease (IBD) and may represent a target for therapeutic intervention for IBD.
Collapse
|
204
|
Hofer U, Speck RF. Disturbance of the gut-associated lymphoid tissue is associated with disease progression in chronic HIV infection. Semin Immunopathol 2009; 31:257-66. [PMID: 19484240 DOI: 10.1007/s00281-009-0158-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 05/14/2009] [Indexed: 02/06/2023]
Abstract
Why and how HIV makes people sick is highly debated. Recent evidence implicates heightened immune activation due to breakdown of the gastrointestinal barrier as a determining factor of lentiviral pathogenesis. HIV-mediated loss of Th17 cells from the gut-associated lymphoid tissue (GALT) impairs mucosal integrity and innate defense mechanisms against gut microbes. Translocation of microbial products from the gut, in turn, correlates with increased immune activation in chronic HIV infection and may further damage the immune system by increasing viral and activation-induced T cell death, by reducing T cell reconstitution due to tissue scarring, and by impairing the function of other cell types, such as gammadelta T cells and epithelial cells. Maintaining a healthy GALT may be the key to reducing the pathogenic potential of HIV.
Collapse
Affiliation(s)
- Ursula Hofer
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
205
|
Nembrini C, Marsland BJ, Kopf M. IL-17-producing T cells in lung immunity and inflammation. J Allergy Clin Immunol 2009; 123:986-94; quiz 995-6. [PMID: 19410688 DOI: 10.1016/j.jaci.2009.03.033] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 03/26/2009] [Accepted: 03/26/2009] [Indexed: 11/19/2022]
Abstract
T(H)17 cells are a recently described effector CD4 T-cell subset characterized by the production of IL-17A, IL-17F, and IL-22, which have been implicated in the pathogenesis of several autoimmune diseases. T(H)17 and other IL-17A-producing T cells, including a population of gammadelta T cells and natural killer T cells, have also been associated with the development of skin, intestinal, and lung inflammatory diseases, such as asthma, granulomatous disease, chronic obstructive pulmonary disease, and cystic fibrosis. On the other hand, IL-17-producing T cells play important roles in protective immunity against some bacterial infections, mainly through the recruitment and activation of neutrophils. Thus, their regulation appears to be critical, and excess or deficient IL-17 elaboration leads either to deficient responses or disease. This review will summarize T(H)17 cell differentiation and discuss the host beneficial and detrimental function of IL-17A and related cytokines produced by different subpopulations of T cells.
Collapse
Affiliation(s)
- Chiara Nembrini
- Institute of Integrative Biology, Molecular Biomedicine, ETH, Zurich, Switzerland
| | | | | |
Collapse
|
206
|
Jafarzadeh A, Mirzaee V, Ahmad-Beygi H, Nemati M, Rezayati MT. Association of the CagA status of Helicobacter pylori and serum levels of interleukin (IL)-17 and IL-23 in duodenal ulcer patients. J Dig Dis 2009; 10:107-12. [PMID: 19426392 DOI: 10.1111/j.1751-2980.2009.00371.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE It has been reported that the cytotoxin-associated gene A (cagA+) H. pylori strains induce severe gastric mucosal inflammation. The aim of this study was to investigate the association of the virulence factor CagA with IL-17 and IL-23 serum levels in duodenal ulcer (DU) patients and H. pylori-infected asymptomatic (AS) carriers. METHODS In total, 45 H. pylori-infected DU patients were enrolled to study: 23 tested positive for the anti-CagA antibody (anti-CagA+) and 22 tested negative for the anti-CagA antibody (anti-CagA-), 30 were AS carriers (15 were anti-CagA+ and 15 were anti-CagA-) and 15 were healthy uninfected participants (as a control group). The IL-17 and IL-23 serum levels of participants were measured by enzyme-linked immunosorbent assay method. RESULTS The mean IL-17 levels in DU patients were significantly higher than those in AS and control groups (P < 0.001 and P < 0.0001 respectively). In the DU group, the mean IL-17 levels in participants testing positive for anti-CagA (10.84 +/- 5.79 pg/mL) were significantly higher than those observed in participants testing negative for anti-CagA (7.65 +/- 4.74 pg/mL; P < 0.05). The mean IL-23 levels in the DU and AS groups were significantly higher than in the control group (P < 0.02 and P < 0.03 respectively) but were not significantly different in participants testing positive and negative for anti-CagA. CONCLUSION These results showed higher IL-17 and IL-23 serum levels in H. pylori-infected participants than in the control group. In the DU group the expression of IL-17 was influenced by the CagA factor.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, Medical School, Rafsanjan University of Medical Sciences and Health Services, Rafsanjan, Iran.
| | | | | | | | | |
Collapse
|
207
|
Marks BR, Craft J. Barrier immunity and IL-17. Semin Immunol 2009; 21:164-71. [PMID: 19386512 DOI: 10.1016/j.smim.2009.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Accepted: 03/17/2009] [Indexed: 01/14/2023]
Abstract
CD4+ T(H)17 cells display a featured role in barrier immunity. This effector population of T cells is important for clearance of microorganisms but can also promote autoimmunity at barrier sites. Recent work has indicated that these effector cells share a pathway with CD4+ regulatory T cells (T(R) cells) that also have a critical function in barrier protection and immune regulation. The development and function of T(H)17 cells, and their relationship with T(R) cells are discussed.
Collapse
Affiliation(s)
- Benjamin R Marks
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
208
|
Mucida D, Salek-Ardakani S. Regulation of TH17 cells in the mucosal surfaces. J Allergy Clin Immunol 2009; 123:997-1003. [PMID: 19362732 DOI: 10.1016/j.jaci.2009.03.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 02/27/2009] [Accepted: 03/09/2009] [Indexed: 01/06/2023]
Abstract
The mucosal surfaces represent the main intersection between jawed vertebrates and the environment. The mucosal surface of the intestine alone forms the largest surface that is exposed to exogenous antigens as well as the largest collection of lymphoid tissue in the body. Therefore, a protective immune activity must coexist with efficient regulatory mechanisms to maintain a health status of these organisms. The discovery of a new lineage of T(H) cells that produce IL-17 has provided valuable new insight into host defense and the pathogenesis of inflammatory diseases at the mucosal surfaces. Of particular interest for these surfaces, it has been reported that peripherally-induced regulatory T cells and T(H)17 effector cells arise in a mutually exclusive fashion, depending on whether they are activated in the presence of TGF-beta or TGF-beta plus inflammatory cytokines such as IL-6. This review addresses the protective and pathogenic roles of T(H)17 cells in the mucosal surfaces and potential regulatory mechanisms that control their development.
Collapse
Affiliation(s)
- Daniel Mucida
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, Calif 92037, USA.
| | | |
Collapse
|
209
|
Curtis MM, Way SS. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology 2009; 126:177-85. [PMID: 19125888 DOI: 10.1111/j.1365-2567.2008.03017.x] [Citation(s) in RCA: 372] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The mammalian immune system is intricately regulated, allowing for potent pathogen-specific immunity to be rapidly activated in response to infection with a broad and diverse array of potential pathogens. As a result of their ability to differentiate into distinct effector lineages, CD4 T cells significantly contribute to pathogen-specific adaptive immune responses. Through the production of effector cytokines, CD4 T helper (Th) cells orchestrate the precise mobilization of specific immune cells to eradicate infection. The protective effects of the newly identified lineage of Th17 cells against pathogens like Klebsiella pneumoniae, Citrobacter rodentium and Candida albicans indicate the capacity of Th17 cells to confer protection against extracellular bacterial and fungal pathogens, filling a critical void in host immunity not covered by the classically described Th1 lineage that activates immunity to intracellular pathogens or the Th2 lineage that is important in protection against mucosal parasitic pathogens. Host defence by Th17 cells extends beyond protection against extracellular bacterial and fungal pathogens, as demonstrated in infections against intracellular bacteria like Listeria monocytogenes and Salmonella enterica, as well as Mycobacterium tuberculosis. Herein, we summarize both experimental data from mouse infection models and epidemiological studies in humans that demonstrate the protective effects of interleukin-17 and Th17 CD4 T cells in immunity to bacterial, mycobacterial and fungal pathogens.
Collapse
Affiliation(s)
- Meredith M Curtis
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | | |
Collapse
|
210
|
Abstract
CD4+ T cells, upon activation and expansion, develop into different T helper cell subsets with different cytokine profiles and distinct effector functions. Until recently, T cells were divided into Th1 or Th2 cells, depending on the cytokines they produce. A third subset of IL-17-producing effector T helper cells, called Th17 cells, has now been discovered and characterized. Here, we summarize the current information on the differentiation and effector functions of the Th17 lineage. Th17 cells produce IL-17, IL-17F, and IL-22, thereby inducing a massive tissue reaction owing to the broad distribution of the IL-17 and IL-22 receptors. Th17 cells also secrete IL-21 to communicate with the cells of the immune system. The differentiation factors (TGF-β plus IL-6 or IL-21), the growth and stabilization factor (IL-23), and the transcription factors (STAT3, RORγt, and RORα) involved in the development of Th17 cells have just been identified. The participation of TGF-β in the differentiation of Th17 cells places the Th17 lineage in close relationship with CD4+CD25+Foxp3+ regulatory T cells (Tregs), as TGF-β also induces differentiation of naive T cells into Foxp3+ Tregs in the peripheral immune compartment. The investigation of the differentiation, effector function, and regulation of Th17 cells has opened up a new framework for understanding T cell differentiation. Furthermore, we now appreciate the importance of Th17 cells in clearing pathogens during host defense reactions and in inducing tissue inflammation in autoimmune disease.
Collapse
Affiliation(s)
- Thomas Korn
- Technical University Munich, Department of Neurology, 81675 Munich, Germany
| | - Estelle Bettelli
- Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115;,
| | - Mohamed Oukka
- Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Cambridge, Massachusetts 02139
| | - Vijay K. Kuchroo
- Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115;,
| |
Collapse
|
211
|
Abstract
IL-17 can impact health in a variety of ways. It is protective for some pathogens but it is also associated with tissue damaging inflammation. By examining the role of IL-17 in a variety of bacterial infections the mechanisms by which this cytokine mediates both protection and damage can be dissected. A key element in understanding the role of this cytokine is determining where and when it is acting. Dissecting its essential protective role from its immunopathologic role will allow for improved intervention in both acute and chronic disease.
Collapse
|
212
|
You QH, Sun GY, Wang N, Shen JL, Wang Y. Interleukin-17F-induced pulmonary microvascular endothelial monolayer hyperpermeability via the protein kinase C pathway. J Surg Res 2009; 162:110-21. [PMID: 19577259 DOI: 10.1016/j.jss.2009.01.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Revised: 01/06/2009] [Accepted: 01/13/2009] [Indexed: 01/05/2023]
Abstract
BACKGROUND Interleukin (IL)-17F is involved in lung inflammation, but the effect of IL-17F on endothelial permeability and its signaling pathway remain ill-defined. The current study sought to investigate the effect of IL-17F on endothelium and assess the role of protein kinase C (PKC) and src-suppressed C kinase substrate (SSeCKS) in this process. METHODS Rat pulmonary microvascular endothelial monolayers were constructed to determine changes of permeability as measured by means of FITC-dextran and Hank's solution flux across monolayers and transendothelial electrical resistance with or without IL-17F and PKC inhibitors. Additional monolayers were stained using FITC-phalloidin for filamentous actin (F-actin). The gene expression of SSeCKS was analyzed by the reverse transcription-polymerase chains. Alterations of SSeCKS protein were investigated by immunoblotting and immunoprecipitation. RESULTS IL-17F increased endothelial monolayer permeability in a dose- and time-dependent manner. F-actin staining revealed that permeability changes were accompanied by reorganization of cytoskeleton. In the presence of PKC inhibitors, the IL-17F-induced hyperpermeability and reorganization of F-actin were attenuated. The gene and protein expression of SSeCKS were conspicuously elevated after IL-17F challenge. The process of SSeCKS phosphorylation followed a time course that mirrored the time course of hyperpermeability induced by IL-17F. IL-17F-induced SSeCKS phosphorylation was abrogated after PKC inhibitors pretreatment. The translocation of SSeCKS from the cytosol to the membrane and a significant increase in the SSeCKS association with the cytoskeleton were found after IL-17F treatment. CONCLUSIONS IL-17F is an important mediator of increased endothelial permeability. PKC and SSeCKS are integral signaling components essential for IL-17F-induced hyperpermeability.
Collapse
Affiliation(s)
- Qing-hai You
- Department of Respiratory Medicine, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, China
| | | | | | | | | |
Collapse
|
213
|
Sieve AN, Meeks KD, Bodhankar S, Lee S, Kolls JK, Simecka JW, Berg RE. A novel IL-17-dependent mechanism of cross protection: respiratory infection with mycoplasma protects against a secondary listeria infection. Eur J Immunol 2009; 39:426-38. [PMID: 19180464 PMCID: PMC2735239 DOI: 10.1002/eji.200838726] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Immune responses to pathogens occur within the context of current and previous infections. Cross protection refers to the phenomena where infection with a particular pathogen provides enhanced resistance to a subsequent unrelated pathogen in an antigen-independent manner. Proposed mechanisms of antigen-independent cross protection have involved the secretion of IFN-gamma, which activates macrophages, thus providing enhanced innate immunity against the secondary viral or bacterial pathogen. Here we provide evidence that a primary infection with the chronic respiratory pathogen, Mycoplasma pulmonis, provides a novel form of cross protection against a secondary infection with Listeria monocytogenes that is not mediated by IFN-gamma, but instead relies upon IL-17 and mobilization of neutrophils. Mice infected with M. pulmonis have enhanced clearance of L. monocytogenes from the spleen and liver, which is associated with increased numbers of Gr-1(+)CD11b(+) cells and higher levels of IL-17. This enhanced clearance of L. monocytogenes was absent in mice depleted of Gr-1(+) cells or in mice deficient in the IL-17 receptor. Additionally, both the IL-17 receptor and neutrophils were essential for optimal clearance of M. pulmonis. Thus, a natural component of the immune response directed against M. pulmonis was able to enhance clearance of L. monocytogenes.
Collapse
Affiliation(s)
- Amy N. Sieve
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Karen D. Meeks
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Sheetal Bodhankar
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Suheung Lee
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Jay K. Kolls
- Division of Pulmonology, Department of Pediatrics, Children's Hospital of Pittsburgh and The University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Jerry W. Simecka
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | - Rance E. Berg
- Department of Molecular Biology and Immunology, The University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| |
Collapse
|
214
|
Wolf K, Plano GV, Fields KA. A protein secreted by the respiratory pathogen Chlamydia pneumoniae impairs IL-17 signalling via interaction with human Act1. Cell Microbiol 2009; 11:769-79. [PMID: 19159390 DOI: 10.1111/j.1462-5822.2009.01290.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chlamydia pneumoniae is a common respiratory pathogen that has been associated with a variety of chronic diseases including asthma and atherosclerosis. Chlamydiae are obligate intracellular parasites that primarily infect epithelial cells where they develop within a membrane-bound vacuole, termed an inclusion. Interactions between the microorganism and eukaryotic cell can be mediated by chlamydial proteins inserted into the inclusion membrane. We describe here a novel C. pneumoniae-specific inclusion membrane protein (Inc) CP0236, which contains domains exposed to the host cytoplasm. We demonstrate that, in a yeast two-hybrid screen, CP0236 interacts with the NFκB activator 1 (Act1) and this interaction was confirmed in HeLa 229 cells where ectopically expressed CP0236 was co-immunoprecipitated with endogenous Act1. Furthermore, we demonstrate that Act1 displays an altered distribution in the cytoplasm of HeLa cells infected with C. pneumoniae where it associates with the chlamydial inclusion membrane. This sequestration of Act1 by chlamydiae inhibited recruitment of the protein to the interleukin-17 (IL-17) receptor upon stimulation of C. pneumoniae-infected cells with IL-17A. Such inhibition of the IL-17 signalling pathway led to protection of Chlamydia-infected cells from NFκB activation in IL-17-stimulated cells. We describe here a unique strategy employed by C. pneumoniae to achieve inhibition of NFκB activation via interaction of CP0236 with mammalian Act1.
Collapse
Affiliation(s)
- Katerina Wolf
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | | | | |
Collapse
|
215
|
Dobbs NA, Odeh AN, Sun X, Simecka JW. THE MULTIFACETED ROLE OF T CELL-MEDIATED IMMUNITY IN PATHOGENESIS AND RESISTANCE TO MYCOPLASMA RESPIRATORY DISEASE. CURRENT TRENDS IN IMMUNOLOGY 2009; 10:1-19. [PMID: 21743780 PMCID: PMC3131222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Mycoplasma respiratory diseases have a significant impact on the economy, health and wildlife. The hallmark of these diseases is the persistence of the mycoplasma infections and chronic inflammatory responses associated with the airways. There is still much that needs to be understood about the immune mechanisms involved in mycoplasma disease and resistance from infection. It is clear that immune responses can contribute to the generation of inflammatory lesions in mycoplasma respiratory disease, as well as provide protection from infection and extrapulmonary dissemination of the organisms. The evolution of this lung disease is under the control innate immune mechanisms and the contrasting effects of different T cell populations. The mechanisms of immunity involved in mycoplasma diseases are multifaceted, and a fascinating story of its complexity is being uncovered. Research in mycoplasma respiratory diseases have underscored the idea that immunity along the respiratory tract against infectious agents is a dynamic process and involves a network of cellular and cytokine signals that determine the type of responses generated, and ultimately, the outcome of infection. The aim of this article is to present on overview of our work on mycoplasma disease and immunity, focusing on the interactions and regulation of T cell responses that influence disease pathogenesis. We will first provide an overview of immune mechanisms involved in controlling infection and participate in the generation of T cell responses, and the role of T cell populations in generating protection and contributing to lesion development will be discussed.
Collapse
Affiliation(s)
- Nicole A Dobbs
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107
| | | | | | | |
Collapse
|
216
|
Abstract
CD4+ T cells, upon activation and expansion, develop into different T helper cell subsets with different cytokine profiles and distinct effector functions. Until recently, T cells were divided into Th1 or Th2 cells, depending on the cytokines they produce. A third subset of IL-17-producing effector T helper cells, called Th17 cells, has now been discovered and characterized. Here, we summarize the current information on the differentiation and effector functions of the Th17 lineage. Th17 cells produce IL-17, IL-17F, and IL-22, thereby inducing a massive tissue reaction owing to the broad distribution of the IL-17 and IL-22 receptors. Th17 cells also secrete IL-21 to communicate with the cells of the immune system. The differentiation factors (TGF-beta plus IL-6 or IL-21), the growth and stabilization factor (IL-23), and the transcription factors (STAT3, RORgammat, and RORalpha) involved in the development of Th17 cells have just been identified. The participation of TGF-beta in the differentiation of Th17 cells places the Th17 lineage in close relationship with CD4+CD25+Foxp3+ regulatory T cells (Tregs), as TGF-beta also induces differentiation of naive T cells into Foxp3+ Tregs in the peripheral immune compartment. The investigation of the differentiation, effector function, and regulation of Th17 cells has opened up a new framework for understanding T cell differentiation. Furthermore, we now appreciate the importance of Th17 cells in clearing pathogens during host defense reactions and in inducing tissue inflammation in autoimmune disease.
Collapse
Affiliation(s)
- Thomas Korn
- Technical University Munich, Department of Neurology, 81675 Munich, Germany.
| | | | | | | |
Collapse
|
217
|
Schulz SM, Köhler G, Schütze N, Knauer J, Straubinger RK, Chackerian AA, Witte E, Wolk K, Sabat R, Iwakura Y, Holscher C, Müller U, Kastelein RA, Alber G. Protective immunity to systemic infection with attenuated Salmonella enterica serovar enteritidis in the absence of IL-12 is associated with IL-23-dependent IL-22, but not IL-17. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:7891-901. [PMID: 19017979 DOI: 10.4049/jimmunol.181.11.7891] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
IL-12 is essential for protective T cell-mediated immunity against Salmonella infection. To characterize the role of the related cytokine IL-23, wild-type (WT) C57BL/6 and p19(-/-) mice were infected systemically with an attenuated strain of Salmonella enterica serovar Enteritidis (S. Enteritidis). IL-23-deficient mice controlled infection with S. Enteritidis similarly as WT mice. Similar IFN-gamma production as compared with WT mice, but defective IL-17A and IL-22 production was found in the absence of IL-23. Nevertheless, although IL-23 is required for T cell-dependent cytokine responses, IL-23 is dispensable for protection against S. Enteritidis when IL-12 is present. To analyze the role of IL-23 in the absence of IL-12, low doses of S. Enteritidis were administered to p35(-/-) mice (lacking IL-12), p35/19(-/-) mice (lacking IL-12 and IL-23), p35/40(-/-) mice (lacking IL-12, IL-23, and homodimeric IL-12p40), or p35/IL-17A(-/-) mice (lacking IL-12 and IL-17A). We found survival of p35(-/-) and p35/IL-17A(-/-) mice, whereas p35/19(-/-) and p35/40(-/-) mice died within 3-6 wk and developed liver necrosis. This indicates that IL-23, but not homodimeric IL-12p40, is required for protection, which, surprisingly, is independent of IL-17A. Moreover, protection was associated with IL-22, but not IL-17F or IL-21 expression or with neutrophil recruitment. Finally, anti-IL-22 treatment of S. Enteritidis-infected p35(-/-) mice resulted in liver necrosis, indicating a central role of IL-22 in hepatocyte protection during salmonellosis. In conclusion, IL-23-dependent IL-22, but not IL-17 production is associated with protection against systemic infection with S. Enteritidis in the absence of IL-12.
Collapse
Affiliation(s)
- Silke M Schulz
- Institute of Immunology, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
The T-helper 17 (Th17) lineage is a recently described subset of memory T cells that is characterized by its CD4(+) status and its ability to make a constellation of cytokines including interleukin-17A (IL-17A), IL-17F, IL-22, and, in humans, IL-26. Although most extensively described in the autoimmunity literature, there is growing evidence that the Th17 lineage plays a significant role in mediating host mucosal immunity to a number of pulmonary pathogens. This review highlights our current understanding of the role of the Th17 lineage and Th17 cytokines in mediating mucosal immunity to both pulmonary and gastrointestinal pathogens. While we have the strongest evidence that the Th17 lineage is centrally involved in mediating the host response to Gram-negative extracellular pulmonary pathogens, this literature is rapidly evolving and demonstrates a central role for Th17 cytokines both in primary infection and in recall responses seen in vaccine studies. In this review, we summarize the current state of this literature and present possible applications of Th17-targeted immunotherapy in the treatment and prevention of infection.
Collapse
Affiliation(s)
- Patricia J Dubin
- Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
219
|
Waites KB, Balish MF, Atkinson TP. New insights into the pathogenesis and detection of Mycoplasma pneumoniae infections. Future Microbiol 2008; 3:635-48. [PMID: 19072181 PMCID: PMC2633477 DOI: 10.2217/17460913.3.6.635] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mycoplasma pneumoniae is a common cause of upper and lower respiratory tract infections in persons of all ages and may be responsible for up to 40% of community-acquired pneumonias. A wide array of extrapulmonary events may accompany the infections caused by this organism, related to autoimmunity or direct spread. This review includes a discussion of the latest knowledge concerning the molecular pathological basis of mycoplasmal respiratory disease, how the organism interacts with the host immune system and its association with the development of chronic conditions such as asthma, recent emergence of macrolide resistance and the status of laboratory diagnostic methods.
Collapse
Affiliation(s)
- Ken B Waites
- University of Alabama at Birmingham, WP 230, 619 19th Street South, Birmingham, AL 35249, USA.
| | | | | |
Collapse
|
220
|
Abstract
Naïve CD4(+) helper T (TH) cells, upon activation by antigen-presenting cells (APC), differentiate into different types of effector cells that are characterized by their distinct cytokine production profiles and immune regulatory functions. In addition to TH1 and TH2 cells, a third subset of effector TH cells has recently been described and termed TH17. Since their identification, TH17 cells have emerged as crucial players in infectious, inflammatory, and autoimmune diseases, and cancer. In this review, we summarize the latest discoveries on the cytokine-mediated regulation and transcriptional programming of TH17 cells and their roles in different immune responses and diseases.
Collapse
Affiliation(s)
- Gustavo J Martinez
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
221
|
Baqir M, Chen CZ, Martin RJ, Thaikoottathil J, Case SR, Minor MN, Bowler R, Chu HW. Cigarette smoke decreases MARCO expression in macrophages: implication in Mycoplasma pneumoniae infection. Respir Med 2008; 102:1604-10. [PMID: 18590957 DOI: 10.1016/j.rmed.2008.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Revised: 04/10/2008] [Accepted: 05/16/2008] [Indexed: 10/21/2022]
Abstract
Bacterial infections including Mycoplasma pneumoniae (Mp) are a major cause of exacerbations in chronic obstructive pulmonary disease (COPD). Cigarette smoke (CS) is the leading cause of COPD, and affects the function of alveolar macrophages that act as the first line of defense against the invading respiratory pathogens. Macrophages express a transmembrane receptor called macrophage receptor with collagenous structure (MARCO) that is involved in the clearance of microorganisms. Whether CS down-regulates MARCO and eventually decreases the clearance of Mp has not been investigated. We utilized human monocytic cell line (THP-1)-derived macrophages to examine the effects of CS extract (CSE) on MARCO expression and Mp growth. Specifically, macrophages were pre-exposed to CSE for 6 h, and then infected with or without Mp for 2 h. MARCO was examined at both mRNA and protein levels by using real-time PCR and immunofluorescent staining, respectively. Mp in the supernatants was quantified by quantitative culture. In addition, a neutralizing MARCO antibody was added to macrophages to test if blockade of MARCO impaired Mp clearance. We found that CSE significantly decreased MARCO expression in a dose-dependant manner at 6 h post-CSE. Mp levels in CSE-treated cells were higher than those in non-CSE-treated cells, indicating a decreased pathogen clearance. Additionally, neutralizing MARCO in macrophages markedly increased Mp levels. Our results indicate that cigarette smoke exposure down-regulates MARCO expression in macrophages, which may be in part responsible for impaired bacterial (e.g., Mp) clearance.
Collapse
Affiliation(s)
- Misbah Baqir
- Department of Medicine, National Jewish Medical and Research Center, University of Colorado Health Sciences Center, Denver, CO 80206, USA
| | | | | | | | | | | | | | | |
Collapse
|
222
|
Interleukin-23 orchestrates mucosal responses to Salmonella enterica serotype Typhimurium in the intestine. Infect Immun 2008; 77:387-98. [PMID: 18955477 DOI: 10.1128/iai.00933-08] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Salmonella enterica serotype Typhimurium causes an acute inflammatory reaction in the ceca of streptomycin-pretreated mice that involves T-cell-dependent induction of gamma interferon (IFN-gamma), interleukin-22 (IL-22), and IL-17 expression (genes Ifn-gamma, Il-22, and Il-17, respectively). We investigated here the role of IL-23 in initiating these inflammatory responses using the streptomycin-pretreated mouse model. Compared to wild-type mice, the expression of IL-17 was abrogated, IL-22 expression was markedly reduced, but IFN-gamma expression was normal in the ceca of IL-23p19-deficient mice during serotype Typhimurium infection. IL-23p19-deficient mice also exhibited a markedly reduced expression of regenerating islet-derived 3 gamma, keratinocyte-derived cytokine, and reduced neutrophil recruitment into the cecal mucosa during infection. Analysis of CD3(+) lymphocytes in the intestinal mucosa by flow cytometry revealed that alphabeta T cells were the predominant cell type expressing the IL-23 receptor in naive mice. However, a marked increase in the number of IL-23 receptor-expressing gammadelta T cells was observed in the lamina propria during serotype Typhimurium infection. Compared to wild-type mice, gammadelta T-cell-receptor-deficient mice exhibited blunted expression of IL-17 during serotype Typhimurium infection, while IFN-gamma expression was normal. These data suggested that gammadelta T cells are a significant source, but not the sole source, of IL-17 in the acutely inflamed cecal mucosa of mice. Collectively, our results point to IL-23 as an important player in initiating a T-cell-dependent amplification of inflammatory responses in the intestinal mucosa during serotype Typhimurium infection.
Collapse
|
223
|
Held KS, Glass WG, Orlovsky YI, Shamberger KA, Petley TD, Branigan PJ, Carton JM, Beck HS, Cunningham MR, Benson JM, Lane TE. Generation of a protective T-cell response following coronavirus infection of the central nervous system is not dependent on IL-12/23 signaling. Viral Immunol 2008; 21:173-88. [PMID: 18570589 DOI: 10.1089/vim.2008.0014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The functional role of IL-12 and IL-23 in host defense and disease following viral infection of the CNS was determined. Instillation of mouse hepatitis virus (MHV, a positive-strand RNA virus) into the CNS of mice results in acute encephalitis followed by a chronic immune-mediated demyelinating disease. Antibody-mediated blocking of either IL-23 (anti-IL-23p19) or IL-12 and IL-23 (anti-IL-12/23p40) signaling did not mute T-cell trafficking into the CNS or antiviral effector responses and mice were able to control viral replication within the brain. Therapeutic administration of either anti-IL-23p19 or anti-IL-12/23p40 to mice with viral-induced demyelination did not attenuate T-cell or macrophage infiltration into the CNS nor improve clinical disease or diminish white matter damage. In contrast, treatment of mice with anti-IL-12/23p40 or anti-IL-23p19 resulted in inhibition of the autoimmune model of demyelination, experimental autoimmune encephalomyelitis (EAE). These data indicate that (1) IL-12 and IL-23 signaling are dispensable in generating a protective T-cell response following CNS infection with MHV, and (2) IL-12 and IL-23 do not contribute to demyelination in a model independent of autoimmune T-cell-mediated pathology. Therefore, therapeutic targeting of IL-12 and/or IL-23 for the treatment of autoimmune diseases may offer unique advantages by reducing disease severity without muting protective responses following viral infection.
Collapse
Affiliation(s)
- Katherine S Held
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697-3900, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
The discovery of the Th1/Th2 paradigm of CD4(+) T-cell subsets redefined our understanding of immunity by highlighting the essential roles of cytokine networks in the induction and regulation of immune responses. Most recently, the identification of an additional subset, known as Th17 cells, has further illustrated the complexity and diversity of effector CD4(+) T cells. Th17 responses have been closely associated with the cytokine interleukin (IL)-23 and, although originally pinpointed as having a deleterious role in autoimmune tissue pathology, the IL-23/Th17 axis has also been associated with protective immunity at mucosal surfaces. Recent progress has highlighted the heterogeneous nature of Th17 responses, has demonstrated diverse cellular sources for Th17-associated cytokines, and has begun to dissect the individual roles of these cytokines in different disease processes. Here, we will review the evidence linking the IL-23/Th17 axis to chronic intestinal inflammation and also will discuss its beneficial roles in intestinal protection and homeostasis.
Collapse
Affiliation(s)
- K J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | | |
Collapse
|
225
|
Atkinson TP, Balish MF, Waites KB. Epidemiology, clinical manifestations, pathogenesis and laboratory detection of Mycoplasma pneumoniae infections. FEMS Microbiol Rev 2008; 32:956-73. [PMID: 18754792 DOI: 10.1111/j.1574-6976.2008.00129.x] [Citation(s) in RCA: 336] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Since its initial description in the 1940s and eventual elucidation as a highly evolved pathogenic bacterium, Mycoplasma pneumoniae has come to be recognized as a worldwide cause of primary atypical pneumonia. Beyond its ability to cause severe lower respiratory illness and milder upper respiratory symptoms it has become apparent that a wide array of extrapulmonary infectious and postinfectious events may accompany the infections in humans caused by this organism. Autoimmune disorders and chronic diseases such as asthma and arthritis are increasingly being associated with this mycoplasma, which frequently persists in individuals for prolonged periods. The reductive evolutionary process that has led to the minimal genome of M. pneumoniae suggests that it exists as a highly specialized parasitic bacterium capable of residing in an intracellular state within the respiratory tissues, occasionally emerging to produce symptoms. This review includes discussion of some of the newer aspects of our knowledge on this pathogen, characteristics of clinical infections, how it causes disease, the recent emergence of macrolide resistance, and the status of laboratory diagnostic methods.
Collapse
|
226
|
Abstract
Acute diarrheal illness is a global health problem that may be exacerbated by concurrent infection. Using Citrobacter rodentium, a murine model of attaching and effacing diarrheagenic Escherichia coli, we demonstrate that persistent Helicobacter hepaticus infection modulates host responses to diarrheal disease, resulting in delayed recovery from weight loss and from tissue damage. Chronic colitis in concurrently infected mice is characterized by macrophage and Foxp3(+) regulatory T-cell accumulation. Prolonged disease is also associated with increased interleukin-17 expression, which may be due to suppression of gamma interferon during the acute phase of diarrheal infection. This new model of polymicrobial infection provides insight into the mechanism by which subclinical infection can exacerbate morbidity due to an unrelated self-limiting infection.
Collapse
|
227
|
Cheung PFY, Wong CK, Lam CWK. Molecular mechanisms of cytokine and chemokine release from eosinophils activated by IL-17A, IL-17F, and IL-23: implication for Th17 lymphocytes-mediated allergic inflammation. THE JOURNAL OF IMMUNOLOGY 2008; 180:5625-35. [PMID: 18390747 DOI: 10.4049/jimmunol.180.8.5625] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
IL-17A and IL-17F are members of the IL-17 family that play crucial roles in allergic inflammation. Recent studies reported that IL-17A and IL-17F production from a distinct Th lymphocyte subset, Th17, was specifically induced by IL-23, which was produced by dendritic cells and macrophages in response to microbial stimuli. The IL-23-IL-17 axis might therefore provide a link between infections and allergic diseases. In the present study, we investigated the effects of IL-17A, IL-17F, and IL-23, alone or in combination, on cytokine and chemokine release from eosinophils and the underlying intracellular mechanisms. Human eosinophils were found to constitutively express receptors for IL-17A, IL-17F, and IL-23 at the protein level. IL-17A, IL-17F, and IL-23 could induce the release of chemokines GRO-alpha/CXCL1, IL-8/CXCL8, and MIP-1beta/CCL4 from eosinophils, while IL-17F and IL-23 could also increase the production of proinflammatory cytokines IL-1beta and IL-6. Synergistic effects were observed in the combined treatment of IL-17F and IL-23 on the release of proinflammatory cytokines, and the effects were dose-dependently enhanced by IL-23, but not IL-17F. Further investigations showed that IL-17A, IL-17F, and IL-23 differentially activated the ERK, p38 MAPK, and NF-kappaB pathways. Moreover, inhibition of these pathways using selective inhibitors could significantly abolish the chemokine release induced by IL-17A, IL-17F, and IL-23 and the synergistic increases on IL-1beta and IL-6 production mediated by combined treatment of IL-17F and IL-23. Taken together, our findings provide insight for the Th17 lymphocyte-mediated activation of eosinophils via differential intracellular signaling cascades in allergic inflammation.
Collapse
Affiliation(s)
- Phyllis F Y Cheung
- Department of Chemical Pathology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | | | | |
Collapse
|
228
|
Wissinger EL, Saldana J, Didierlaurent A, Hussell T. Manipulation of acute inflammatory lung disease. Mucosal Immunol 2008; 1:265-78. [PMID: 19079188 PMCID: PMC7100270 DOI: 10.1038/mi.2008.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 02/26/2008] [Indexed: 02/04/2023]
Abstract
Inflammatory lung disease to innocuous antigens or infectious pathogens is a common occurrence and in some cases, life threatening. Often, the inflammatory infiltrate that accompanies these events contributes to pathology by deleterious effects on otherwise healthy tissue and by compromising lung function by consolidating (blocking) the airspaces. A fine balance, therefore, exists between a lung immune response and immune-mediated damage, and in some the "threshold of ignorance" may be set too low. In most cases, the contributing, potentially offending, cell population or immune pathway is known, as are factors that regulate them. Why then are targeted therapeutic strategies to manipulate them not more commonplace in clinical medicine? This review highlights immune homeostasis in the lung, how and why this is lost during acute lung infection, and strategies showing promise as future immune therapeutics.
Collapse
Affiliation(s)
- E L Wissinger
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| | - J Saldana
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| | - A Didierlaurent
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
- Present Address: Present address: GlaxoSmithKline Biologicals, Rue de l'Institut 89, Rixensart B-1330, Belgium,
| | - T Hussell
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| |
Collapse
|
229
|
Abstract
Although the precise aetiology of inflammatory bowel disease (IBD) remains unclear, recent discoveries have led to an improved understanding of disease pathogenesis. Whilst these findings have underscored the central role of innate and adaptive immune responses in intestinal inflammation, they have also precipitated a paradigm shift in the key cytokine pathways that drive disease. The prevailing dogma that IBD was mediated by interleukin (IL)-12-driven T-helper (Th)1 CD4 T cell responses towards the bacterial flora has been largely dispelled by findings that the closely related cytokine IL-23 appears to be the key mediator of intestinal inflammation. IL-23 is associated with a novel subset of IL-17-secreting CD4 T cells termed Th17 cells and rodent studies have implicated the IL-23/IL-17 axis in autoimmune inflammation. Genome-wide association studies in IBD patients have confirmed the predominant role of the IL-23 pathway, indicating that this could represent an important future therapeutic target.
Collapse
Affiliation(s)
- Kevin J Maloy
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
230
|
Rutitzky LI, Bazzone L, Shainheit MG, Joyce-Shaikh B, Cua DJ, Stadecker MJ. IL-23 is required for the development of severe egg-induced immunopathology in schistosomiasis and for lesional expression of IL-17. THE JOURNAL OF IMMUNOLOGY 2008; 180:2486-95. [PMID: 18250458 DOI: 10.4049/jimmunol.180.4.2486] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In infection with the trematode helminth Schistosoma mansoni, the severity of CD4 T cell-mediated hepatic granulomatous and fibrosing inflammation against parasite eggs varies considerably in humans and among mouse strains. In mice, either the natural high pathology, or high pathology induced by concomitant immunization with schistosome egg Ags (SEA) in CFA (SEA/CFA), results from a failure to contain a net proinflammatory cytokine environment. We previously demonstrated that the induction of severe immunopathology was dependent on the IL-12/IL-23 common p40 subunit, and correlated with an increase in IL-17, thus implying IL-23 in the pathogenesis. We now show that mice lacking the IL-23-specific subunit p19 are impaired in developing severe immunopathology following immunization with SEA/CFA, which is associated with a marked drop of IL-17 in the granulomas, but not in the draining mesenteric lymph nodes, and with a markedly suppressed SEA-specific IFN-gamma response regulated by a striking increase in IL-10. The granulomas are characterized by a significant reduction in Gr-1(+) cell recruitment and by alternative macrophage activation. Taken together, these results demonstrate that IL-23 per se is not necessary for the generation of IL-17-producing T cells, but is essential for the development of severe schistosome egg-induced immunopathology, and its absence cannot be overcome with other possible compensatory mechanisms.
Collapse
Affiliation(s)
- Laura I Rutitzky
- Department of Pathology, School of Medicine, Tufts University, 150 Harrison Avenue, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
231
|
Respiratory Francisella tularensis live vaccine strain infection induces Th17 cells and prostaglandin E2, which inhibits generation of gamma interferon-positive T cells. Infect Immun 2008; 76:2651-9. [PMID: 18391003 DOI: 10.1128/iai.01412-07] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Two key routes of Francisella tularensis infection are through the skin and airway. We wished to understand how the route of inoculation influenced the primary acute adaptive immune response. We show that an intranasal inoculation of the F. tularensis live vaccine strain (LVS) with a 1,000-fold-smaller dose than an intradermal dose results in similar growth kinetics and peak bacterial burdens. In spite of similar bacterial burdens, we demonstrate a difference in the quality, magnitude, and kinetics of the primary acute T-cell response depending on the route of inoculation. Further, we show that prostaglandin E(2) secretion in the lung is responsible for the difference in the gamma interferon (IFN-gamma) response. Intradermal inoculation led to a large number of IFN-gamma(+) T cells 7 days after infection in both the spleen and the lung. In contrast, intranasal inoculation induced a lower number of IFN-gamma(+) T cells in the spleen and lung but an increased number of Th17 cells in the lung. Intranasal infection also led to a significant increase of prostaglandin E(2) (PGE(2)) in the bronchoalveolar lavage fluid. Inhibition of PGE(2) production with indomethacin treatment resulted in increased numbers of IFN-gamma(+) T cells and decreased bacteremia in the lungs of intranasally inoculated mice. This research illuminates critical differences in acute adaptive immune responses between inhalational and dermal infection with F. tularensis LVS mediated by the innate immune system and PGE(2).
Collapse
|
232
|
Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, Pociask DA, Reinhart TA, McAllister F, Edeal J, Gaus K, Husain S, Kreindler JL, Dubin PJ, Pilewski JM, Myerburg MM, Mason CA, Iwakura Y, Kolls JK. IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat Med 2008; 14:275-81. [PMID: 18264110 PMCID: PMC2901867 DOI: 10.1038/nm1710] [Citation(s) in RCA: 938] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 12/17/2007] [Indexed: 12/11/2022]
Abstract
Emerging evidence supports the concept that T helper type 17 (T(H)17) cells, in addition to mediating autoimmunity, have key roles in mucosal immunity against extracellular pathogens. Interleukin-22 (IL-22) and IL-17A are both effector cytokines produced by the T(H)17 lineage, and both were crucial for maintaining local control of the Gram-negative pulmonary pathogen, Klebsiella pneumoniae. Although both cytokines regulated CXC chemokines and granulocyte colony-stimulating factor production in the lung, only IL-22 increased lung epithelial cell proliferation and increased transepithelial resistance to injury. These data support the concept that the T(H)17 cell lineage and its effector molecules have evolved to effect host defense against extracellular pathogens at mucosal sites.
Collapse
Affiliation(s)
- Shean J Aujla
- Children's Hospital of Pittsburgh, Suite 3765, 3705 Fifth Avenue, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Yoo J, Chang HH, Bae YH, Seong CN, Choe NH, Lillehoj HS, Park JH, Min W. Monoclonal antibodies reactive with chicken interleukin-17. Vet Immunol Immunopathol 2008; 121:359-63. [DOI: 10.1016/j.vetimm.2007.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 07/28/2007] [Accepted: 10/05/2007] [Indexed: 11/30/2022]
|
234
|
Wu Q, Martin RJ, Lafasto S, Efaw BJ, Rino JG, Harbeck RJ, Chu HW. Toll-like receptor 2 down-regulation in established mouse allergic lungs contributes to decreased mycoplasma clearance. Am J Respir Crit Care Med 2008; 177:720-9. [PMID: 18202345 DOI: 10.1164/rccm.200709-1387oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Respiratory Mycoplasma pneumoniae (Mp) infection is involved in asthma pathobiology, but whether the established allergic airway inflammation compromises lung innate immunity and subsequently predisposes patients with asthma to Mp infection remains unknown. OBJECTIVES To test whether the established allergic airway inflammation compromises host innate immunity (e.g., Toll-like receptor 2 [TLR2]) to hinder the elimination of Mp from the lungs. METHODS We used mouse models of ovalbumin (OVA)-induced allergic airway inflammation with an ensuing Mp infection, and cultures of mouse primary lung dendritic cells (DCs) and bone marrow-derived DCs. MEASUREMENTS AND MAIN RESULTS Lung Mp clearance in allergic mice and TLR2 and IL-6 levels in lung cells, including DCs as well as cultured primary lung DCs and bone marrow-derived DCs, were assessed. The established OVA-induced allergic airway inflammation, or the prominent Th2 cytokines IL-4 and IL-13, inhibited TLR2 expression and IL-6 production in lung cells, including lung DCs, and eventually led to impaired host defense against Mp. Studies in IL-6 knockout mice indicated that IL-6 directly promoted Mp clearance from the lungs. IL-4- and IL-13-induced suppression of TLR2 was mediated by inhibiting nuclear factor-kappaB activation through signal transducer and activator of transcription 6 (STAT6) signaling pathway. CONCLUSIONS The established OVA-induced allergic airway inflammation impairs TLR2 expression and host defense cytokine (e.g., IL-6) production, and subsequently delays lung bacterial clearance. This could offer novel therapeutic strategies to reinstate TLR2 activation by using TLR2 ligands and/or blocking IL-4 and IL-13 to ameliorate persisting respiratory bacterial infections in allergic lungs.
Collapse
Affiliation(s)
- Qun Wu
- National Jewish Medical and Research Center, 1400 Jackson Street, Room A639, Denver, CO 80206, USA.
| | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
Recently, a paradigm shift has emerged in T-cell-mediated adaptive immunity. On the heels of the discovery of T cells with immunosuppressive function, so-called regulatory T cells (Tregs), the diversity of effector cells has expanded to include a third helper T cell, termed Th17. The appreciation that Th17 cells are products of a distinct effector pathway depended critically on observations made during investigations of mouse models of autoimmunity, advanced by discovery of the cytokines IL-17 and IL-23. These studies understandably led investigators to highlight the role played by Th17 cells in autoimmunity. Yet while the dysfunctional behavior of this phenotype as a contributor to inflammatory disease remains a central issue, this pathway evolved to meet a need for host protection against potential pathogens. It has become apparent that the Th17 pathway promotes host defense against certain extracellular bacteria and fungi, but more recent studies also implicate a role in protection against some protozoa and viruses. Here we review the experimental history that ultimately uncovered the existence and nature of Th17 cells, and then turn the reader's attention to what is currently known about Th17 cells as a bulwark against pathogens.
Collapse
|
236
|
Uyttenhove C, Sommereyns C, Théate I, Michiels T, Van Snick J. Anti-IL-17A autovaccination prevents clinical and histological manifestations of experimental autoimmune encephalomyelitis. Ann N Y Acad Sci 2007; 1110:330-6. [PMID: 17911448 DOI: 10.1196/annals.1423.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Excessive or inappropriate production of IL-17A has been reported in diseases such as rheumatoid arthritis, asthma, and multiple sclerosis. The potential clinical relevance of these correlations was suggested by the protective effects of anti-IL-17A monoclonal antibodies in various mouse disease models. However, the chronic nature of the corresponding human afflictions raises great challenges for Ab-based therapies. An alternative to passive Ab therapy is autovaccination. Covalent association of self-cytokines with foreign proteins has been reported to induce the production of antibodies capable of neutralizing the biological activity of the target cytokine. We recently reported that cross-linking of IL-17A to ovalbumin produced highly immunogenic complexes that induced long-lasting IL-17A-neutralizing antibodies. Vaccinated SJL mice were completely protected against experimental autoimmune encephalomyelitis (EAE) induced by proteolipid protein peptide (PLP 139-151), and a monoclonal anti-IL-17A Ab (MM17F3), derived from C57Bl/6 mice vaccinated against IL-17A-OVA, also prevented disease development. Here we report that this Ab also protects C57Bl/6 mice from myelin oligdendrocyte glycoprotein (MOG)-induced EAE. Histological analysis of brain sections of C57Bl/6 mice treated with MM17F3 showed a complete absence of inflammatory infiltrates and evidence for a marked inhibition of chemokine and cytokine messages in the spinal cord. These results further extend the analytical and therapeutic potential of the autovaccine procedure.
Collapse
Affiliation(s)
- Catherine Uyttenhove
- Ludwig Institute for Cancer Research, Brussels Branch, 74 av. Hippocrate UCL 7459, B-1200 Brussels, Belgium.
| | | | | | | | | |
Collapse
|
237
|
Abstract
Th17 cells are a new lineage of T-cells that are controlled by the transcription factor RORgammat and develop independent of GATA-3, T-bet, Stat 4 and Stat 6. Novel effector molecules produced by these cells include IL-17A, IL-17F, IL-22, and IL-26. IL-17RA binds IL-17A and IL-17F and is critical for host defense against extracellular planktonic bacteria by regulating chemokine gradients for neutrophil emigration into infected tissue sites as well as host granulopoiesis. Moreover, IL-17 and IL-22 regulate the production of antimicrobial proteins in mucosal epithelium. Although TGF-beta1 and IL-6 have been shown to be critical for development of Th17 cells from naive precursors, IL-23 is also important in regulating IL-17 release in mucosal tissues in response to infectious stimuli. Compared to Th1 cells, IL-23 and IL-17 show limited roles in controlling host defense against primary infections with intracellular bacteria such as Mycobacterium tuberculosis suggesting a predominate role of the Th17 lineage in host defense against extracellular pathogens. However, in the setting of chronic biofilm infections, as that occurs with cystic fibrosis or bronchiectasis, Th17 cells may be key contributors of tissue injury.
Collapse
|