Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Sci Rep ; 4: 5865, 2014 Aug 11.
Article in English | MEDLINE | ID: mdl-25109392

ABSTRACT

In a murine model of allergic asthma, we found that Tyk-2((-/-)) asthmatic mice have induced peribronchial collagen deposition, mucosal type mast cells in the lung, IRF4 and hyperproliferative lung Th2 CD4(+) effector T cells over-expressing IL-3, IL-4, IL-5, IL-10 and IL-13. We also observed increased Th9 cells expressing IL-9 and IL-10 as well as T helper cells expressing IL-6, IL-10 and IL-21 with a defect in IL-17A and IL-17F production. This T helper phenotype was accompanied by increased SOCS3 in the lung of Tyk-2 deficient asthmatic mice. Finally, in vivo treatment with rIL-17A inhibited local CD4(+)CD25(+)Foxp3(+) T regulatory cells as well as Th2 cytokines without affecting IL-9 in the lung. These results suggest a role of Tyk-2 in different subsets of T helper cells mediated by SOCS3 regulation that is relevant for the treatment of asthma, cancer and autoimmune diseases.


Subject(s)
Asthma/pathology , T-Lymphocytes, Helper-Inducer/metabolism , TYK2 Kinase/metabolism , Th17 Cells/metabolism , Animals , Asthma/immunology , Asthma/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-17/pharmacology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Phenotype , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , TYK2 Kinase/genetics , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism
2.
Sci Rep ; 3: 1754, 2013.
Article in English | MEDLINE | ID: mdl-23628948

ABSTRACT

IL-6 plays a central role in supporting pathological T(H2) and T(H17) cell development and inhibiting the protective T regulatory cells in allergic asthma. T(H17) cells have been demonstrated to regulate allergic asthma in general and T-bet-deficiency-induced asthma in particular. Here we found an inverse correlation between T-bet and Il-6 mRNA expression in asthmatic children. Moreover, experimental subcutaneous immunotherapy (SIT) in T-bet((-/-)) mice inhibited IL-6, IL-21R and lung T(H17) cells in a setting of asthma. Finally, local delivery of an anti-IL-6R antibody in T-bet((-/-)) mice resulted in the resolution of this allergic trait. Noteworthy, BATF, crucial for the immunoglobulin-class-switch and T(H2),T(H17) development, was found down-regulated in the lungs of T-bet((-/-)) mice after SIT and after treatment with anti-IL-6R antibody, indicating a critical role of IL-6 in controlling BATF/IRF4 integrated functions in T(H2), T(H17) cells and B cells also in a T-bet independent fashion in allergic asthma.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Interferon Regulatory Factors/immunology , Interleukin-6/immunology , T-Box Domain Proteins/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Transcription Factors/immunology , Animals , Antibodies/metabolism , Asthma/genetics , Asthma/immunology , Asthma/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Child , Child, Preschool , Female , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunotherapy/methods , Interferon Regulatory Factors/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/immunology , Lung/metabolism , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Interleukin-21/genetics , Receptors, Interleukin-21/immunology , Receptors, Interleukin-21/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Receptors, Interleukin-6/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Th17 Cells/metabolism , Th2 Cells/metabolism , Transcription Factors/metabolism
3.
Cancer Res ; 72(17): 4340-50, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22738913

ABSTRACT

NFAT transcription factors control T-cell activation and function. Specifically, the transcription factor NFATc2 affects the regulation of cell differentiation and growth and plays a critical role in the development of colonic inflammation. Here, we used an experimental model of colitis-associated colorectal carcinoma to investigate the contribution of NFATc2 to the promotion of colonic tumors. Compared with wild-type animals that readily presented with multiple colon tumors, NFATc2-deficient mice were protected from tumor development. This observed decrease in colonic tumor progression was associated with reduced endoscopic inflammation, increased apoptosis of lamina propria T lymphocytes, and significantly reduced levels of the critical proinflammatory cytokines interleukin (IL)-21 and IL-6. Administration of hyper IL-6 abrogated protection from tumor progression in NFATc2-knockout mice and restored tumor incidence to control levels. Taken together, our findings highlight a pivotal role for NFATc2 in the establishment of inflammation-associated colorectal tumors mediated by control of IL-6 expression.


Subject(s)
Colitis/genetics , Colonic Neoplasms/genetics , Interleukin-6/immunology , NFATC Transcription Factors/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Colitis/complications , Colitis/immunology , Colonic Neoplasms/etiology , Colonic Neoplasms/immunology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , NFATC Transcription Factors/immunology
4.
J Allergy Clin Immunol ; 129(4): 1126-35, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22236728

ABSTRACT

BACKGROUND: Humanized murine models comprise a new tool to analyze novel therapeutic strategies for allergic diseases of the intestine. OBJECTIVE: In this study we developed a human PBMC-engrafted murine model of allergen-driven gut inflammation and analyzed the underlying immunologic mechanisms. METHODS: Nonobese diabetic (NOD)-scid-γc(-/-) mice were injected intraperitoneally with human PBMCs from allergic donors together with the respective allergen or not. Three weeks later, mice were challenged with the allergen orally or rectally, and gut inflammation was monitored with a high-resolution video miniendoscopic system, as well as histologically. RESULTS: Using the aeroallergens birch or grass pollen as model allergens and, for some donors, also hazelnut allergen, we show that allergen-specific human IgE in murine sera and allergen-specific proliferation and cytokine production of human CD4(+) T cells recovered from spleens after 3 weeks could only be measured in mice treated with PBMCs plus allergen. Importantly, these mice had the highest endoscopic scores evaluating translucent structure, granularity, fibrin, vascularity, and stool after oral or rectal allergen challenge and a strong histologic inflammation of the colon. Analyzing the underlying mechanisms, we demonstrate that allergen-associated colitis was dependent on IgE, human IgE receptor-expressing effector cells, and the mediators histamine and platelet-activating factor. CONCLUSION: These results demonstrate that allergic gut inflammation can be induced in human PBMC-engrafted mice, allowing the investigation of pathophysiologic mechanisms of allergic diseases of the intestine and evaluation of therapeutic interventions.


Subject(s)
Allergens/immunology , Gastritis/immunology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Leukocytes, Mononuclear/transplantation , Administration, Oral , Administration, Rectal , Allergens/administration & dosage , Animals , Antibody Specificity/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Disease Models, Animal , Gastritis/pathology , Gastritis/prevention & control , Histamine Antagonists/metabolism , Humans , Hypersensitivity/pathology , Hypersensitivity/prevention & control , Immunoglobulin E/blood , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, SCID , Platelet Membrane Glycoproteins/antagonists & inhibitors , Pollen/immunology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, IgE/metabolism , Spleen/immunology
5.
EMBO Mol Med ; 3(6): 348-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538995

ABSTRACT

IL-28 (IFN-λ) cytokines exhibit potent antiviral and antitumor function but their full spectrum of activities remains largely unknown. Recently, IL-28 cytokine family members were found to be profoundly down-regulated in allergic asthma. We now reveal a novel role of IL-28 cytokines in inducing type 1 immunity and protection from allergic airway disease. Treatment of wild-type mice with recombinant or adenovirally expressed IL-28A ameliorated allergic airway disease, suppressed Th2 and Th17 responses and induced IFN-γ. Moreover, abrogation of endogenous IL-28 cytokine function in IL-28Rα(-/-) mice exacerbated allergic airway inflammation by augmenting Th2 and Th17 responses, and IgE levels. Central to IL-28A immunoregulatory activity was its capacity to modulate lung CD11c(+) dendritic cell (DC) function to down-regulate OX40L, up-regulate IL-12p70 and promote Th1 differentiation. Consistently, IL-28A-mediated protection was absent in IFN-γ(-/-) mice or after IL-12 neutralization and could be adoptively transferred by IL-28A-treated CD11c(+) cells. These data demonstrate a critical role of IL-28 cytokines in controlling T cell responses in vivo through the modulation of lung CD11c(+) DC function in experimental allergic asthma.


Subject(s)
Asthma/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Th1 Cells/immunology , Animals , Asthma/pathology , Asthma/therapy , CD11c Antigen/metabolism , Cytokines/genetics , Down-Regulation , Lung/cytology , Lung/immunology , Mice , OX40 Ligand/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Th1 Cells/cytology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
6.
Inflamm Bowel Dis ; 17(6): 1343-58, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21305677

ABSTRACT

BACKGROUND: The transcription factor IRF4 is involved in several T-cell-dependent chronic inflammatory diseases. To elucidate the mechanisms for pathological cytokine production in colitis, we addressed the role of the IRF transcription factors in human inflammatory bowel disease (IBD) and experimental colitis. METHODS: IRF levels and cytokine production in IBD patients were studied as well as the effects of IRF4 deficiency in experimental colitis. RESULTS: In contrast to IRF1, IRF5, and IRF8, IRF4 expression in IBD was augmented in the presence of active inflammation. Furthermore, IRF4 levels significantly correlated with IL-6 and IL-17 mRNA expression and to a lesser extent with IL-22 mRNA expression in IBD. To further explore the role of IRF4 under in vivo conditions, we studied IRF4-deficient and wildtype mice in experimental colitis. In contrast to DSS colitis, IRF4 deficiency was protective in T-cell-dependent transfer colitis associated with reduced RORα/γt levels and impaired IL-6, IL-17a, and IL-22 production, suggesting that IRF4 acts as a master regulator of mucosal Th17 cell differentiation. Subsequent mechanistic studies using database analysis, chromatin immunoprecipitation, and electrophoretic mobility shift assays identified a novel IRF4 binding site in the IL-17 gene promoter. Overexpression of IRF4 using retroviral infection induced IL-17 production and IL-17 together with IL-6 induced RORγt expression. CONCLUSIONS: IRF4 can directly bind to the IL-17 promotor and induces mucosal RORγt levels and IL-17 gene expression thereby controlling Th17-dependent colitis. Targeting of this molecular mechanism may lead to novel therapeutic approaches in human IBD.


Subject(s)
Colitis/genetics , Inflammatory Bowel Diseases/genetics , Interferon Regulatory Factors/genetics , Interleukin-17/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th17 Cells/metabolism , Adult , Animals , Colitis/metabolism , Colitis, Ulcerative/genetics , Colitis, Ulcerative/metabolism , Crohn Disease/genetics , Crohn Disease/metabolism , Electrophoretic Mobility Shift Assay , Female , Humans , Inflammatory Bowel Diseases/metabolism , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/physiology , Interleukin-17/metabolism , Interleukin-17/physiology , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-6/physiology , Male , Mice , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Th17 Cells/physiology
7.
Int Immunol ; 23(1): 1-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21135031

ABSTRACT

Signal transducer and activator of transcription (STAT)-3 inhibitors play an important role in regulating immune responses. Galiellalactone (GL) is a fungal secondary metabolite known to interfere with the binding of phosphorylated signal transducer and activator of transcription (pSTAT)-3 as well of pSTAT-6 dimers to their target DNA in vitro. Intra nasal delivery of 50 µg GL into the lung of naive Balb/c mice induced FoxP3 expression locally and IL-10 production and IL-12p40 in RNA expression in the airways in vivo. In a murine model of allergic asthma, GL significantly suppressed the cardinal features of asthma, such as airway hyperresponsiveness, eosinophilia and mucus production, after sensitization and subsequent challenge with ovalbumin (OVA). These changes resulted in induction of IL-12p70 and IL-10 production by lung CD11c(+) dendritic cells (DCs) accompanied by an increase of IL-3 receptor α chain and indoleamine-2,3-dioxygenase expression in these cells. Furthermore, GL inhibited IL-4 production in T-bet-deficient CD4(+) T cells and down-regulated the suppressor of cytokine signaling-3 (SOCS-3), also in the absence of STAT-3 in T cells, in the lung in a murine model of asthma. In addition, we found reduced amounts of pSTAT-5 in the lung of GL-treated mice that correlated with decreased release of IL-2 by lung OVA-specific CD4(+) T cells after treatment with GL in vitro also in the absence of T-bet. Thus, GL treatment in vivo and in vitro emerges as a novel therapeutic approach for allergic asthma by modulating lung DC phenotype and function resulting in a protective response via CD4(+)FoxP3(+) regulatory T cells locally.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Lactones/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Administration, Intranasal , Animals , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/chemistry , Anti-Asthmatic Agents/isolation & purification , Anti-Asthmatic Agents/pharmacology , Asthma/immunology , CD11c Antigen/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-4/biosynthesis , Lactones/administration & dosage , Lactones/chemistry , Lung/immunology , Mice , Mice, Inbred BALB C , Receptors, Interleukin-3/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Box Domain Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
8.
Cancer Res ; 69(7): 3069-76, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318584

ABSTRACT

The Ca(2+)-regulated calcineurin/nuclear factor of activated T cells (NFAT) cascade controls alternative pathways of T-cell activation and peripheral tolerance. Here, we describe reduction of NFATc2 mRNA expression in the lungs of patients with bronchial adenocarcinoma. In a murine model of bronchoalveolar adenocarcinoma, mice lacking NFATc2 developed more and larger solid tumors than wild-type littermates. The extent of central tumor necrosis was decreased in the tumors in NFATc2((-/-)) mice, and this finding was associated with reduced tumor necrosis factor-alpha and interleukin-2 (IL-2) production by CD8(+) T cells. Adoptive transfer of CD8(+) T cells of NFATc2((-/-)) mice induced transforming growth factor-beta(1) in the airways of recipient mice, thus supporting CD4(+)CD25(+)Foxp-3(+)glucocorticoid-induced tumor necrosis factor receptor (GITR)(+) regulatory T (T(reg)) cell survival. Finally, engagement of GITR in NFATc2((-/-)) mice induced IFN-gamma levels in the airways, reversed the suppression by T(reg) cells, and costimulated effector CD4(+)CD25(+) (IL-2Ralpha) and memory CD4(+)CD127(+) (IL-7Ralpha) T cells, resulting in abrogation of carcinoma progression. Agonistic signaling through GITR, in the absence of NFATc2, thus emerges as a novel possible strategy for the treatment of human bronchial adenocarcinoma in the absence of NFATc2 by enhancing IL-2Ralpha(+) effector and IL-7Ralpha(+) memory-expressing T cells.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/immunology , Bronchial Neoplasms/genetics , Bronchial Neoplasms/immunology , CD8-Positive T-Lymphocytes/immunology , NFATC Transcription Factors/biosynthesis , Adenocarcinoma/metabolism , Animals , Bronchial Neoplasms/metabolism , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Glucocorticoid-Induced TNFR-Related Protein , Humans , Interferon-gamma , Interleukin-2/biosynthesis , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Receptors, Interleukin-7/biosynthesis , Receptors, Interleukin-7/immunology , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/immunology , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/immunology , Transcription, Genetic , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/immunology , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
9.
J Immunol ; 181(9): 6148-57, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941205

ABSTRACT

EBV-induced gene 3 (EBI-3) codes for a soluble type I receptor homologous to the p40 subunit of IL-12 that is expressed by APCs following activation. In this study, we assessed the role of EBI-3 in a model of lung melanoma metastasis. Intravenous injection of the B16-F10 cell line resulted in a significant reduction of lung tumor metastasis in EBI-3(-/-) recipient mice compared with wild-type mice. The immunological finding accompanying this effect was the expansion of a newly described cell subset called IFN-gamma producing killer dendritic cells associated with CD8(+) T cell responses in the lung of EBI-3(-/-) mice including IFN-gamma release and TNF-alpha-induced programmed tumor cell death. Depletion of CD8(+) T cells as well as targeting T-bet abrogated the protective effects of EBI-3 deficiency on lung melanoma metastases. Finally, adoptive transfer of EBI-3(-/-) CD8(+) T cells into tumor bearing wild-type mice inhibited lung metastasis in recipient mice. Taken together, these data demonstrate that targeting EBI-3 leads to a T-bet-mediated antitumor CD8(+) T cell responses in the lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Cytotoxicity, Immunologic/genetics , Gene Knockout Techniques , Immunologic Surveillance/genetics , Injections, Intravenous , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Neoplasm Transplantation , Receptors, Cytokine/physiology , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology
10.
J Exp Med ; 205(9): 2099-110, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18710929

ABSTRACT

The nuclear factor of activated T cells (NFAT) family of transcription factors controls calcium signaling in T lymphocytes. In this study, we have identified a crucial regulatory role of the transcription factor NFATc2 in T cell-dependent experimental colitis. Similar to ulcerative colitis in humans, the expression of NFATc2 was up-regulated in oxazolone-induced chronic intestinal inflammation. Furthermore, NFATc2 deficiency suppressed colitis induced by oxazolone administration. This finding was associated with enhanced T cell apoptosis in the lamina propria and strikingly reduced production of IL-6, -13, and -17 by mucosal T lymphocytes. Further studies using knockout mice showed that IL-6, rather than IL-23 and -17, are essential for oxazolone colitis induction. Administration of hyper-IL-6 blocked the protective effects of NFATc2 deficiency in experimental colitis, suggesting that IL-6 signal transduction plays a major pathogenic role in vivo. Finally, adoptive transfer of IL-6 and wild-type T cells demonstrated that oxazolone colitis is critically dependent on IL-6 production by T cells. Collectively, these results define a unique regulatory role for NFATc2 in colitis by controlling mucosal T cell activation in an IL-6-dependent manner. NFATc2 in T cells thus emerges as a potentially new therapeutic target for inflammatory bowel diseases.


Subject(s)
Interleukin-6/metabolism , NFATC Transcription Factors/metabolism , T-Lymphocytes/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Humans , Interleukin-13/biosynthesis , Interleukin-17/biosynthesis , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Models, Biological , Oxazolone/pharmacology
11.
J Clin Invest ; 118(7): 2415-26, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18535667

ABSTRACT

The proinflammatory cytokine IL-6 seems to have an important role in the intestinal inflammation that characterizes inflammatory bowel diseases (IBDs) such as Crohn disease and ulcerative colitis. However, little is known about the molecular mechanisms regulating IL-6 production in IBD. Here, we assessed the role of the transcriptional regulator IFN regulatory factor-4 (IRF4) in this process. Patients with either Crohn disease or ulcerative colitis exhibited increased IRF4 expression in lamina propria CD3+ T cells as compared with control patients. Consistent with IRF4 having a regulatory function in T cells, in a mouse model of IBD whereby colitis is induced in RAG-deficient mice by transplantation with CD4+CD45RB(hi) T cells, adoptive transfer of wild-type but not IRF4-deficient T cells resulted in severe colitis. Furthermore, IRF4-deficient mice were protected from T cell-dependent chronic intestinal inflammation in trinitrobenzene sulfonic acid- and oxazolone-induced colitis. In addition, IRF4-deficient mice with induced colitis had reduced mucosal IL-6 production, and IRF4 was required for IL-6 production by mucosal CD90+ T cells, which it protected from apoptosis. Finally, the protective effect of IRF4 deficiency could be abrogated by systemic administration of either recombinant IL-6 or a combination of soluble IL-6 receptor (sIL-6R) plus IL-6 (hyper-IL-6). Taken together, our data identify IRF4 as a key regulator of mucosal IL-6 production in T cell-dependent experimental colitis and suggest that IRF4 might provide a therapeutic target for IBDs.


Subject(s)
Colitis/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-6/metabolism , T-Lymphocytes/metabolism , Adoptive Transfer , Adult , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/metabolism , Colitis/chemically induced , Colitis/pathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interferon Regulatory Factors/genetics , Interleukin-6/genetics , Interleukin-6/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oxazolone/pharmacology , Receptors, Interleukin-6 , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/transplantation , Trinitrobenzenesulfonic Acid/pharmacology
12.
J Allergy Clin Immunol ; 121(4): 992-9.e6, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18329088

ABSTRACT

BACKGROUND: The transcriptional regulation of cytokines released and controlled by memory T cells is not well understood. Defective IFN-gamma production in allergic asthma correlates in human beings with the risk of wheezing in childhood. OBJECTIVE: To understand the role of the transcription factor nuclear factor of activated T cells 2 (NFATc2) in memory and effector T cells in the airways in experimental allergic asthma. METHODS: We used murine models of allergic asthma and adoptive cell transfer of fluorescence-activated sorted cells in a disease model. RESULTS: Mice lacking NFATc2 developed an increase in airwayhyperresponsiveness (AHR), remodeling, and serum IgE levelson ovalbumin sensitization. This phenotype was associated withCD81CD1222 T cells deficient in IFN-g production in theairways. The origin of this phenotype in NFATc2(2/2) mice wasrelated to an expanded population of lung CD81CD1221(IL-2Rb chain) CD127hi (IL-7 receptor [R] a chain1) long-livedmemory cells. Adoptive transfer of ovalbumin-specific CD81NFATc2(2/2) T cells enhanced the AHR generated byNFATc2(2/2) CD41 T cells in immunodeficient mice, increasedIL-17, and reduced IFN-g production in the reconstituted mice. Depletion of the memory CD81CD1221IL-7Rhigh T-cellpopulation corrected the defect in IFN-g production by lungNFATc2(2/2) CD81CD1222 cells and abrogated the increasedAHR observed in NFATc2(2/2) CD81 T-cell-reconstituted micewith a severe combined immunodeficiency disorder. CONCLUSION: Taken together, our results suggest that NFATc2 expression in long-lived memory CD8+ T cells controls IL-2 and IFN-gamma production in lung CD8+ T cells, which then limits TH17 and TH2 development in the airways during allergen challenge.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Hypersensitivity/immunology , Hypersensitivity/prevention & control , Immunologic Memory , NFATC Transcription Factors/physiology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/prevention & control , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Hypersensitivity/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-17/biosynthesis , Interleukin-2 Receptor beta Subunit/biosynthesis , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Receptors, Interleukin-7/biosynthesis , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Up-Regulation/immunology
13.
Eur J Cell Biol ; 86(11-12): 781-99, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17499880

ABSTRACT

Members of the transforming growth factor beta (TGF-beta) superfamily of signaling molecules are involved in the regulation of many developmental processes that involve the interaction between mesenchymal and epithelial tissues. Smad7 is a potent inhibitor of many members of the TGF-beta family, notably TGF-beta and activin. In this study, we show that embryonic overexpression of Smad7 in stratified epithelia using a keratin 5 promoter, results in severe morphogenetic defects in skin and teeth and leads to embryonic and perinatal lethality. To further analyze the functions of Smad7 in epithelial tissues of adult mice, we used an expression system that allowed a controlled overexpression of Smad7 in terms of both space and time. Skin defects in adult mice overexpressing Smad7 were characterized by hyper-proliferation and missing expression of early markers of keratinocyte differentiation. Upon Smad7-mediated blockade of TGF-beta superfamily signaling, ameloblasts failed to produce an enamel layer in incisor teeth. In addition, TGF-beta blockade in adult mice altered the pattern of thymic T cell differentiation and the number of thymic T cells was significantly reduced. This study shows that TGF-beta superfamily signaling is essential for development of hair, tooth and T-cells as well as differentiation and proliferation control in adult tissues.


Subject(s)
Hair/cytology , Hair/embryology , Morphogenesis , Signal Transduction , Tooth/cytology , Tooth/embryology , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Embryonic Development , Gene Expression Regulation , Genes, Reporter , Hair/abnormalities , Hair/pathology , Humans , Integrases/metabolism , Intermediate Filament Proteins/metabolism , Lac Operon , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin , Rabbits , Smad7 Protein/metabolism , Survival Analysis , Tooth/pathology , Tooth Abnormalities/pathology , Transgenes
14.
Eur J Immunol ; 37(6): 1663-77, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17506035

ABSTRACT

Epstein-Barr virus-induced gene (EBI)-3 codes for a soluble type 1 cytokine receptor homologous to the p40 subunit of IL-12 that is expressed by antigen-presenting cells following activation. Here, we analyzed the functional role of EBI-3 in a murine model of asthma associated with airway hyper-responsiveness (AHR) in ovalbumin-sensitized mice. Upon allergen challenge, EBI-3-/- mice showed less severe AHR, decreased numbers and degranulation of eosinophils and a significantly reduced number of VCAM-1+ cells in the lungs as compared to wild-type littermates. We thus analyzed lung CD11c+ cells before and after allergen challenge in these mice and found that before allergen challenge, lung CD11c+ cells isolated from EBI-3-/- mice express markers of a more plasmacytoid phenotype without releasing IFN-alpha as compared to those from wild-type littermates. Moreover, allergen challenge induced the development of myeloid CD11c+ cells in the lungs of EBI-3-/- mice, which released increased amounts of IL-10 and IL-12 while not expressing IFN-alpha. Finally, inhibition of EBI-3 expression in lung DC could prevent AHR in adoptive transfer studies by suppressing mediator release of effector cells into the airways. These results indicate a novel role for EBI-3 in controlling local immune responses in the lungs in experimental asthma.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , CD11c Antigen/analysis , Dendritic Cells/immunology , Lung/immunology , Receptors, Cytokine/genetics , Animals , Asthma/chemically induced , Asthma/metabolism , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cell Transplantation/methods , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Eosinophils/metabolism , Eosinophils/pathology , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-12/metabolism , Interleukin-4/metabolism , Interleukin-5/metabolism , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , STAT4 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
15.
Dig Dis Sci ; 52(9): 2396-402, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17415659

ABSTRACT

Hepatocyte apoptosis was recently described for NASH patients. The pathomechanisms are incompletely understood, but upregulation of the death receptor Fas was detectable on hepatocytes of NASH patients. We analyzed the sensitivity of fatty liver against CD95/Fas-mediated apoptotic cell death by injection of agonistic anti-Fas antibody (Jo2) in obese ob/ob mice and lean control animals. Ob/ob mice died within 12 hrs, whereas control animals survived. Liver enzymes were significantly increased compared to those in control mice (P < 0.001). Histological analysis and also TUNEL assay of liver sections from ob/ob mice exhibited massive liver injury. Activity of caspase 3 was significantly more enhanced in livers of ob/ob mice after Jo2 challenge. The increased sensitivity was confirmed in vitro by using ob/ob-derived primary hepatocytes. CD95 expression was similar in ob/ob and control mice. However, hepatocytes from ob/ob mice revealed a decreased mitochondrial membrane potential, suggesting that mitochondria play a potential role in this increased susceptibility.


Subject(s)
Apoptosis/immunology , Fatty Liver/pathology , Hepatocytes/pathology , fas Receptor/immunology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Fatty Liver/chemically induced , Fatty Liver/mortality , Hepatocytes/drug effects , Hepatocytes/metabolism , In Situ Nick-End Labeling , Leptin/deficiency , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Mice , Mice, Inbred C57BL , Severity of Illness Index , Survival Rate , fas Receptor/biosynthesis , fas Receptor/toxicity
17.
J Immunol ; 176(8): 5015-22, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16585598

ABSTRACT

Inhibitors of histone deacetylases (HDAC) are being studied for their antiproliferative effects in preclinical cancer trials. Recent studies suggest an anti-inflammatory role for this class of compounds. Because inflammatory bowel disease is associated with an increased risk of malignancies, agents with antiproliferative and anti-inflammatory properties would be of therapeutic interest. HDAC inhibitors from various classes were selected and evaluated for their in vitro capacity to suppress cytokine production and to induce apoptosis and histone acetylation. Valproic acid (VPA) and suberyolanilide hydroxamic acid (SAHA) were chosen for further studies in dextran sulfate sodium- and trinitrobenzene sulfonic acid-induced colitis in mice. In vitro, inhibition of HDAC resulted in a dose-dependent suppression of cytokine synthesis and apoptosis induction requiring higher concentrations of HDAC inhibitors for apoptosis induction compared with cytokine inhibition. Oral administration of either VPA or SAHA reduced disease severity in dextran sulfate sodium-induced colitis. The macroscopic and histologic reduction of disease severity was associated with a marked suppression of colonic proinflammatory cytokines. In parallel to the beneficial effect observed, a dose-dependent increase in histone 3 acetylation at the site of inflammation was shown under VPA treatment. Furthermore, SAHA as well as VPA treatment resulted in amelioration of trinitrobenzene sulfonic acid-induced colitis, which was associated with an increase of apoptosis of lamina propria lymphocytes. Inhibitors of HDAC reveal strong protective effects in different models of experimental colitis by inducing apoptosis and suppressing proinflammatory cytokines, thereby representing a promising class of compounds for clinical studies in human inflammatory bowel disease.


Subject(s)
Colitis/drug therapy , Colitis/metabolism , Histones/chemistry , Histones/metabolism , Acetylation , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Colitis/etiology , Colitis/pathology , Cytokines/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Valproic Acid/administration & dosage , Valproic Acid/pharmacology , Vorinostat
18.
Int Immunol ; 17(8): 993-1007, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16000330

ABSTRACT

Mice with a targeted deletion of the T-bet gene exhibit spontaneous airway hyperresponsiveness (AHR), airway inflammation, enhanced recovery of T(h)2 cytokines from bronchoalveolar lavage fluid, sub-epithelial collagen deposition and myofibroblast transformation. Here we analyze the mechanisms responsible for the chronic airway remodeling observed in these mice. CD4+ T cells isolated from the lung of T-bet-deficient mice were spontaneously activated CD44(high)CD69(high) memory T cells, with a typical T(h)2 cytokine profile. Neutralization of IL-13 but not IL-4 resulted in amelioration of AHR in airways of mice lacking T-bet. IL-13 blockade also led to reduced eosinophilia and decreased vimentin, transforming growth factor beta (TGF-beta) and alpha smooth muscle actin (alphaSMA) levels. T-bet(-/-) lung fibroblasts proliferated very rapidly and released increased amounts of TGF-beta. Interestingly, neutralization of TGF-beta ameliorated aspects of the chronic airway remodeling phenotype but did not reduce AHR. These data highlight a T-bet-directed function for IL-13 in controlling lung remodeling that is both dependent on and independent of its interaction with TGF-beta in the asthmatic airway.


Subject(s)
Asthma/etiology , Interleukin-13/metabolism , Transcription Factors/deficiency , Actins/metabolism , Animals , Asthma/genetics , Asthma/immunology , Asthma/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cells, Cultured , Cytokines/biosynthesis , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Fibroblasts/immunology , Fibroblasts/pathology , Immunologic Memory , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Lung/immunology , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Smad3 Protein , Smad7 Protein , T-Box Domain Proteins , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/biosynthesis , Vimentin/metabolism
19.
Oncogene ; 24(19): 3100-9, 2005 Apr 28.
Article in English | MEDLINE | ID: mdl-15735678

ABSTRACT

The human gene, human giant larvae (Hugl-1/Llg1/Lgl1) has significant homology to the Drosophila tumour suppressor gene lethal(2)giant larvae (lgl). The lgl gene codes for a cortical cytoskeleton protein, Lgl, that binds Myosin II and is involved in maintaining cell polarity and epithelial integrity. The human protein, Hugl-1 contains several conserved functional domains found in Lgl, suggesting that these proteins may have closely related functions. Whether loss of Hugl expression plays a role in human tumorigenesis has so far not been extensively investigated. Thus, we evaluated tumour tissues from 94 patients undergoing surgery for colorectal cancer (CRC) for loss of Hugl-1 transcription and compared our findings with the clinical data from each of these patients. We found that Hugl-1 was lost in 75% of tumour samples and these losses were associated with advanced stage and particularly with lymph node metastases. Reduced Hugl-1 expression during the adenoma-carcinoma sequence occurring as early as in colorectal adenomas was detected by both immunohistochemical and reverse transcription-polymerase chain reaction analysis. Functional assays with ecdysone-inducible cell lines revealed that Hugl-1 expression increased cell adhesion and decreased cell migration. Our studies thus indicate that downregulation of Hugl-1 contributes to CRC progression.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Proteins/metabolism , Adenoma/metabolism , Adult , Animals , Blotting, Western , Caco-2 Cells , Carcinoma/metabolism , Cell Adhesion , Cell Cycle , Cell Differentiation , Cell Line , Cell Line, Tumor , Cell Movement , Cytoskeletal Proteins , Cytoskeleton/metabolism , Disease Progression , Down-Regulation , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Male , Microscopy, Fluorescence , Middle Aged , Neoplasms/metabolism , Protein Structure, Tertiary , Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
20.
J Clin Invest ; 115(2): 313-25, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15668741

ABSTRACT

The cytokine IL-6 acts via a specific receptor complex that consists of the membrane-bound IL-6 receptor (mIL-6R) or the soluble IL-6 receptor (sIL-6R) and glycoprotein 130 (gp130). In this study, we investigated the role of IL-6R components in asthma. We observed increased levels of sIL-6R in the airways of patients with allergic asthma as compared to those in controls. In addition, local blockade of the sIL-6R in a murine model of late-phase asthma after OVA sensitization by gp130-fraction constant led to suppression of Th2 cells in the lung. By contrast, blockade of mIL-6R induced local expansion of Foxp3-positive CD4+CD25+ Tregs with increased immunosuppressive capacities. CD4+CD25+ but not CD4+CD25- lung T cells selectively expressed the IL-6R alpha chain and showed IL-6-dependent STAT-3 phosphorylation. Finally, in an in vivo transfer model of asthma in immunodeficient Rag1 mice, CD4+CD25+ T cells isolated from anti-IL-6R antibody-treated mice exhibited marked immunosuppressive and antiinflammatory functions. IL-6 signaling therefore controls the balance between effector cells and Tregs in the lung by means of different receptor components. Furthermore, inhibition of IL-6 signaling emerges as a novel molecular approach for the treatment of allergic asthma.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Lung/immunology , Receptors, Interleukin-2/immunology , Receptors, Interleukin-6/immunology , Th2 Cells/immunology , Adult , Animals , Antibodies/administration & dosage , Antibodies/immunology , Asthma/pathology , DNA-Binding Proteins/immunology , Female , Forkhead Transcription Factors , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Hypersensitivity/pathology , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Male , Mice , Mice, Knockout , Ovalbumin/metabolism , Receptors, Cytokine/immunology , STAT3 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Th2 Cells/pathology , Trans-Activators/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...