1351
|
Blumberg H, Dinh H, Dean C, Trueblood ES, Bailey K, Shows D, Bhagavathula N, Aslam MN, Varani J, Towne JE, Sims JE. IL-1RL2 and its ligands contribute to the cytokine network in psoriasis. THE JOURNAL OF IMMUNOLOGY 2010; 185:4354-62. [PMID: 20833839 DOI: 10.4049/jimmunol.1000313] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Psoriasis is a common immune-mediated disease in European populations; it is characterized by inflammation and altered epidermal differentiation leading to redness and scaling. T cells are thought to be the main driver, but there is also evidence for an epidermal contribution. In this article, we show that treatment of mouse skin overexpressing the IL-1 family member, IL-1F6, with phorbol ester leads to an inflammatory condition with macroscopic and histological similarities to human psoriasis. Inflammatory cytokines thought to be important in psoriasis, such as TNF-α, IL-17A, and IL-23, are upregulated in the mouse skin. These cytokines are induced by and can induce IL-1F6 and related IL-1 family cytokines. Inhibition of TNF or IL-23 inhibits the increased epidermal thickness, inflammation, and cytokine production. Blockade of IL-1F6 receptor also resolves the inflammatory changes in human psoriatic lesional skin transplanted onto immunodeficient mice. These data suggest a role for IL-1F family members in psoriasis.
Collapse
|
1352
|
Khader SA, Gopal R. IL-17 in protective immunity to intracellular pathogens. Virulence 2010; 1:423-7. [PMID: 21178483 PMCID: PMC2953849 DOI: 10.4161/viru.1.5.12862] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 06/28/2010] [Accepted: 06/30/2010] [Indexed: 01/04/2023] Open
Abstract
The identification of a new T cell subset referred to as T helper 17 (Th17) cells and its role in protective immunity against extracellular bacterial infections is well established. In contrast, initial studies suggested that the IL-23-IL-17 pathway was not required for protection against intracellular pathogens such as mycobacterial infections. However, recent studies demonstrate that Th17-IL-23 pathway may play a crucial role in protective immunity against other intracellular pathogens by regulating the innate and adaptive immune responses. The current outlook on the role of IL-23-IL-17 pathway in protective immunity to intracellular pathogens is discussed here.
Collapse
Affiliation(s)
- Shabaana A Khader
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | |
Collapse
|
1353
|
Eyerich S, Eyerich K, Cavani A, Schmidt-Weber C. IL-17 and IL-22: siblings, not twins. Trends Immunol 2010; 31:354-61. [PMID: 20691634 DOI: 10.1016/j.it.2010.06.004] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 06/10/2010] [Accepted: 06/21/2010] [Indexed: 01/25/2023]
Abstract
T helper (Th) cell subsets secrete cytokines that regulate other immune cells. Interleukin (IL)-17 and IL-22 belong to a new class of cytokines with predominant effects on epithelial cells. Thus, these cytokines are key molecules in several disease processes. IL-17 and IL-22 are released by leukocytes such as Th and natural killer cell populations. Both IL-17 and IL-22 induce an innate immune response in epithelial cells, but their functional spectra are generally distinct. IL-17 induces an inflammatory tissue response and is involved in the pathogenesis of several autoimmune diseases, whereas IL-22 is protective/regenerative. This review juxtaposes IL-17 and IL-22 and describes overlaps and differences regarding their cellular sources, biochemical structure, signaling cascades in target cells, and function.
Collapse
Affiliation(s)
- Stefanie Eyerich
- Molecular Immunology, Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College, London, UK.
| | | | | | | |
Collapse
|
1354
|
Kopf M, Bachmann MF, Marsland BJ. Averting inflammation by targeting the cytokine environment. Nat Rev Drug Discov 2010; 9:703-18. [DOI: 10.1038/nrd2805] [Citation(s) in RCA: 185] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
1355
|
Ramirez JM, Brembilla NC, Sorg O, Chicheportiche R, Matthes T, Dayer JM, Saurat JH, Roosnek E, Chizzolini C. Activation of the aryl hydrocarbon receptor reveals distinct requirements for IL-22 and IL-17 production by human T helper cells. Eur J Immunol 2010; 40:2450-9. [PMID: 20706985 DOI: 10.1002/eji.201040461] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ligands of the aryl hydrocarbon receptor (AHR), a transcription factor mediating the effects of dioxin, favor Th17 differentiation and exacerbate autoimmunity in mice. We investigated how AHR ligands affected human T-cell polarization. We found that the high affinity and stable AHR-ligand dioxin as well as the natural AHR-ligand 6-formylinolo[3,2-b] carbazole induced the downstream AHR-target cytochrome P450A1, and without affecting IFN-gamma, they enhanced IL-22 while simultaneously decreasing IL-17A production by CD4(+) T cells. The specific AHR-inhibitor CH-223191 abolished these effects. Furthermore, blockade of IL-23 and IL-1, important for Th17 expansion, profoundly decreased IL-17A but not IL-22 production. AHR agonists reduced the expression of the Th17 master transcription factor retinoic acid-related orphan receptor C (RORC), without affecting T-bet, GATA-3 and Foxp3. They also decreased the expression of the IL-23 receptor. Importantly, AHR-ligation did not only decrease the number of Th17 cells but also primed naïve CD4(+) T cells to produce IL-22 without IL-17 and IFN-gamma. Furthermore, IL-22 single producers did not express CD161, which distinguished them from the CD161(+) Th17 cells. Hence, our data provide compelling evidence that AHR activation participates in shaping human CD4(+) T-cell polarization favoring the emergence of a distinct subset of IL-22-producing cells that are independent from the Th17 lineage.
Collapse
Affiliation(s)
- Jean-Marie Ramirez
- Immunology and Allergy, Swiss Centre for Applied Human Toxicology, University Hospital and School of Medicine, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
1356
|
Chmill S, Kadow S, Winter M, Weighardt H, Esser C. 2,3,7,8-Tetrachlorodibenzo-p-dioxin impairs stable establishment of oral tolerance in mice. Toxicol Sci 2010; 118:98-107. [PMID: 20729464 DOI: 10.1093/toxsci/kfq232] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The toxic environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent immunomodulatory chemical. TCDD activates the aryl hydrocarbon receptor (AhR) and suppresses peripheral humoral and cellular adaptive immune responses. Though the major route of uptake is via food, little is known until now on the immunotoxic effects of TCDD on the gut-associated lymphoid tissue. We show here that AhR is strongly expressed along the small intestine, especially in intestinal epithelial cells (IEC). The AhR marker gene cyp1a1 is induced in IEC by oral TCDD exposure. We asked how TCDD affects oral tolerance, a unique function of mucosal immunity. C57BL/6 mice were injected with 10 μg/kg body weight TCDD and fed with ovalbumin (OVA) in a high-dose tolerization protocol. Mice were immunized and boosted with OVA on days 12, 23, and 55 after tolerization. Five of 14, 6 of 15, and 13 of 14 TCDD-treated mice generated OVA-specific immunoglobulin (Ig)G1 antibodies after the first, second, and third immunization with OVA, respectively. Only one mouse harbored anti-OVA IgG1 antibodies in the control group even after the third immunization with OVA. OVA-specific IgA in fecal samples of tolerized and TCDD-exposed mice could be detected at the levels of nontolerized mice, whereas completely absent in tolerant control mice. Correlated to this, we found in TCDD-treated mice an increase in interleukin-6 producing CD103+ dendritic cells (DC) present in the gut-draining mesenteric lymph nodes (MLN) and a small increase in the frequency of Th17 cells. Neither the frequencies nor the absolute numbers of immune cells in the lamina propria (LP) or in intraepithelial lymphocytes were changed by TCDD treatment. Our data not only have implications for food allergies in settings of environmental exposure but also raise concerns regarding the harmlessness of overdosing potential AhR agonist in food, which needs to be studied further.
Collapse
Affiliation(s)
- Stefanie Chmill
- Institute for Environmental Medical Research, Molecular Immunology Unit, 40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
1357
|
Onishi RM, Park SJ, Hanel W, Ho AW, Maitra A, Gaffen SL. SEF/IL-17R (SEFIR) is not enough: an extended SEFIR domain is required for il-17RA-mediated signal transduction. J Biol Chem 2010; 285:32751-32759. [PMID: 20729198 DOI: 10.1074/jbc.m110.121418] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IL-17, the hallmark cytokine of the Th17 population, mediates immunity to extracellular pathogens and promotes autoimmune immunopathology. The signaling mechanisms triggered by the IL-17 receptor (IL-17RA) and related receptors are strikingly different from other cytokine subclasses. Namely, IL-17Rs contain a conserved SEF/IL-17R (SEFIR) subdomain that engages Act1, leading to activation of TRAF6, NF-κB, and other events. Although the SEFIR is critical for signaling, the molecular details of the functional subdomains within IL-17RA remain poorly characterized. Here, we provide a detailed structure-function analysis delineating the C-terminal boundary of the SEFIR-containing region of IL-17RA. We show that functionality of this domain requires a large extension to the previously identified SEFIR motif. In contrast to the SEFIR, this extension is not conserved among IL-17R family members. Surprisingly, Act1 recruitment is not sufficient for downstream signaling activation, whereas ubiquitination of TRAF6 correlates tightly with functional receptors. We further demonstrate that IL-17RA exhibits signaling properties that are nonredundant with other IL-17R family members. Finally, we report that IL-17 signals synergistically with lymphotoxin-α3, using the same signaling motifs within IL-17RA. These studies provide new insight into the structure-function relationships of IL-17RA and reveal distinct signaling differences among IL-17R family members.
Collapse
Affiliation(s)
- Reiko M Onishi
- From the University of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania 15261
| | - Sangmi J Park
- From the University of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania 15261
| | - Walter Hanel
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Allen W Ho
- From the University of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania 15261; Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Amarnath Maitra
- Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York 14214
| | - Sarah L Gaffen
- From the University of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Pittsburgh, Pennsylvania 15261; Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, New York 14214.
| |
Collapse
|
1358
|
Abstract
Secondary lymphoid organs are important locations for the initiation of adaptive immune responses. They develop before birth, and their formation requires interaction between lymphotoxin-α₁ß₂-expressing lymphoid-tissue inducer cells and lymphotoxin-ß receptor-expressing stromal organizer cells. Here, we discuss new insights into the earliest phases of peripheral lymph node and Peyer's patch formation that occur before lymphotoxin-ß receptor signalling and suggest a role for the developing nervous system. In addition, we discuss the differing requirements for the postnatal formation of mucosa-associated lymphoid tissues and tertiary lymphoid structures that develop at sites of chronic inflammation.
Collapse
|
1359
|
Puel A, Picard C, Cypowyj S, Lilic D, Abel L, Casanova JL. Inborn errors of mucocutaneous immunity to Candida albicans in humans: a role for IL-17 cytokines? Curr Opin Immunol 2010; 22:467-74. [PMID: 20674321 PMCID: PMC3770911 DOI: 10.1016/j.coi.2010.06.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
Abstract
The various clinical manifestations of chronic mucocutaneous candidiasis (CMC) often result from acquired T-cell immunodeficiencies. More rarely, CMC results from inborn errors of immunity, the recent dissection of which has shed light on the molecular mechanisms of mucocutaneous immunity to Candida albicans. CMC may accompany various other infectious diseases in patients with almost any broad and profound T-cell primary immunodeficiency. By contrast, CMC is one of the few key infections in patients with autosomal dominant hyper IgE syndrome (mutations in STAT3), and in rare patients with autosomal recessive predisposition to mucocutaneous and invasive fungal infections (mutation in CARD9). In patients with mutations in STAT3 and CARD9, the development of IL-17-producing T cells is impaired. Moreover, CMC is the principal, if not only, infection in patients with autosomal recessive autoimmune polyendocrinopathy syndrome-I (mutations in AIRE). Patients with this condition have high titers of neutralizing autoantibodies (auto-Abs) against the IL-17 cytokines IL-17A, IL-17F, and IL-22. Collectively, these data suggest that human IL-17A, IL-17F, and IL-22 are essential for mucocutaneous immunity to C. albicans. They also suggest that the distinct syndrome of isolated CMC, without auto-immunity or other infections, may be caused by inborn errors of IL-17 immunity.
Collapse
Affiliation(s)
- Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Inserm U980, University Paris Descartes, Necker Medical School, Paris, France, EU.
| | | | | | | | | | | |
Collapse
|
1360
|
Sieve AN, Meeks KD, Lee S, Berg RE. A novel immunoregulatory function for IL-23: Inhibition of IL-12-dependent IFN-γ production. Eur J Immunol 2010; 40:2236-47. [PMID: 20458705 PMCID: PMC3039303 DOI: 10.1002/eji.200939759] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Most studies investigating the function of IL-23 have concluded that it promotes IL-17-secreting T cells. Although some reports have also characterized IL-23 as having redundant pro-inflammatory effects with IL-12, we have instead found that IL-23 antagonizes IL-12-induced secretion of IFN-γ. When splenocytes or purified populations of T cells were cultured with IL-23, IFN-γ secretion in response to IL-12 was dramatically reduced. The impact of IL-23 was most prominent in CD8(+) T cells, but was also observed in NK and CD4(+) T cells. Mechanistically, the IL-23 receptor was not required for this phenomenon, and IL-23 inhibited signaling through the IL-12 receptor by reducing IL-12-induced signal transducer and activator of transcription 4 (STAT4) phosphorylation. IL-23 was also able to reduce IFN-γ secretion by antagonizing endogenously produced IL-12 from Listeria monocytogenes (LM)-infected macrophages. In vivo, LM infection induced higher serum IFN-γ levels and a greater percentage of IFN-γ(+)CD8(+) T cells in IL-23p19-deficient mice as compared with WT mice. This increase in IFN-γ production coincided with increased LM clearance at days 2 and 3 post-infection. Our data suggest that IL-23 may be a key factor in determining the responsiveness of lymphocytes to IL-12 and their subsequent secretion of IFN-γ.
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
| | - Suheung Lee
- 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
|
1361
|
Yang L, Zhang Y, Wang L, Fan F, Zhu L, Li Z, Ruan X, Huang H, Wang Z, Huang Z, Huang Y, Yan X, Chen Y. Amelioration of high fat diet induced liver lipogenesis and hepatic steatosis by interleukin-22. J Hepatol 2010; 53:339-47. [PMID: 20452699 DOI: 10.1016/j.jhep.2010.03.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 02/26/2010] [Accepted: 03/01/2010] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Interleukin-22 (IL-22) is a Th17-related cytokine within the IL-10 family and plays an important role in host defense and inflammatory responses in orchestration with other Th17 cytokines. IL-22 exerts its functions in non-immune cells as its functional receptor IL-22R1 is restricted in peripheral tissues but not in immune cells. It was recently found that IL-22 serves as a protective molecule to counteract the destructive nature of the T cell-mediated immune response to liver damage. However, it is currently unknown whether IL-22 has an effect on lipid metabolism in the liver. METHODS In this study, we demonstrate that IL-22 alleviates hepatic steatosis induced by high fat diet (HFD). RESULTS Administration of recombinant murine IL-22 (rmIL-22) was able to stimulate STAT3 phosphorylation in HepG2 cells and mouse liver. The activation of STAT3 by rmIL-22 was reduced by the over-expression of a dominant negative IL-22R1. Within hours after rmIL-22 treatment, the expression of lipogenesis-related genes including critical transcription factors and enzymes for lipid synthesis in the liver was significantly down-regulated. The levels of triglyceride and cholesterol in the liver were significantly reduced by long-term treatment of rmIL-22 in C57BL/6 and ob/ob mice fed with HFD. The HFD-induced increases of ALT and AST in ob/ob mice were ameliorated by rmIL-22 treatment. In addition, the expression of fatty acid synthase and TNF-alpha in the liver was decreased by long-term rmIL-22 administration. CONCLUSIONS Collectively, these data indicate that IL-22, in addition to its known functions in host defense and inflammation, has a protective role in HFD-induced hepatic steatosis via its regulation on lipid metabolism in the liver.
Collapse
Affiliation(s)
- Ling Yang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1362
|
Caipang CMA, Lazado CC, Brinchmann MF, Kiron V. Infection-induced changes in expression of antibacterial and cytokine genes in the gill epithelial cells of Atlantic cod, Gadus morhua during incubation with bacterial pathogens. Comp Biochem Physiol B Biochem Mol Biol 2010; 156:319-25. [DOI: 10.1016/j.cbpb.2010.04.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2010] [Revised: 04/18/2010] [Accepted: 04/19/2010] [Indexed: 12/28/2022]
|
1363
|
Guo H, Topham DJ. Interleukin-22 (IL-22) production by pulmonary Natural Killer cells and the potential role of IL-22 during primary influenza virus infection. J Virol 2010; 84:7750-9. [PMID: 20504940 PMCID: PMC2897635 DOI: 10.1128/jvi.00187-10] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 05/14/2010] [Indexed: 11/20/2022] Open
Abstract
We set out to test the hypothesis that interleukin-22 (IL-22), a cytokine crucial for epithelial cell homeostasis and recovery from tissue injury, would be protective during influenza virus infection. Recent studies have identified phenotypically and functionally unique intestinal NK cells capable of producing the cytokine IL-22. Unlike gut NK cells that produce IL-22, the surface phenotypes of lung NK cells were similar to those of spleen NK cells and were characteristically mature. With mitogen stimulation, both single and double IL-22- and gamma interferon (IFN-gamma)-producing lung NK cells were detected. However, only the IL-22(+) IFN-gamma(-) lung NK subset was observed after stimulation with IL-23. IL-23 receptor (IL-23R) blocking dramatically inhibited IL-22 production, but not IFN-gamma production. Furthermore, we found that NK1.1(+) or CD27(-) lung NK cells were the primary sources of IL-22. After influenza virus infection, lung NK cells were quickly activated to produce both IFN-gamma and IL-22 and had increased cytotoxic potential. The level of IL-22 in the lung tissue declined shortly after infection, gradually returning to the baseline after virus clearance, although the IL-22 gene expression was maintained. Furthermore, depletion of NK cells with or without influenza virus infection reduced the protein level of IL-22 in the lung. Anti-IL-22 neutralization in vivo did not dramatically affect weight loss and survival after virus clearance. Unexpectedly, anti-IL-22-treated mice had reduced virus titers. Our data suggest that during primary respiratory viral infection, IL-22 seems to a play a marginal role for protection, indicating a differential requirement of this cytokine for bacterial and viral infections.
Collapse
Affiliation(s)
- Hailong Guo
- Department of Microbiology and Immunology and the David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, New York 14642
| | - David J. Topham
- Department of Microbiology and Immunology and the David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, New York 14642
| |
Collapse
|
1364
|
Thompson CL, Plummer SJ, Tucker TC, Casey G, Li L. Interleukin-22 genetic polymorphisms and risk of colon cancer. Cancer Causes Control 2010; 21:1165-70. [PMID: 20339910 DOI: 10.1007/s10552-010-9542-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Accepted: 03/11/2010] [Indexed: 10/19/2022]
Abstract
Interleukin-22 (IL-22) is a member of the IL-10 family of anti-inflammatory cytokines that mediates epithelial immunity. IL-22 expression is enhanced in inflamed colon mucosa in individuals with inflammatory bowel disease. We carried out an association study to examine the hypothesis that common variation in the IL-22 gene is associated with risk of colon cancer. Seven tagging SNPs were genotyped in 561 colon cancer cases and 722 population controls. Information on lifestyle risk factors was collected via a self-administered questionnaire. The rs1179251 SNP conferred an estimated odds ratio (OR) of 1.46 (95% CI = 1.04-2.05) and 2.10 (95% CI = 0.66-6.66), respectively, for those heterozygous and homozygous for the G variant (p (additive) = 0.013) after adjustment for age, gender, and race; the OR assuming a dominant model was 1.50 (95% CI = 1.05-2.08, p (dominant) = 0.016). No other SNP was statistically significantly associated with colon cancer risk. Haplotype analysis found that one haplotype containing the rs1179251 G allele gave an estimated 52% increase in risk of colon cancer for individuals with at least one copy (OR = 1.52, 95% CI = 1.12-2.06, p = 0.0073). Our findings suggest that the rs1179251 SNP in IL-22 is associated with risk of colon cancer.
Collapse
Affiliation(s)
- Cheryl L Thompson
- Department of Family Medicine-Research Division, Case Western Reserve University/University Hospitals of Cleveland, 11001 Cedar Ave., Suite 306, Cleveland, OH 44106-7136, USA.
| | | | | | | | | |
Collapse
|
1365
|
The other T helper cells in asthma pathogenesis. J Allergy (Cairo) 2010; 2010:519298. [PMID: 20976014 PMCID: PMC2957587 DOI: 10.1155/2010/519298] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/18/2010] [Indexed: 12/16/2022] Open
Abstract
The complex phenotype of allergic bronchial asthma involves a variable degree of bronchoobstruction, increased mucus production, and airway remodeling. So far it is suggested that it arises from multiple interactions of infiltrating and structural cells in the context of chronic airway inflammation that is orchestrated by T helper 2 (TH2) cells. By secreting a plethora of typical mediators such as interleukin (IL) 4, IL-5, and IL-13, these cells hold a key position in asthma pathogenesis. However, therapeutic approaches targeting these TH2-type mediators failed to improve asthma symptoms and impressively showed that asthma pathogenesis cannot be reduced by TH2 cell functions. Recently, other T helper cells, that is, TH9 and TH17 cells, have been identified and these cells also contribute to asthma pathogenesis, the processes leading to formation or aggravation of asthma. Furthermore, TH25 cells, TH3 cells, and regulatory T cells have also been implicated in asthma pathogenesis. This paper aims at summarizing recent insights about these new T helper cells in asthma pathogenesis.
Collapse
|
1366
|
Winter SE, Keestra AM, Tsolis RM, Bäumler AJ. The blessings and curses of intestinal inflammation. Cell Host Microbe 2010; 8:36-43. [PMID: 20638640 PMCID: PMC2918243 DOI: 10.1016/j.chom.2010.06.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 04/10/2010] [Accepted: 06/07/2010] [Indexed: 12/19/2022]
Abstract
The intestinal immune system has to strike a delicate balance between initiating inflammatory responses against invading bacterial pathogens and avoiding their induction against microbiota colonizing the lumen. Adequate inflammatory responses against bacterial invasion result in the lumenal secretion of antimicrobial peptides, as well as the release of cytokines in tissue that recruit and activate phagocytes. However, pathogens have evolved to utilize these environmental changes in the inflamed intestine to promote colonization. This review focuses on the costs and benefits of intestinal inflammation and the fine interplay between the host, its microbiota, and enteric pathogens.
Collapse
Affiliation(s)
- Sebastian E. Winter
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - A. Marijke Keestra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - Renée M. Tsolis
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| | - Andreas J. Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, One Shields Ave., Davis, CA, USA
| |
Collapse
|
1367
|
Van Maele L, Carnoy C, Cayet D, Songhet P, Dumoutier L, Ferrero I, Janot L, Erard F, Bertout J, Leger H, Sebbane F, Benecke A, Renauld JC, Hardt WD, Ryffel B, Sirard JC. TLR5 signaling stimulates the innate production of IL-17 and IL-22 by CD3(neg)CD127+ immune cells in spleen and mucosa. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:1177-85. [PMID: 20566828 PMCID: PMC3060348 DOI: 10.4049/jimmunol.1000115] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In adaptive immunity, Th17 lymphocytes produce the IL-17 and IL-22 cytokines that stimulate mucosal antimicrobial defenses and tissue repair. In this study, we observed that the TLR5 agonist flagellin induced swift and transient transcription of genes encoding IL-17 and IL-22 in lymphoid, gut, and lung tissues. This innate response also temporarily enhanced the expression of genes associated with the antimicrobial Th17 signature. The source of the Th17-related cytokines was identified as novel populations of CD3(neg)CD127(+) immune cells among which CD4-expressing cells resembling lymphoid tissue inducer cells. We also demonstrated that dendritic cells are essential for expression of Th17-related cytokines and so for stimulation of innate cells. These data define that TLR-induced activation of CD3(neg)CD127(+) cells and production of Th17-related cytokines may be crucial for the early defenses against pathogen invasion of host tissues.
Collapse
Affiliation(s)
- Laurye Van Maele
- CIL, Centre d'infection et d'immunité de Lille
INSERM : U1019Institut Pasteur de LilleLille,FR
- UMR 8204
CNRS : UMR 8204Université Lille NordLille,FR
| | - Christophe Carnoy
- CIL, Centre d'infection et d'immunité de Lille
INSERM : U1019Institut Pasteur de LilleLille,FR
- UMR 8204
CNRS : UMR 8204Université Lille NordLille,FR
| | - Delphine Cayet
- CIL, Centre d'infection et d'immunité de Lille
INSERM : U1019Institut Pasteur de LilleLille,FR
- UMR 8204
CNRS : UMR 8204Université Lille NordLille,FR
| | - Pascal Songhet
- ETH Zürich, Eldgenössische Technische Hochschule Zürich
ETH ZurichHauptgebäude Rämistrasse 101 8092 Zürich Schweiz Telefon: +41 44 632 11 11 Telefax: +41 44 632 10 10,CH
| | - Laure Dumoutier
- Ludwig Institute for Cancer Research
Ludwig Institute for Cancer ResearchBrussels Branch, Brussels,BE
- Université Catholique de Louvain
Université Catholique de LouvainBE
| | - Isabel Ferrero
- Ludwig Institute for Cancer Research
Ludwig Institute for Cancer ResearchUniversité de LausanneEpalinges,CH
| | - Laure Janot
- IEM, Immunologie et embryologie moléculaires
CNRS : UMR6218Université d'Orléans3B rue de la Ferollerie 45071 ORLEANS CEDEX 2,FR
| | - François Erard
- IEM, Immunologie et embryologie moléculaires
CNRS : UMR6218Université d'Orléans3B rue de la Ferollerie 45071 ORLEANS CEDEX 2,FR
| | - Julie Bertout
- MCM, Médecine cellulaire et moléculaire
CNRS : IFR142INSERMInstitut Pasteur de LilleUniversité des Sciences et Technologies de Lille - Lille IUniversité du Droit et de la Santé - Lille II1 rue du Professeur Calmette 59019 LILLE CEDEX,FR
| | - Hélène Leger
- IHES, Institut des Hautes Etudes Scientifiques
IHES35 route de Chartres 91440 Bures sur Yvette,FR
- USTL, Institut de Recherche Interdisciplinaire
CNRS : USR3078Université Lille NordLille,FR
| | - Florent Sebbane
- CIL, Centre d'infection et d'immunité de Lille
INSERM : U1019Institut Pasteur de LilleLille,FR
- UMR 8204
CNRS : UMR 8204Université Lille NordLille,FR
| | - Arndt Benecke
- IHES, Institut des Hautes Etudes Scientifiques
IHES35 route de Chartres 91440 Bures sur Yvette,FR
| | | | - Wolf-Dietrich Hardt
- ETH Zürich, Eldgenössische Technische Hochschule Zürich
ETH ZurichHauptgebäude Rämistrasse 101 8092 Zürich Schweiz Telefon: +41 44 632 11 11 Telefax: +41 44 632 10 10,CH
| | - Bernhard Ryffel
- IEM, Immunologie et embryologie moléculaires
CNRS : UMR6218Université d'Orléans3B rue de la Ferollerie 45071 ORLEANS CEDEX 2,FR
- USTL, Institut de Recherche Interdisciplinaire
CNRS : USR3078Université Lille NordLille,FR
| | - Jean-Claude Sirard
- CIL, Centre d'infection et d'immunité de Lille
INSERM : U1019Institut Pasteur de LilleLille,FR
- UMR 8204
CNRS : UMR 8204Université Lille NordLille,FR
| |
Collapse
|
1368
|
Ho AW, Shen F, Conti HR, Patel N, Childs EE, Peterson A, Hernández-Santos N, Kolls JK, Kane LP, Ouyang W, Gaffen SL. IL-17RC is required for immune signaling via an extended SEF/IL-17R signaling domain in the cytoplasmic tail. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:1063-70. [PMID: 20554964 PMCID: PMC2897912 DOI: 10.4049/jimmunol.0903739] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
IL-17 mediates essential inflammatory responses in host defense and autoimmunity. The IL-17A-IL-17F signaling complex is composed of IL-17RA and IL-17RC, both of which are necessary for signal transduction. To date, the specific contribution of IL-17RC to downstream signaling remains poorly understood. To define the regions within the IL-17RC cytoplasmic tail required for signal transduction, we assayed signaling by a panel of IL-17RC deletion mutants. These findings reveal that IL-17RC inducibly associates with a specific glycosylated IL-17RA isoform, in a manner independent of the IL-17RC cytoplasmic tail. Using expression of the IL-17 target genes IL-6 and 24p3/lipocalin-2 as a readout, functional reconstitution of signaling in IL-17RC(-/-) fibroblasts required the SEF/IL-17R signaling domain (SEFIR), a conserved motif common to IL-17R family members. Unexpectedly, the IL-17RC SEFIR alone was not sufficient to reconstitute IL-17-dependent signaling. Rather, an additional sequence downstream of the SEFIR was also necessary. We further found that IL-17RC interacts directly with the adaptor/E3 ubiquitin ligase Act1, and that the functional IL-17RC isoforms containing the extended SEFIR region interact specifically with a phosphorylated isoform of Act1. Finally, we show that IL-17RC is required for in vivo IL-17-dependent responses during oral mucosal infections caused by the human commensal fungus Candida albicans. These results indicate that IL-17RC is vital for IL-17-dependent signaling both in vitro and in vivo. Insight into the mechanisms by which IL-17RC signals helps shed light on IL-17-dependent inflammatory responses and may ultimately provide an avenue for therapeutic intervention in IL-17-mediated diseases.
Collapse
Affiliation(s)
- Allen W. Ho
- University at Buffalo, SUNY, Dept. of Oral Biology, Buffalo NY
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
| | - Fang Shen
- University at Buffalo, SUNY, Dept. of Oral Biology, Buffalo NY
- Genentech Inc., South San Francisco, CA
| | | | - Nayan Patel
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
| | - Erin E. Childs
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
| | - Alanna Peterson
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
| | - Nydiaris Hernández-Santos
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
| | - Jay K. Kolls
- LSU Health Sciences Center, Dept. of Genetics, New Orleans, LA
| | | | | | - Sarah L. Gaffen
- University at Buffalo, SUNY, Dept. of Oral Biology, Buffalo NY
- University of Pittsburgh, Dept. of Medicine, Division of Rheumatology & Clinical Immunology, Pittsburgh PA
- University of Pittsburgh, Dept. of Immunology, Pittsburgh PA
| |
Collapse
|
1369
|
Abstract
PURPOSE OF REVIEW The present review summarizes key studies on the effects of major abdominal surgery on the host response to infection published during the last 18 months. RECENT FINDINGS Surgical trauma causes stereotyped systemic proinflammatory and compensatory anti-inflammatory reactions. It is leukocyte reprogramming rather than general immune suppression. The list of recent findings is long. Preoperative infectious challenge was found to increase survival. Obesity is associated with increased production of interleukin-17A in peritonitis. Abdominal surgery alters expression of toll-like receptors (TLRs). The acute phase reaction down-regulates the transcription factor carbohydrate response element binding protein. Myosin light chain kinase activation is a final pathway of acute tight junction regulation of gut barrier and zonula occludens 1 protein is an essential effector. The brain is involved in regulating the immune and gut system. Elimination of lipopolysaccharide is challenging. Th1/Th2 ratio is lowered in patients with postoperative complications. Cholinergic anti-inflammatory pathways can inhibit tissue damage. The new substance PXL01 prevents adhesions. Postoperative infection causes incisional hernias. Hypothermia reduced human leukocyte antigen DR surface expression and delayed tumor necrosis factor clearance. Systems biology identified interferon regulatory factor 3 as the negative regulator of TLR signaling. Protective immunity could contribute defeating surgical infections. SUMMARY Systemic inflammation is the usual response to trauma. All organs seem to be involved and linked up in cybernetic systems aiming at reconstitution of homeostasis. Although knowledge is still fragmentary, it is already difficult to integrate known facts and new technologies are required for information processing. Defining criteria to develop therapeutic strategies requires much more insight into molecular mechanisms and cybernetics of organ systems.
Collapse
|
1370
|
De Luca A, Zelante T, D'Angelo C, Zagarella S, Fallarino F, Spreca A, Iannitti RG, Bonifazi P, Renauld JC, Bistoni F, Puccetti P, Romani L. IL-22 defines a novel immune pathway of antifungal resistance. Mucosal Immunol 2010; 3:361-73. [PMID: 20445503 DOI: 10.1038/mi.2010.22] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The role of IL-17 and Th17 cells in immunity vs. pathology associated with the human commensal Candida albicans remains controversial. Both positive and negative effects on immune resistance have been attributed to IL-17/Th17 in experimental candidiasis. In this study, we provide evidence that IL-22, which is also produced by Th17 cells, has a critical, first-line defense in candidiasis by controlling the growth of infecting yeasts as well as by contributing to the host's epithelial integrity in the absence of acquired Th1-type immunity. The two pathways are reciprocally regulated, and IL-22 is upregulated under Th1 deficiency conditions and vice versa. Whereas both IL-17A and F are dispensable for antifungal resistance, IL-22 mediates protection in IL-17RA-deficient mice, in which IL-17A contributes to disease susceptibility. Thus, our findings suggest that protective immunity to candidiasis is made up of a staged response involving an early, IL-22-dominated response followed by Th1/Treg reactivity that will prevent fungal dissemination and supply memory.
Collapse
Affiliation(s)
- A De Luca
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1371
|
Cai X, Wang J, Wang Y, Yang Y, Gao J, Fu W, Wang J, Xu D. Expression, purification and characterization of recombinant human interleukin-22 in Pichia pastoris. Mol Biol Rep 2010; 37:2609-13. [PMID: 19760099 DOI: 10.1007/s11033-009-9785-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 08/19/2009] [Indexed: 10/20/2022]
Abstract
Interleukin-22 (IL-22) is a member of the IL-10 family. Its potential in clinical use has been highlighted for its important roles in promoting antimicrobial defense and preventing epithelial damages. Previous studies have reported that IL-22 can be expressed using prokaryotic systems and purified from inclusion bodies, however the recovery rate was poor. To produce functional IL-22 with a high yield, human IL-22 was inserted into the eukaryotic expression vector pPICZalphaA and transformed into Pichia pastoris. The expression of recombinant human IL-22 (rhIL-22) was induced by methanol and accounted for about 85% of the total secreted proteins. A simple purification strategy was established to purify the rhIL-22 from the culture supernatant, yielding 100 mg/l at 90% purity by chromatography with a SP Sepharose FF column. Bioactivity analysis showed the purified rhIL-22 demonstrated a specific activity that was comparable with the commercial one. This study provides a new strategy for large-scale production of bioactive IL-22 for use in basic studies and therapeutic applications.
Collapse
Affiliation(s)
- Xin Cai
- Laboratory of Molecular Genetics, Beijing Institute of Basic Medical Sciences, 100850 Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
1372
|
Abstract
The cytokine interleukin-17 (IL-17) has received considerable attention since the discovery of a distinct CD4(+) T helper (T(H)) cell subset that produces it, known as the T(H)17 cell subset. Despite the fact that most of the recent literature describes IL-17 as a T cell-secreted cytokine, much of the IL-17 released during an inflammatory response is produced by innate immune cells. In this Review, we explore the many innate immune cell populations that are an early source of IL-17 in response to stress, injury or pathogens. These early sources have been shown to have a central role in the initiation of IL-17-dependent immune responses, even before the first CD4(+)T cell sees its cognate antigen and initiates the T(H)17 cell developmental programme.
Collapse
|
1373
|
Ness-Schwickerath KJ, Jin C, Morita CT. Cytokine requirements for the differentiation and expansion of IL-17A- and IL-22-producing human Vgamma2Vdelta2 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:7268-80. [PMID: 20483730 PMCID: PMC2965829 DOI: 10.4049/jimmunol.1000600] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human gammadelta T cells expressing the Vgamma2Vdelta2 TCR play important roles in immune responses to microbial pathogens by monitoring prenyl pyrophosphate isoprenoid metabolites. Most adult Vgamma2Vdelta2 cells are memory cytotoxic cells that produce IFN-gamma. Recently, murine gammadelta T cells were found to be major sources of IL-17A in antimicrobial and autoimmune responses. To determine if primate gammadelta T cells play similar roles, we characterized IL-17A and IL-22 production by Vgamma2Vdelta2 cells. IL-17A-producing memory Vgamma2Vdelta2 cells exist at low but significant frequencies in adult humans (1:2762 T cells) and at even higher frequencies in adult rhesus macaques. Higher levels of Vgamma2Vdelta2 cells produce IL-22 (1:1864 T cells), although few produce both IL-17A and IL-22. Unlike adult humans, in whom many IL-17A+ Vgamma2Vdelta2 cells also produce IFN-gamma (Tgammadelta1/17), the majority of adult macaques IL-17A+ Vdelta2 cells (Tgammadelta17) do not produce IFN-gamma. To define the cytokine requirements for Tgammadelta17 cells, we stimulated human neonatal Vgamma2Vdelta2 cells with the bacterial Ag, (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate, and various cytokines and mAbs in vitro. We find that IL-6, IL-1beta, and TGF-beta are required to generate Tgammadelta17 cells in neonates, whereas Tgammadelta1/17 cells additionally required IL-23. In adults, memory Tgammadelta1/17 and Tgammadelta17 cells required IL-23, IL-1beta, and TGF-beta, but not IL-6. IL-22-producing cells showed similar requirements. Both neonatal and adult IL-17A+ Vgamma2Vdelta2 cells expressed elevated levels of retinoid-related orphan receptor gammat. Our data suggest that, like Th17 alphabeta T cells, Vgamma2Vdelta2 T cells can be polarized into Tgammadelta17 and Tgammadelta1/17 populations with distinct cytokine requirements for their initial polarization and later maintenance.
Collapse
Affiliation(s)
- Kristin J. Ness-Schwickerath
- Division of Rheumatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, EMRB 400F, Iowa City, IA 52242 USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, EMRB 400F, Iowa City, IA 52242 USA
| | - Chenggang Jin
- Division of Rheumatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, EMRB 400F, Iowa City, IA 52242 USA
| | - Craig T. Morita
- Division of Rheumatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, EMRB 400F, Iowa City, IA 52242 USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa Carver College of Medicine, EMRB 400F, Iowa City, IA 52242 USA
| |
Collapse
|
1374
|
Mizoguchi A, Mizoguchi E. Animal models of IBD: linkage to human disease. Curr Opin Pharmacol 2010; 10:578-87. [PMID: 20860919 DOI: 10.1016/j.coph.2010.05.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2010] [Revised: 05/11/2010] [Accepted: 05/16/2010] [Indexed: 12/19/2022]
Abstract
Spontaneous development of intestinal inflammation in many different kinds of genetically engineered mice as well as the presence of numerous susceptibility genes in humans suggests that inflammatory bowel disease (IBD) is mediated by more complicated mechanisms than previously predicted. The human genetic studies implicate some major pathways in the pathogenesis of IBD, including epithelial defense against commensal microbiota, the IL-23/Th17 axis, and immune regulation. Murine IBD models, which are genetically engineered to lack some susceptibility genes, have been generated, and have provided useful insights into the therapeutic potential of targeting the susceptibility genes directly or their downstream pathways indirectly for IBD. This review summarizes current information related to the function of IBD-associated genes as derived from genetically engineered mouse models.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Molecular Pathology Unit, Massachusetts General Hospital, Boston, MA, USA.
| | | |
Collapse
|
1375
|
Sonnenberg GF, Nair MG, Kirn TJ, Zaph C, Fouser LA, Artis D. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. THE JOURNAL OF EXPERIMENTAL MEDICINE 2010. [PMID: 20498020 DOI: 10.1084/jem-20092054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-22 has both proinflammatory and tissue-protective properties depending on the context in which it is expressed. However, the factors that influence the functional outcomes of IL-22 expression remain poorly defined. We demonstrate that after administration of a high dose of bleomycin that induces acute tissue damage and airway inflammation and is lethal to wild-type (WT) mice, Th17 cell-derived IL-22 and IL-17A are expressed in the lung. Bleomycin-induced disease was ameliorated in Il22-/- mice or after anti-IL-22 monoclonal antibody (mAb) treatment of WT mice, indicating a proinflammatory/pathological role for IL-22 in airway inflammation. However, despite increased bleomycin-induced IL-22 production, Il17a-/- mice were protected from airway inflammation, suggesting that IL-17A may regulate the expression and/or proinflammatory properties of IL-22. Consistent with this, IL-17A inhibited IL-22 production by Th17 cells, and exogenous administration of IL-22 could only promote airway inflammation in vivo by acting in synergy with IL-17A. Anti-IL-22 mAb was delivered to Il17a-/- mice and was found to exacerbate bleomycin-induced airway inflammation, indicating that IL-22 is tissue protective in the absence of IL-17A. Finally, in an in vitro culture system, IL-22 administration protected airway epithelial cells from bleomycin-induced apoptosis, and this protection was reversed after coadministration of IL-17A. These data identify that IL-17A can regulate the expression, proinflammatory properties, and tissue-protective functions of IL-22, and indicate that the presence or absence of IL-17A governs the proinflammatory versus tissue-protective properties of IL-22 in a model of airway damage and inflammation.
Collapse
Affiliation(s)
- Gregory F Sonnenberg
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
1376
|
Sonnenberg GF, Nair MG, Kirn TJ, Zaph C, Fouser LA, Artis D. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. J Exp Med 2010; 207:1293-305. [PMID: 20498020 PMCID: PMC2882840 DOI: 10.1084/jem.20092054] [Citation(s) in RCA: 304] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 04/08/2010] [Indexed: 12/11/2022] Open
Abstract
IL-22 has both proinflammatory and tissue-protective properties depending on the context in which it is expressed. However, the factors that influence the functional outcomes of IL-22 expression remain poorly defined. We demonstrate that after administration of a high dose of bleomycin that induces acute tissue damage and airway inflammation and is lethal to wild-type (WT) mice, Th17 cell-derived IL-22 and IL-17A are expressed in the lung. Bleomycin-induced disease was ameliorated in Il22-/- mice or after anti-IL-22 monoclonal antibody (mAb) treatment of WT mice, indicating a proinflammatory/pathological role for IL-22 in airway inflammation. However, despite increased bleomycin-induced IL-22 production, Il17a-/- mice were protected from airway inflammation, suggesting that IL-17A may regulate the expression and/or proinflammatory properties of IL-22. Consistent with this, IL-17A inhibited IL-22 production by Th17 cells, and exogenous administration of IL-22 could only promote airway inflammation in vivo by acting in synergy with IL-17A. Anti-IL-22 mAb was delivered to Il17a-/- mice and was found to exacerbate bleomycin-induced airway inflammation, indicating that IL-22 is tissue protective in the absence of IL-17A. Finally, in an in vitro culture system, IL-22 administration protected airway epithelial cells from bleomycin-induced apoptosis, and this protection was reversed after coadministration of IL-17A. These data identify that IL-17A can regulate the expression, proinflammatory properties, and tissue-protective functions of IL-22, and indicate that the presence or absence of IL-17A governs the proinflammatory versus tissue-protective properties of IL-22 in a model of airway damage and inflammation.
Collapse
Affiliation(s)
| | - Meera G. Nair
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Thomas J. Kirn
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Colby Zaph
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Lynette A. Fouser
- Inflammation and Immunology–Pfizer BioTherapeutics Research and Development, Cambridge, MA 02140
| | - David Artis
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
1377
|
Kang YJ, Otsuka M, van den Berg A, Hong L, Huang Z, Wu X, Zhang DW, Vallance BA, Tobias PS, Han J. Epithelial p38alpha controls immune cell recruitment in the colonic mucosa. PLoS Pathog 2010; 6:e1000934. [PMID: 20532209 PMCID: PMC2880565 DOI: 10.1371/journal.ppat.1000934] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 05/03/2010] [Indexed: 11/19/2022] Open
Abstract
Intestinal epithelial cells (IECs) compose the first barrier against microorganisms in the gastrointestinal tract. Although the NF-kappaB pathway in IECs was recently shown to be essential for epithelial integrity and intestinal immune homeostasis, the roles of other inflammatory signaling pathways in immune responses in IECs are still largely unknown. Here we show that p38alpha in IECs is critical for chemokine expression, subsequent immune cell recruitment into the intestinal mucosa, and clearance of the infected pathogen. Mice with p38alpha deletion in IECs suffer from a sustained bacterial burden after inoculation with Citrobacter rodentium. These animals are normal in epithelial integrity and immune cell function, but fail to recruit CD4(+) T cells into colonic mucosal lesions. The expression of chemokines in IECs is impaired, which appears to be responsible for the impaired T cell recruitment. Thus, p38alpha in IECs contributes to the host immune responses against enteric bacteria by the recruitment of immune cells.
Collapse
Affiliation(s)
- Young Jun Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Motoyuki Otsuka
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Arjen van den Berg
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Lixin Hong
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe Huang
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiurong Wu
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Duan-Wu Zhang
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Bruce A. Vallance
- Division of Gastroenterology, BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Peter S. Tobias
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jiahuai Han
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
- The Key Laboratory of the Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
1378
|
Shen W, Durum SK. Synergy of IL-23 and Th17 cytokines: new light on inflammatory bowel disease. Neurochem Res 2010; 35:940-6. [PMID: 19915978 PMCID: PMC7241863 DOI: 10.1007/s11064-009-0091-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2009] [Indexed: 01/01/2023]
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, involve an interplay between host genetics and environmental factors including intestinal microbiota. Animal models of IBD have indicated that chronic inflammation can result from over-production of inflammatory responses or deficiencies in key negative regulatory pathways. Recent research advances in both T-helper 1 (Th1) and T-helper 17 (Th17) effect responses have offered new insights on the induction and regulation of mucosal immunity which is linked to the development of IBD. Th17 cytokines, such as IL-17 and IL-22, in combination with IL-23, play crucial roles in intestinal protection and homeostasis. IL-23 is expressed in gut mucosa and tends to orchestrate T-cell-independent pathways of intestinal inflammation as well as T cell dependent pathways mediated by cytokines produced by Th1 and Th17 cells. Th17 cells, generally found to be proinflammatory, have specific functions in host defense against infection by recruiting neutrophils and macrophages to infected tissues. Here we will review emerging data on those cytokines and their related regulatory networks that appear to govern the complex development of chronic intestinal inflammation; we will focus on how IL-23 and Th17 cytokines act coordinately to influence the balance between tolerance and immunity in the intestine.
Collapse
Affiliation(s)
- Wei Shen
- Laboratory of Molecular Immunoregulation, Cancer Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | | |
Collapse
|
1379
|
Yersinia enterocolitica promotes robust mucosal inflammatory T-cell immunity in murine neonates. Infect Immun 2010; 78:3595-608. [PMID: 20515925 DOI: 10.1128/iai.01272-09] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Mucosal immunity to gastrointestinal pathogens in early life has been studied only slightly. Recently, we developed an infection model in murine neonates using the gastroenteric pathogen Yersinia enterocolitica. Here, we report that oral infection of neonatal mice with low doses of virulent Y. enterocolitica leads to vigorous intestinal and systemic adaptive immunity. Y. enterocolitica infection promoted the development of anti-LcrV memory serum IgG1 and IgG2a responses of comparable affinity and magnitude to adult responses. Strikingly, neonatal mesenteric lymph node CD4(+) T cells produced Yersinia-specific gamma interferon (IFN-gamma) and interleukin-17A (IL-17A), exceeding adult levels. The robust T- and B-cell responses elicited in neonates exposed to Y. enterocolitica were associated with long-term protection against mucosal challenge with this pathogen. Using genetically deficient mice, we found that IFN-gamma and CD4(+) cells, but not B cells, are critical for protection of neonates during primary Y. enterocolitica infection. In contrast, adults infected with low bacterial doses did not require either cell population for protection. CD4-deficient neonatal mice adoptively transferred with CD4(+) cells from wild-type, IFN-gamma-deficient, or IL-17AF-deficient mice were equally protected from infection. These data demonstrate that inflammatory CD4(+) T cells are required for protection of neonatal mice and that this protection may not require CD4-derived IFN-gamma, IL-17A, or IL-17F. Overall, these studies support the idea that Y. enterocolitica promotes the development of highly inflammatory mucosal responses in neonates and that intestinal T-cell function may be a key immune component in protection from gastrointestinal pathogens in early life.
Collapse
|
1380
|
Onishi RM, Gaffen SL. Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology 2010; 129:311-21. [PMID: 20409152 DOI: 10.1111/j.1365-2567.2009.03240.x] [Citation(s) in RCA: 689] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Interleukin-17 (IL-17) has emerged as a central player in the mammalian immune system. Although this cytokine exerts a host-defensive role in many infectious diseases, it promotes inflammatory pathology in autoimmunity and other settings. A myriad of studies have focused on how IL-17-producing cells are generated. However, the means by which IL-17 achieves its effects, either for the benefit or the detriment of the host, are due in large part to the induction of new gene expression. Whereas many IL-17 target genes are common to different disease states, in some cases the effects of IL-17 differ depending on the target cell, infectious site or pathogen. Gene products induced by IL-17 include cytokines (IL-6, granulocyte-colony-stimulating factor, tumour necrosis factor-alpha), chemokines (CXCL1, CXCL2, CCL20, among many others), inflammatory effectors (acute-phase protesins, complement) and antimicrobial proteins (defensins, mucins). Different cell types appear to respond differently to IL-17 in terms of target gene expression, with notable differences seen in mesenchymal and epithelial cells compared with cells of haematopoietic origin. Here, we summarize the major IL-17 target genes that mediate this cytokine's activities in both autoimmune and chronic diseases as well as during various types of infections.
Collapse
Affiliation(s)
- Reiko M Onishi
- Department of Medicine, Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | |
Collapse
|
1381
|
|
1382
|
Bergstrom KSB, Kissoon-Singh V, Gibson DL, Ma C, Montero M, Sham HP, Ryz N, Huang T, Velcich A, Finlay BB, Chadee K, Vallance BA. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog 2010; 6:e1000902. [PMID: 20485566 PMCID: PMC2869315 DOI: 10.1371/journal.ppat.1000902] [Citation(s) in RCA: 469] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 04/08/2010] [Indexed: 12/20/2022] Open
Abstract
Despite recent advances in our understanding of the pathogenesis of attaching and effacing (A/E) Escherichia coli infections, the mechanisms by which the host defends against these microbes are unclear. The goal of this study was to determine the role of goblet cell-derived Muc2, the major intestinal secretory mucin and primary component of the mucus layer, in host protection against A/E pathogens. To assess the role of Muc2 during A/E bacterial infections, we inoculated Muc2 deficient (Muc2−/−) mice with Citrobacter rodentium, a murine A/E pathogen related to diarrheagenic A/E E. coli. Unlike wildtype (WT) mice, infected Muc2−/− mice exhibited rapid weight loss and suffered up to 90% mortality. Stool plating demonstrated 10–100 fold greater C. rodentium burdens in Muc2−/− vs. WT mice, most of which were found to be loosely adherent to the colonic mucosa. Histology of Muc2−/− mice revealed ulceration in the colon amid focal bacterial microcolonies. Metabolic labeling of secreted mucins in the large intestine demonstrated that mucin secretion was markedly increased in WT mice during infection compared to uninfected controls, suggesting that the host uses increased mucin release to flush pathogens from the mucosal surface. Muc2 also impacted host-commensal interactions during infection, as FISH analysis revealed C. rodentium microcolonies contained numerous commensal microbes, which was not observed in WT mice. Orally administered FITC-Dextran and FISH staining showed significantly worsened intestinal barrier disruption in Muc2−/− vs. WT mice, with overt pathogen and commensal translocation into the Muc2−/− colonic mucosa. Interestingly, commensal depletion enhanced C. rodentium colonization of Muc2−/− mice, although colonic pathology was not significantly altered. In conclusion, Muc2 production is critical for host protection during A/E bacterial infections, by limiting overall pathogen and commensal numbers associated with the colonic mucosal surface. Such actions limit tissue damage and translocation of pathogenic and commensal bacteria across the epithelium. Enteropathogenic E. coli (EPEC) and Enterohemorrhagic E. coli (EHEC) are important causes of diarrheal disease and other serious complications worldwide. Despite many studies addressing the pathogenic strategies used by these microbes, how the host protects itself from these pathogens is poorly understood. A critical question we address here is whether the thick mucus layer that overlies the intestinal surface plays a role in host protection. Since EPEC and EHEC do not infect mice efficiently, we used a related mouse pathogen called Citrobacter rodentium to infect and compare responses between wildtype mice and Muc2-deficient mice, which are defective in mucus production. We show that Muc2-deficient mice are extremely susceptible to C. rodentium infection-induced mortality and disease. Muc2-deficient mice were also colonized faster and had higher pathogen burdens throughout the experiment. Resident (non-pathogenic) bacteria were found to interact with C. rodentium and host tissues in Muc2-deficient mice, indicating Muc2 regulates all forms of intestinal microbiota at the gut surface. Deficiency in mucus production also contributed to increased leakiness of the gut, which allowed microbes to enter mucosal tissues. Our study shows that Muc2-dependent mucus production is critical for effective management of both pathogenic and non-pathogenic bacteria during infection by an EPEC/EHEC-like pathogen.
Collapse
Affiliation(s)
- Kirk S. B. Bergstrom
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Vanessa Kissoon-Singh
- Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Deanna L. Gibson
- Department of Biology and Physical Geography, Irving K. Barber School of Arts and Sciences, University of British Columbia-Okanagan, Kelowna, British Columbia, Canada
| | - Caixia Ma
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Marinieve Montero
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Ho Pan Sham
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Natasha Ryz
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Tina Huang
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Anna Velcich
- Department of Oncology, Albert Einstein Cancer Center/Montefiore Medical Center, Bronx, New York, United States of America
| | - B. Brett Finlay
- Michael Smith Laboratories and Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kris Chadee
- Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (KC); (BAV)
| | - Bruce A. Vallance
- Department of Pediatrics, Division of Gastroenterology, Child and Family Research Institute, Vancouver, British Columbia, Canada
- * E-mail: (KC); (BAV)
| |
Collapse
|
1383
|
Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu Rev Immunol 2010; 28:243-73. [PMID: 20192805 DOI: 10.1146/annurev-immunol-030409-101314] [Citation(s) in RCA: 386] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In mammals, the gastrointestinal tract harbors an extraordinarily dense and complex community of microorganisms. The gut microbiota provide strong selective pressure to the host to evolve adaptive immune responses required for the maintenance of local and systemic homeostasis. The continuous antigenic presence in the gut imposes a dynamic remodeling of gut-associated lymphoid tissues (GALT) and the selection of multiple layered strategies for immunoglobulin (Ig) A production. The composite and dynamic gut environment also necessitates heterogeneous, versatile, and convertible T cells, capable of inhibiting (Foxp3(+) T cells) or helping (T(FH) cells) local immune responses. In this review, we describe recent advances in our understanding of dynamic pathways that lead to IgA synthesis, in gut follicular structures and in extrafollicular sites, by T cell-dependent and T cell-independent mechanisms. We discuss the finely tuned regulatory mechanisms for IgA production and emphasize the role of mucosal IgA in the selection and maintenance of the appropriate microbial composition that is necessary for immune homeostasis.
Collapse
|
1384
|
Torchinsky MB, Blander JM. T helper 17 cells: discovery, function, and physiological trigger. Cell Mol Life Sci 2010; 67:1407-21. [PMID: 20054607 PMCID: PMC11115816 DOI: 10.1007/s00018-009-0248-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 12/17/2009] [Accepted: 12/22/2009] [Indexed: 01/19/2023]
Abstract
In the few years since their discovery, T helper 17 cells (T(H)17) have been shown to play an important role in host defense against infections, and in tissue inflammation during autoimmunity. T(H)17 cells produce IL-17, IL-21, IL-10, and IL-22 cytokines, and thus have broad effects on a variety of tissues. Notably, the requirement for the immunosuppressive cytokine TGF-beta along with the pro-inflammatory cytokine IL-6 for T(H)17 differentiation supports the intimate relationship between the T(H)17 subset and FOXP3(+) regulatory T cells. Here, we discuss current knowledge on effector functions and differentiation of the T(H)17 lineage. Furthermore, we now know of a physiological stimulus for T(H)17 differentiation: innate immune recognition of cells undergoing apoptosis as a direct result of infection induces unique development of this subset. As our knowledge of T(H)17 and T regulatory cells grows, we are building on a new framework for the understanding of effector T cell differentiation and the biology of CD4(+) T cell adaptive immune responses.
Collapse
Affiliation(s)
- Miriam Beer Torchinsky
- Department of Medicine, Immunology Institute, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029 USA
| | - J. Magarian Blander
- Department of Medicine, Immunology Institute, Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029 USA
| |
Collapse
|
1385
|
Abstract
Commensal bacteria are crucial for maturation and function of the mucosal immune system. However, the mechanisms of these interactions are poorly understood. In addition, the role of the composition of the microbiota and the importance of individual species in this community in stimulating different types of immunity are major unanswered questions. We recently showed that the balance between two major effector T cell populations in the intestine, IL-17(+) Th17 cells and Foxp3(+) Tregs, requires signals from commensal bacteria and is dependent on the composition of the intestinal microbiota. Comparison of microbiota from Th17 cell-deficient and Th17 cell-sufficient mice identified segmented filamentous bacteria (SFB) as capable of specifically inducing Th17 cells in the gut. SFB represent the first example of a commensal species that can skew the mucosal effector T cell balance and thus affect the immune fitness of the individual.
Collapse
Affiliation(s)
- Ivaylo I. Ivanov
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Dan R. Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
- Howard Hughes Medical Institute, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
1386
|
Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe HR, Magorien JE, Blauvelt A, Kolls JK, Cheung AL, Cheng G, Modlin RL, Miller LS. IL-17 is essential for host defense against cutaneous Staphylococcus aureus infection in mice. J Clin Invest 2010; 120:1762-73. [PMID: 20364087 PMCID: PMC2860944 DOI: 10.1172/jci40891] [Citation(s) in RCA: 512] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 01/27/2010] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus is the most common cause of skin and soft tissue infections, and rapidly emerging antibiotic-resistant strains are creating a serious public health concern. If immune-based therapies are to be an alternative to antibiotics, greater understanding is needed of the protective immune response against S. aureus infection in the skin. Although neutrophil recruitment is required for immunity against S. aureus, a role for T cells has been suggested. Here, we used a mouse model of S. aureus cutaneous infection to investigate the contribution of T cells to host defense. We found that mice deficient in gammadelta but not alphabeta T cells had substantially larger skin lesions with higher bacterial counts and impaired neutrophil recruitment compared with WT mice. This neutrophil recruitment was dependent upon epidermal Vgamma5+ gammadelta T cell production of IL-17, but not IL-21 and IL-22. Furthermore, IL-17 induction required IL-1, TLR2, and IL-23 and was critical for host defense, since IL-17R-deficient mice had a phenotype similar to that of gammadelta T cell-deficient mice. Importantly, gammadelta T cell-deficient mice inoculated with S. aureus and treated with a single dose of recombinant IL-17 had lesion sizes and bacterial counts resembling those of WT mice, demonstrating that IL-17 could restore the impaired immunity in these mice. Our study defines what we believe to be a novel role for IL-17-producing epidermal gammadelta T cells in innate immunity against S. aureus cutaneous infection.
Collapse
Affiliation(s)
- John S. Cho
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Eric M. Pietras
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Nairy C. Garcia
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Romela Irene Ramos
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - David M. Farzam
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Holly R. Monroe
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Julie E. Magorien
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Andrew Blauvelt
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Jay K. Kolls
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Ambrose L. Cheung
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Genhong Cheng
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Robert L. Modlin
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Lloyd S. Miller
- Division of Dermatology and
Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, California, USA.
Dermatology Service, Veterans Affairs Medical Center, Portland, Oregon, USA.
Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA.
Department of Microbiology and Immunology, Dartmouth Medical School, Hanover, New Hampshire, USA
| |
Collapse
|
1387
|
Dectin-2 Recognition of α-Mannans and Induction of Th17 Cell Differentiation Is Essential for Host Defense against Candida albicans. Immunity 2010; 32:681-91. [DOI: 10.1016/j.immuni.2010.05.001] [Citation(s) in RCA: 582] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/19/2010] [Accepted: 05/03/2010] [Indexed: 11/24/2022]
|
1388
|
Elevated IL-17 produced by TH17 cells promotes myeloma cell growth and inhibits immune function in multiple myeloma. Blood 2010; 115:5385-92. [PMID: 20395418 DOI: 10.1182/blood-2009-10-246660] [Citation(s) in RCA: 283] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Elevated cytokines in bone marrow (BM) micro-environment (interleukin-6 [IL-6], transforming growth factor-beta [TGF-beta], and IL-1beta) may play an important role in observed immune dysfunction in multiple myeloma (MM). As IL-6 and TGF-beta are important for the generation of T-helper 17 (T(H)17) cells, we evaluated and observed a significantly elevated baseline and induced frequency of T(h)17 cells in peripheral blood mononuclear cells (PBMCs) and BM mononuclear cells (BMMCs) from MM patients compared with healthy donors. We observed significant increase in levels of serum IL-17, IL-21, IL-22, and IL-23 in blood and BM in MM compared with healthy donors. We also observed that myeloma PBMCs after T(H)17 polarization significantly induced IL-1alpha, IL-13, IL-17, and IL-23 production compared with healthy donor PBMCs. We next observed that IL-17 promotes myeloma cell growth and colony formation via IL-17 receptor, adhesion to bone marrow stromal cells (BMSCs) as well as increased growth in vivo in murine xenograft model of human MM. Additionally, we have observed that combination of IL-17 and IL-22 significantly inhibited the production of T(H)1-mediated cytokines, including interferon-gamma (IFN-gamma), by healthy donor PBMCs. In conclusion, IL-17-producing T(h)17 cells play an important role in MM pathobiology and may be an important therapeutic target for anti-MM activity and to improve immune function.
Collapse
|
1389
|
Wilson MS, Feng CG, Barber DL, Yarovinsky F, Cheever AW, Sher A, Grigg M, Collins M, Fouser L, Wynn TA. Redundant and pathogenic roles for IL-22 in mycobacterial, protozoan, and helminth infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:4378-90. [PMID: 20220096 PMCID: PMC3170015 DOI: 10.4049/jimmunol.0903416] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
IL-22 is a member of the IL-10 cytokine family and signals through a heterodimeric receptor composed of the common IL-10R2 subunit and the IL-22R subunit. IL-10 and IL-22 both activate the STAT3 signaling pathway; however, in contrast to IL-10, relatively little is known about IL-22 in the host response to infection. In this study, using IL-22(-/-) mice, neutralizing Abs to IL-22, or both, we show that IL-22 is dispensable for the development of immunity to the opportunistic pathogens Toxoplasma gondii and Mycobacterium avium when administered via the i.p. or i.v. route, respectively. IL-22 also played little to no role in aerosol infections with Mycobacterium tuberculosis and in granuloma formation and hepatic fibrosis following chronic percutaneous infections with the helminth parasite Schistosoma mansoni. A marked pathogenic role for IL-22 was, however, identified in toxoplasmosis when infections were established by the natural oral route. Anti-IL-22 Ab-treated mice developed significantly less intestinal pathology than control Ab-treated mice even though both groups displayed similar parasite burdens. The decreased gut pathology was associated with reduced IL-17A, IL-17F, TNF-alpha, and IFN-gamma expression. In contrast to the prior observations of IL-22 protective effects in the gut, these distinct findings with oral T. gondii infection demonstrate that IL-22 also has the potential to contribute to pathogenic inflammation in the intestine. The IL-22 pathway has emerged as a possible target for control of inflammation in certain autoimmune diseases. Our findings suggest that few if any infectious complications would be expected with the suppression of IL-22 signaling.
Collapse
MESH Headings
- Animals
- Genetic Predisposition to Disease
- Inflammation Mediators/physiology
- Interleukins/deficiency
- Interleukins/physiology
- Intestinal Diseases, Parasitic/genetics
- Intestinal Diseases, Parasitic/immunology
- Intestinal Diseases, Parasitic/pathology
- Liver Diseases, Parasitic/genetics
- Liver Diseases, Parasitic/immunology
- Liver Diseases, Parasitic/pathology
- Meningitis/genetics
- Meningitis/immunology
- Meningitis/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Mycobacterium avium-intracellulare Infection/genetics
- Mycobacterium avium-intracellulare Infection/immunology
- Schistosomiasis mansoni/genetics
- Schistosomiasis mansoni/immunology
- Toxoplasmosis, Animal/genetics
- Toxoplasmosis, Animal/immunology
- Tuberculosis/genetics
- Tuberculosis/immunology
- Interleukin-22
Collapse
Affiliation(s)
- Mark S. Wilson
- Immunopathogensis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Carl G. Feng
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Daniel L. Barber
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Felix Yarovinsky
- University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390
| | | | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Michael Grigg
- Molecular Parasitology Unit, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Mary Collins
- Wyeth Research-Inflammation, Cambridge, MA 02140
| | | | - Thomas A. Wynn
- Immunopathogensis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
1390
|
Koutruba N, Emer J, Lebwohl M. Review of ustekinumab, an interleukin-12 and interleukin-23 inhibitor used for the treatment of plaque psoriasis. Ther Clin Risk Manag 2010; 6:123-41. [PMID: 20421912 PMCID: PMC2857612 DOI: 10.2147/tcrm.s5599] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of psoriasis is unknown, although it is generally accepted that this chronic inflammatory skin disorder is a complex autoimmune condition similar to other T-cell mediated disorders. Psoriasis imposes a heavy burden on the lifestyle of those affected due to the psychological, arthritic, and cutaneous morbidities; thus significant research has focused on the genetic and immunologic features of psoriasis in anticipation of more targeted, efficacious, and safe therapies. Recently, CD4(+) T helper (Th) 17 cells and interleukins (IL)-12 and -23 have been important in the pathogenesis of T-cell mediated disorders such as psoriasis and has influenced the development of medications that specifically target these key immunological players. Ustekinumab is a monoclonal antibody belonging to a newly developed class of biological, anti-cytokine medications that notably targets the p40 subunit of both IL-12 and -23, both naturally occurring proteins that are important in regulating the immune system and are understood to play a role in immune-mediated inflammatory disorders. Ustekinumab's safety and efficacy has been evaluated for the treatment of moderate-to-severe plaque psoriasis in 3 phase III clinical trials, 2 placebo-controlled (PHOENIX 1 and 2), and 1 comparator-controlled (ACCEPT) study which proved advantageous in patients who were treatment-naive, previously failed other immunosuppressive medications including cyclosporine or methotrexate, were unresponsive to phototherapy, or were unable to use or tolerate other therapies. Ustekinumab has also been investigated for other indications such as psoriatic arthritis, Crohn's disease, and relapsing/remitting multiple sclerosis. We present a concise review evaluating the evidence that supports the use of ustekinumab in the treatment of plaque psoriasis and other conditions.
Collapse
|
1391
|
Abstract
Interleukin-17 (IL-17) and IL-17-producing cells have been shown to play important roles in inflammation and the immune response. IL-17 is believed to be mainly produced by T helper 17 (Th17) cells, a unique helper T-cell subset different from Th1 and Th2 cells. Other subsets of T cells such as gammadeltaT and natural killer T (NKT) cells have also been found to produce IL-17 in response to innate stimuli. IL-17 acts as a proinflammatory cytokine that can induce the release of certain chemokines, cytokines, matrix metalloproteinases (MMPs) and antimicrobial peptides from mesenchymal and myeloid cells. This leads to the expansion and accumulation of neutrophils in the innate immune system and links innate and adaptive immunity in vivo. Furthermore, increasing evidence indicates that IL-17 and IL-17-producing cells are involved in the pathogenesis of various diseases such as allergies, autoimmune diseases, allograft transplantation and even malignancy. They may also play protective roles in host defense against infectious diseases and promote induction of cytotoxic T lymphocyte (CTL) responses against cancer. Targeting of the IL-17 axis is under investigation for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Sheng Xu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, China
| | | |
Collapse
|
1392
|
Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell 2010; 140:845-58. [PMID: 20303875 DOI: 10.1016/j.cell.2010.02.021] [Citation(s) in RCA: 821] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 02/11/2010] [Accepted: 02/13/2010] [Indexed: 12/22/2022]
Abstract
The vertebrate immune system is poised in a state of equilibrium that permits accurate and rapid protective responses against pathogens but curtails potential for causing harm to the host through targeting of "self" and provoking overexuberant inflammatory processes. In this Review we discuss this balance achieved in large part by interactions of different classes of T lymphocytes that have potent pro- or anti-inflammatory activity in the context of genetic and environmental factors, particularly the commensal microbiota.
Collapse
Affiliation(s)
- Dan R Littman
- Howard Hughes Medical Institute, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, 10016, USA.
| | | |
Collapse
|
1393
|
Thompson JS, Chu Y, Glass JF, Brown SA. Absence of IL-23p19 in donor allogeneic cells reduces mortality from acute GVHD. Bone Marrow Transplant 2010; 45:712-22. [PMID: 19718070 DOI: 10.1038/bmt.2009.215] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Revised: 06/17/2009] [Accepted: 07/09/2009] [Indexed: 02/05/2023]
Abstract
The p19 dimer of interleukin 23 (IL-23) has been reported to have a major role in the pathogenesis of many experimental and clinical autoimmune diseases and may also have a prominent role in transplantation. We reasoned that deficiency of p19 in the allogeneic donor transplant might reduce the inflammation caused by acute GVHD (aGVHD). The major histocompatibility complex-2 (H2(d)) BALB/c mice were subjected to 8.5 Gy TBI, followed by transplantation with 10 x 10(6) BM and 2.5 x 10(6) spleen cells from H2(d) BALB/c, H2(b) C57Bl/6 (B6) or H2(b) p19-/- donors. In all, 75% of the p19-/- transplanted mice survived, compared with only 12.5% of the B6 transplanted mice. This superior survival is correlated with significantly less severe aGVHD, absence of p19 after transplantation, less upregulation of mRNA and lower serum levels of IL-17 as compared with the B6 transplants. TBI alone significantly upregulated transforming growth factor-beta (TGF-beta), IL-6 and p19 mRNA levels in host BALB/c mice, possibly providing the milieu to induce IL-17 in p19-/- donor cells. IL-22, another cytokine, the induction of which in T-helper 17 (Th17) cells is supported by p19, was upregulated in BALB/c hosts but not in transplanted B6 or p19 donor cells, and may not have had a major role in modifying aGVHD.
Collapse
Affiliation(s)
- J S Thompson
- Veterans Affairs Medical Center, Department of Internal Medicine, University of Kentucky, Lexington, KY 40502, USA.
| | | | | | | |
Collapse
|
1394
|
Baba N, Rubio M, Sarfati M. Interplay between CD45RA+ regulatory T cells and TNF-alpha in the regulation of human Th17 differentiation. Int Immunol 2010; 22:237-44. [PMID: 20181653 DOI: 10.1093/intimm/dxp130] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The balance between effector CD4(+) T cells secreting IL-17 (T(h)17) and regulatory T cells (Treg) plays an important role in autoimmune disorders that include rheumatoid arthritis (RA) and Crohn's disease. Tumor necrosis factor (TNF)-alpha is a key pro-inflammatory cytokine that contributes to disease pathogenesis. We investigated the interplay between CD45RA(+) Treg and TNF-alpha in the regulation of human T(h)17 differentiation. We found that CD45RA(+) Treg promoted while TNF-alpha inhibited naive CD4(+) T-cell differentiation into IL-17 and CCL20 co-expressing T(h)17 cells without influencing their IL-22 release. Unexpectedly, CD45RA(+) Treg depletion abrogated TNF-alpha suppressive function. Finally, dendritic cell-derived TNF-alpha suppressed the development of IL-17(+)CCL20(+) expressing T(h)17 cells. In conclusion, CD45RA(+) Treg positively governs human T(h)17 development, which is impaired by TNF-alpha. We propose that TNF-alpha may represent a negative feedback mechanism to control IL-17/CCL20- but not IL-22-associated autoimmune pathologies.
Collapse
Affiliation(s)
- Nobuyasu Baba
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), University of Montreal, Montreal, Quebec, Canada
| | | | | |
Collapse
|
1395
|
Alam MS, Maekawa Y, Kitamura A, Tanigaki K, Yoshimoto T, Kishihara K, Yasutomo K. Notch signaling drives IL-22 secretion in CD4+ T cells by stimulating the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2010; 107:5943-8. [PMID: 20231432 PMCID: PMC2851859 DOI: 10.1073/pnas.0911755107] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
CD4(+) helper T (Th) cells differentiate toward distinct effector cell lineages characterized by their distinct cytokine expression patterns and functions. Multiple Th cell populations secrete IL-22 that contributes to both protective and pathological inflammatory responses. Although the differentiation of IL-22-producing Th cells is controlled by the aryl hydrocarbon receptor (AhR), little is known about the regulatory mechanisms inducing physiological stimulators for AhR. Here, we show that Notch signaling enhances IL-22 production by CD4(+) T cells by a mechanism involving AhR stimulation. Notch-mediated stimulation of CD4(+) T cells increased the production of IL-22 even in the absence of STAT3. CD4(+) T cells from RBP-J-deficient mice had little ability to produce IL-22 through T cell receptor-mediated stimulation. RBP-J-deficient mice were highly susceptible to the detrimental immunopathology associated with ConA-induced hepatitis with little IL-22 production by CD4(+) T cells. Exogenous IL-22 protected RBP-J-deficient mice from ConA-induced hepatitis. Notch signaling promoted production of endogenous stimulators for AhR, which further augmented IL-22 secretion. Our studies identify a Notch-AhR axis that regulates IL-22 expression and fine-tunes immune system control of inflammatory responses.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Differentiation
- Female
- Hepatitis, Animal/etiology
- Hepatitis, Animal/immunology
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/deficiency
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Interleukins/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Structure, Tertiary
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Notch/chemistry
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
- STAT3 Transcription Factor/deficiency
- STAT3 Transcription Factor/genetics
- Signal Transduction
- Interleukin-22
Collapse
Affiliation(s)
- Muhammad Shamsul Alam
- Department of Immunology and Parasitology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Yoichi Maekawa
- Department of Immunology and Parasitology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Akiko Kitamura
- Department of Immunology and Parasitology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan
| | - Kenji Tanigaki
- Research Institute, Shiga Medical Center, Shiga 520-2192, Japan
| | - Takayuki Yoshimoto
- Intractable Immune System Disease Research Center, Tokyo Medical University, Tokyo 160-8402, Japan; and
| | - Kenji Kishihara
- Department of Immunology and Parasitology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan
- Department of Immunology, Nagasaki International University, Saseho 859-3298, Japan
| | - Koji Yasutomo
- Department of Immunology and Parasitology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8503, Japan
| |
Collapse
|
1396
|
Infection and apoptosis as a combined inflammatory trigger. Curr Opin Immunol 2010; 22:55-62. [PMID: 20137905 DOI: 10.1016/j.coi.2010.01.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 01/05/2010] [Accepted: 01/15/2010] [Indexed: 01/04/2023]
Abstract
While inflammatory phagocytosis of microbial pathogens and non-inflammatory phagocytosis of apoptotic cells have each been studied extensively, the consequences of innate immune recognition of host cells undergoing apoptosis as a direct result of infection are unclear. In this situation, the innate immune system is confronted with mixed signals, those from apoptotic cells and those from the infecting pathogen. Nuclear receptor activation has been implicated downstream of apoptotic cell recognition while Toll-like receptors are the prototypical inflammatory receptors engaged during infection. When the two signals combine, a new set of events takes place beginning with transrepression of a subset of inflammatory-response genes and ending with the induction of a T helper-17 adaptive immune response. This response is best suited for clearing the infecting pathogen and repairing the damage that occurred to the host tissue during infection.
Collapse
|
1397
|
Gamma interferon produced by antigen-specific CD4+ T cells regulates the mucosal immune responses to Citrobacter rodentium infection. Infect Immun 2010; 78:2653-66. [PMID: 20351140 DOI: 10.1128/iai.01343-09] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Citrobacter rodentium, a murine model pathogen for enteropathogenic Escherichia coli, colonizes the surface of intestinal epithelial cells and causes mucosal inflammation. This bacterium is an ideal model for investigating pathogen-host immune interactions in the gut. It is well known that gene transcripts for Th1 cytokines are highly induced in colonic tissue from mice infected with C. rodentium. However, it remains to be seen whether the Th1 or Th2 cytokines produced by antigen-specific CD4(+) T cells provide effective regulation of the host immune defense against C. rodentium infection. To investigate the antigen-specific immune responses, C. rodentium expressing ovalbumin (OVA-C. rodentium), a model antigen, was generated and used to define antigen-specific responses under gamma interferon (IFN-gamma)-deficient or interleukin-4 (IL-4)-deficient conditions in vivo. The activation of antigen-specific CD4(+) T cells and macrophage phagocytosis were evaluated in the presence of IFN-gamma or IL-4 in vitro. IFN-gamma-deficient mice exhibited a loss of body weight and a higher bacterial concentration in feces during OVA-C. rodentium infection than C57BL/6 (wild type) or IL-4-deficient mice. This occurred through the decreased efficiency of macrophage phagocytosis and the activation of antigen-specific CD4(+) T cells. Furthermore, a deficiency in antigen-specific CD4(+) T-cell-expressed IFN-gamma led to a higher susceptibility to mucosal and gut-derived systemic OVA-C. rodentium infection. These results show that the IFN-gamma produced by antigen-specific CD4(+) T cells plays an important role in the defense against C. rodentium.
Collapse
|
1398
|
Wang Y, Koroleva EP, Kruglov AA, Kuprash DV, Nedospasov SA, Fu YX, Tumanov AV. Lymphotoxin beta receptor signaling in intestinal epithelial cells orchestrates innate immune responses against mucosal bacterial infection. Immunity 2010; 32:403-13. [PMID: 20226692 PMCID: PMC2878123 DOI: 10.1016/j.immuni.2010.02.011] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 12/16/2009] [Accepted: 01/21/2010] [Indexed: 11/26/2022]
Abstract
Epithelial cells provide the first line of defense against mucosal pathogens; however, their coordination with innate and adaptive immune cells is not well understood. Using mice with conditional gene deficiencies, we found that lymphotoxin (LT) from innate cells expressing transcription factor RORgammat, but not from adaptive T and B cells, was essential for the control of mucosal C. rodentium infection. We demonstrate that the LTbetaR signaling was required for the regulation of the early innate response against infection. Furthermore, we have revealed that LTbetaR signals in gut epithelial cells and hematopoietic-derived cells coordinate to protect the host from infection. We further determined that LTbetaR signaling in intestinal epithelial cells was required for recruitment of neutrophils to the infection site early during infection via production of CXCL1 and CXCL2 chemokines. These results support a model wherein LT from RORgammat(+) cells orchestrates the innate immune response against mucosal microbial infection.
Collapse
Affiliation(s)
- Yugang Wang
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | | | - Andrei A. Kruglov
- German Rheumatism Research Center (DRFZ), the Leibnitz Institute, Berlin, Germany
| | - Dmitry V. Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergei A. Nedospasov
- German Rheumatism Research Center (DRFZ), the Leibnitz Institute, Berlin, Germany
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Yang-Xin Fu
- The University of Chicago, Department of Pathology, Chicago, Illinois
| | - Alexei V. Tumanov
- The University of Chicago, Department of Pathology, Chicago, Illinois
| |
Collapse
|
1399
|
Abstract
Natural killer (NK) cells are a subset of lymphocytes that kill virus-infected or cancerous cells and influence adaptive immune responses via production of inflammatory cytokines. Unlike B and T lymphocytes, no transcription factors have been identified that are essential for the emergence of NK cell progenitors from their multipotent precursors. We argue that this dearth of essential factors is because of the expression of redundant transcription factors that function at the earliest stages of development. However, multiple essential transcription factors have been identified at later stages of development. Recent studies have revealed novel subsets of NK cells with differing potential for target cell lysis and cytokine production. How these subsets arise from the conventional pathway of NK cell development and identification of the transcriptional networks that control their development are major challenges for future studies.
Collapse
|
1400
|
Mortimer L, Chadee K. The immunopathogenesis of Entamoeba histolytica. Exp Parasitol 2010; 126:366-80. [PMID: 20303955 DOI: 10.1016/j.exppara.2010.03.005] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2009] [Revised: 03/08/2010] [Accepted: 03/12/2010] [Indexed: 12/17/2022]
Abstract
Amebiasis is the disease caused by the enteric dwelling protozoan parasite Entamoeba histolytica. The WHO considers amebiasis as one of the major health problems in developing countries; it is surpassed by only malaria and schistosomiasis for death caused by parasitic infection. E. histolytica primarily lives in the colon as a harmless commensal, but is capable of causing devastating dysentery, colitis and liver abscess. What triggers the switch to a pathogenic phenotype and the onset of disease is unknown. We are becoming increasingly aware of the complexity of the host-parasite interaction. During chronic stages of amebiasis, the host develops an immune response that is incapable of eliminating tissue resident parasites, while the parasite actively immunosuppresses the host. However, most individuals with symptomatic infections succumb only to an episode of dysentery. Why most halt invasion and a minority progress to chronic disease remains poorly understood. This review presents a current understanding of the immune processes that shape the outcome of E. histolytica infections during its different stages.
Collapse
Affiliation(s)
- Leanne Mortimer
- Faculty of Medicine, Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Centre, 3330 Hospital Dr. NW, Calgary, Alberta, Canada
| | | |
Collapse
|