Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Crohns Colitis ; 14(4): 508-524, 2020 May 21.
Article in English | MEDLINE | ID: mdl-31630153

ABSTRACT

BACKGROUND AND AIMS: The topically applied Toll-like receptor 9 [TLR9] agonist cobitolimod is a first-in-class DNA-based oligonucleotide with demonstrated therapeutic efficacy in clinical trials with ulcerative colitis [UC] patients. We here characterized its anti-inflammatory mechanism in UC. METHODS: Luminal cobitolimod administration was evaluated in an experimental dextran sodium sulfate [DSS]-induced colitis model. Cultured blood and mucosal cells from UC patients were treated with cobitolimod and analysed via microarray, quantitative real-time PCR, ELISA and flow cytometry. Intestinal slides of cobitolimod-treated UC patients were analysed by immunohistochemistry. RESULTS: Cobitolimod administration markedly suppressed experimental colitis activity, and microarray analyses demonstrated mucosal IL10 upregulation and suppression of IL17 signalling pathways. Cobitolimod treatment was associated with significant induction of mucosal IL10+Tr1 and Treg cells and suppression of Th17 cells. TLR9 knockout mice indicated that cobitolimod requires TLR9 signalling for IL10 induction. In UC patients, mucosal TLR9 levels correlated with severity of inflammation. Cobitolimod inhibited IL17A and IL17F, but increased IL10 and FoxP3 expression in cultured intestinal UC T cells. Cobitolimod-mediated suppression of intestinal IL17+T cells was abrogated by IL10 blockade. Furthermore, cobitolimod led to heightened IL10 production by wound healing macrophages. Immunohistochemistry in intestinal biopsies of cobitolimod-treated UC patients indicated increased presence of IL10+mononuclear and regulatory T cells, as well as reduction of IL17+cells. CONCLUSION: Activation of TLR9 via cobitolimod might represent a novel therapeutic approach in UC, as it suppresses Th17 cells and induces anti-inflammatory IL10+macrophages and regulatory T cells, thereby modifying the dysregulated intestinal cytokine balance. PODCAST: This article has an associated podcast which can be accessed at https://academic.oup.com/ecco-jcc/pages/podcast.


Subject(s)
Colitis, Ulcerative , Intestinal Mucosa , Macrophages , Oligodeoxyribonucleotides , T-Lymphocytes, Regulatory , Th17 Cells , Toll-Like Receptor 9/agonists , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Cell Culture Techniques , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Disease Models, Animal , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/pharmacokinetics , Gene Expression Regulation , Humans , Immunomodulation , Interleukin-10/analysis , Interleukin-17/analysis , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Middle Aged , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacokinetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Tissue Array Analysis/methods
2.
Gut ; 68(5): 814-828, 2019 05.
Article in English | MEDLINE | ID: mdl-29848778

ABSTRACT

OBJECTIVE: Anti-tumour necrosis factor (TNF) antibodies are successfully used for treatment of Crohn's disease. Nevertheless, approximately 40% of patients display failure to anti-TNF therapy. Here, we characterised molecular mechanisms that are associated with endoscopic resistance to anti-TNF therapy. DESIGN: Mucosal and blood cells were isolated from patients with Crohn's disease prior and during anti-TNF therapy. Cytokine profiles, cell surface markers, signalling proteins and cell apoptosis were assessed by microarray, immunohistochemistry, qPCR, ELISA, whole organ cultures and FACS. RESULTS: Responders to anti-TNF therapy displayed a significantly higher expression of TNF receptor 2 (TNFR2) but not IL23R on T cells than non-responders prior to anti-TNF therapy. During anti-TNF therapy, there was a significant upregulation of mucosal IL-23p19, IL23R and IL-17A in anti-TNF non-responders but not in responders. Apoptosis-resistant TNFR2+IL23R+ T cells were significantly expanded in anti-TNF non-responders compared with responders, expressed the gut tropic integrins α4ß7, and exhibited increased expression of IFN-γ, T-bet, IL-17A and RORγt compared with TNFR2+IL23R- cells, indicating a mixed Th1/Th17-like phenotype. Intestinal TNFR2+IL23R+ T cells were activated by IL-23 derived from CD14+ macrophages, which were significantly more present in non-responders prior to anti-TNF treatment. Administration of IL-23 to anti-TNF-treated mucosal organ cultures led to the expansion of CD4+IL23R+TNFR2+ lymphocytes. Functional studies demonstrated that anti-TNF-induced apoptosis in mucosal T cells is abrogated by IL-23. CONCLUSIONS: Expansion of apoptosis-resistant intestinal TNFR2+IL23R+ T cells is associated with resistance to anti-TNF therapy in Crohn's disease. These findings identify IL-23 as a suitable molecular target in patients with Crohn's disease refractory to anti-TNF therapy.


Subject(s)
Crohn Disease/metabolism , Drug Resistance , Gastrointestinal Agents/therapeutic use , Receptors, Interleukin/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , T-Lymphocytes/physiology , Adalimumab/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Crohn Disease/drug therapy , Crohn Disease/pathology , Humans , Infliximab/therapeutic use , Interleukin-17/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , Young Adult
3.
Front Immunol ; 9: 1700, 2018.
Article in English | MEDLINE | ID: mdl-30131801

ABSTRACT

Background and aims: Despite proven clinical efficacy of vedolizumab (VDZ) for inducing and maintaining remission in patients with Crohn's disease (CD) and ulcerative colitis (UC), subgroups of patients have no therapeutic benefit from anti-α4ß7 integrin therapy with VDZ. Within this study, we aimed to identify genetic, cellular, and immunological mechanisms that define response and failure to VDZ treatment. Methods: Intestinal RNA sequencing was performed in UC and CD patients before and at week 14 of VDZ therapy. α4ß7 expression on peripheral and mucosal immune cells was assessed by flow cytometry and immunohistochemistry. Cellular modes of VDZ-mediated action were analyzed ex vivo and in VDZ-treated inflammatory bowel disease patients. Results: Transcriptome analysis showed an impairment of signaling cascades associated with adhesion, diapedesis, and migration of granulocytes and agranulocytes upon VDZ therapy. In non-remitters to VDZ therapy, a tissue destructive and leukocyte-mediated inflammatory activity with activation of TNF-dependent pathways was present, all of which were inhibited in remitters to VDZ. Clinical remission was associated with a significant reduction of α4ß7 expression on Th2 and Th17 polarized mucosal CD4+ T cells at week 14 of VDZ therapy and with significantly higher numbers of α4ß7-expressing mucosal cells prior to the initiation of VDZ therapy compared with non-remitters. Conclusion: Intestinal α4ß7 expression prior to VDZ therapy might represent a biomarker that predicts therapeutic response to subsequent VDZ treatment. Due to high activation of TNF signaling in VDZ non-remitters, anti-TNF treatment might represent a promising therapeutic strategy in VDZ refractory patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cytokines/metabolism , Inflammatory Bowel Diseases/drug therapy , Integrins/antagonists & inhibitors , Signal Transduction/drug effects , Adult , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Gastrointestinal Agents/therapeutic use , HeLa Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Integrins/genetics , Integrins/metabolism , Male , Middle Aged , Remission Induction , Signal Transduction/genetics , Signal Transduction/immunology , Transcriptome/drug effects , Transcriptome/genetics , Transcriptome/immunology , Young Adult
4.
World J Gastroenterol ; 22(42): 9300-9313, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27895418

ABSTRACT

Anti-tumor necrosis factor (TNF) antibodies are successfully used in the therapy of inflammatory bowel diseases (IBD). However, the molecular mechanism of action of these agents is still a matter of debate. Apart from neutralization of TNF, influence on the intestinal barrier function, induction of apoptosis in mucosal immune cells, formation of regulatory macrophages as well as other immune modulating properties have been discussed as central features. Nevertheless, clinically effective anti-TNF antibodies were shown to differ in their mode-of-action in vivo and in vitro. Furthermore, the anti-TNF agent etanercept is effective in the treatment of rheumatoid arthritis but failed to induce clinical response in Crohn's disease patients, suggesting different contributions of TNF in the pathogenesis of these inflammatory diseases. In the following, we will review different aspects regarding the mechanism of action of anti-TNF agents in general and analyze comparatively different effects of each anti-TNF agent such as TNF neutralization, modulation of the immune system, reverse signaling and induction of apoptosis. We discuss the relevance of the membrane-bound form of TNF compared to the soluble form for the immunopathogenesis of IBD. Furthermore, we review reports that could lead to personalized medicine approaches regarding treatment with anti-TNF antibodies in chronic intestinal inflammation, by predicting response to therapy.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies/therapeutic use , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Gastrointestinal Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Intestines/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antibody-Dependent Cell Cytotoxicity/drug effects , Apoptosis/drug effects , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Crohn Disease/diagnosis , Crohn Disease/immunology , Crohn Disease/metabolism , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/pathology , Signal Transduction/drug effects , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
5.
J Crohns Colitis ; 10(10): 1132-43, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27112707

ABSTRACT

BACKGROUND AND AIMS: The clinical use of azathioprine and 6-mercaptopurine is limited by their delayed onset of action and potential side effects such as myelosuppression and hepatotoxicity. As these drugs specifically target the Vav1/Rac1 signalling pathway in T lamina propria lymphocytes via their metabolite 6-thio-GTP, we studied expression and optimised suppression of this pathway in inflammatory bowel diseases [IBD]. METHODS: Rac1 and Vav1 expressions were analysed in mucosal immune cells in IBD patients. Targeted molecular modelling of the 6-thio-GTP molecule was performed to optimise Rac1 blockade; 44 modified designer thiopurine-analogues were tested for apoptosis induction, potential toxicity, and immunosuppression. Activation of the Vav1/Rac1 pathway in lymphocytes was studied in IBD patients and in lamina propria immune cells in the presence or absence of thiopurine-analogues. RESULTS: Several thiopurine-analogues induced significantly higher T cell apoptosis than 6-mercaptopurine. We identified a compound, denoted B-0N, based on its capacity to mediate earlier and stronger induction of T cell apoptosis than 6-mercaptopurine. B-0N-treatment resulted in accelerated inhibition of Rac1 activity in primary peripheral blood T cells as well as in intestinal lamina propria immune cells. Compared with 6-thio-GTP and 6-mercaptopurine, B-0N-treatment was associated with decreased myelo- and hepatotoxicity. CONCLUSIONS: The Vav1/Rac1 pathway is activated in mucosal immune cells in IBD. The designer thiopurine-analogue B-0N induces immunosuppression more potently than 6-mercaptopurine.


Subject(s)
Designer Drugs/pharmacology , Immunosuppressive Agents/pharmacology , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Mercaptopurine/analogs & derivatives , Mercaptopurine/pharmacology , Apoptosis/drug effects , Biomarkers/metabolism , Case-Control Studies , Designer Drugs/therapeutic use , Drug Design , Humans , Immunosuppressive Agents/therapeutic use , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mercaptopurine/therapeutic use , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction/drug effects , T-Lymphocytes/metabolism , rac1 GTP-Binding Protein/metabolism
6.
Nat Commun ; 7: 10973, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964500

ABSTRACT

Ductal occlusion has been postulated to precipitate focal pancreatic inflammation, while the nature of the primary occluding agents has remained elusive. Neutrophils make use of histone citrullination by peptidyl arginine deiminase-4 (PADI4) in contact to particulate agents to extrude decondensed chromatin as neutrophil extracellular traps (NETs). In high cellular density, NETs form macroscopically visible aggregates. Here we show that such aggregates form inside pancreatic ducts in humans and mice occluding pancreatic ducts and thereby driving pancreatic inflammation. Experimental models indicate that PADI4 is critical for intraductal aggregate formation and that PADI4-deficiency abrogates disease progression. Mechanistically, we identify the pancreatic juice as a strong instigator of neutrophil chromatin extrusion. Characteristic single components of pancreatic juice, such as bicarbonate ions and calcium carbonate crystals, induce aggregated NET formation. Ductal occlusion by aggregated NETs emerges as a pathomechanism with relevance in a plethora of inflammatory conditions involving secretory ducts.


Subject(s)
Chromatin/immunology , Cytokines/immunology , Extracellular Traps/immunology , Interleukin-17/pharmacology , Neutrophils/immunology , Pancreas/drug effects , Pancreatic Ducts/immunology , Pancreatitis/immunology , Animals , Ceruletide/toxicity , Disease Models, Animal , Flow Cytometry , Humans , Hydrolases/metabolism , Immunohistochemistry , Mice , Pancreas/immunology , Pancreas/pathology , Pancreatic Ducts/pathology , Pancreatic Juice/enzymology , Pancreatic Juice/immunology , Pancreatitis/chemically induced , Pancreatitis/pathology , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Reverse Transcriptase Polymerase Chain Reaction
7.
J Clin Invest ; 126(2): 611-26, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26752649

ABSTRACT

Although defects in intestinal barrier function are a key pathogenic factor in patients with inflammatory bowel diseases (IBDs), the molecular pathways driving disease-specific alterations of intestinal epithelial cells (IECs) are largely unknown. Here, we addressed this issue by characterizing the transcriptome of IECs from IBD patients using a genome-wide approach. We observed disease-specific alterations in IECs with markedly impaired Rho-A signaling in active IBD patients. Localization of epithelial Rho-A was shifted to the cytosol in IBDs, and inflammation was associated with suppressed Rho-A activation due to reduced expression of the Rho-A prenylation enzyme geranylgeranyltransferase-I (GGTase-I). Functionally, we found that mice with conditional loss of Rhoa or the gene encoding GGTase-I, Pggt1b, in IECs exhibit spontaneous chronic intestinal inflammation with accumulation of granulocytes and CD4+ T cells. This phenotype was associated with cytoskeleton rearrangement and aberrant cell shedding, ultimately leading to loss of epithelial integrity and subsequent inflammation. These findings uncover deficient prenylation of Rho-A as a key player in the pathogenesis of IBDs. As therapeutic triggering of Rho-A signaling suppressed intestinal inflammation in mice with GGTase-I-deficient IECs, our findings suggest new avenues for treatment of epithelial injury and mucosal inflammation in IBD patients.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Prenylation , Signal Transduction , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/pathology , Mice , Mice, Mutant Strains , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/genetics
8.
World J Gastroenterol ; 21(45): 12963-9, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26668517

ABSTRACT

We present the case of a 53-year-old woman with long-standing ulcerative colitis and severe, steroid-dependent disease course unresponsive to treatment with azathioprine, methotrexate, anti-TNF antibodies (infliximab, adalimumab) and tacrolimus, who refused colectomy as a therapeutic option. As the pro-inflammatory cytokine interleukin-6 (IL-6) had been identified as a crucial regulator in the immunopathogenesis of inflammatory bowel diseases, we treated the patient with biweekly intravenous infusions of an anti-IL-6R antibody (tocilizumab) for 12 wk. However, no clinical improvement of disease activity was noted. In fact, endoscopic, histological and endomicroscopic assessment demonstrated exacerbation of mucosal inflammation and ulcer formation upon anti-IL-6R therapy. Mechanistic studies revealed that tocilizumab treatment failed to suppress intestinal IL-6 production, impaired epithelial barrier function and induced production of pro-inflammatory cytokines such as TNF, IL-21 and IFN-γ. Inhibition of IL-6 by tocilizumab had no clinical benefit in this patient with intractable ulcerative colitis and even led to exacerbation of mucosal inflammation. Our findings suggest that anti-IL-6R antibody therapy may lead to aggravation of anti-TNF resistant ulcerative colitis. When targeting IL-6, the differential responsiveness of target cells has to be taken into account, as IL-6 on the one side promotes acute and chronic mucosal inflammation via soluble IL-6R signaling but on the other side also strongly contributes to epithelial cell survival via membrane bound IL-6R signaling.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Colitis, Ulcerative/drug therapy , Gastrointestinal Agents/adverse effects , Receptors, Interleukin-6/antagonists & inhibitors , Anti-Inflammatory Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Biopsy , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colonoscopy , Cytokines/genetics , Cytokines/immunology , Disease Progression , Female , Gastrointestinal Agents/administration & dosage , Humans , Middle Aged , Molecular Targeted Therapy , Receptors, Interleukin-6/immunology , Treatment Outcome
9.
J Immunol ; 195(4): 1753-62, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26170386

ABSTRACT

Plasmacytoid dendritic cells (pDCs) efficiently produce large amounts of type I IFN in response to TLR7 and TLR9 ligands, whereas conventional DCs (cDCs) predominantly secrete high levels of the cytokines IL-10 and IL-12. The molecular basis underlying this distinct phenotype is not well understood. In this study, we identified the MAPK phosphatase Dusp9/MKP-4 by transcriptome analysis as selectively expressed in pDCs, but not cDCs. We confirmed the constitutive expression of Dusp9 at the protein level in pDCs generated in vitro by culture with Flt3 ligand and ex vivo in sorted splenic pDCs. Dusp9 expression was low in B220(-) bone marrow precursors and was upregulated during pDC differentiation, concomitant with established pDC markers. Higher expression of Dusp9 in pDCs correlated with impaired phosphorylation of the MAPK ERK1/2 upon TLR9 stimulation. Notably, Dusp9 was not expressed at detectable levels in human pDCs, although these displayed similarly impaired activation of ERK1/2 MAPK compared with cDCs. Enforced retroviral expression of Dusp9 in mouse GM-CSF-induced cDCs increased the expression of TLR9-induced IL-12p40 and IFN-ß, but not of IL-10. Conditional deletion of Dusp9 in pDCs was effectively achieved in Dusp9(flox/flox); CD11c-Cre mice at the mRNA and protein levels. However, the lack of Dusp9 in pDC did not restore ERK1/2 activation after TLR9 stimulation and only weakly affected IFN-ß and IL-12p40 production. Taken together, our results suggest that expression of Dusp9 is sufficient to impair ERK1/2 activation and enhance IFN-ß expression. However, despite selective expression in pDCs, Dusp9 is not essential for high-level IFN-ß production by these cells.


Subject(s)
Dendritic Cells/metabolism , Dual-Specificity Phosphatases/genetics , Gene Expression , Interferon-beta/biosynthesis , Animals , Cell Differentiation/genetics , Cluster Analysis , Computational Biology/methods , Dendritic Cells/cytology , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Humans , Interferon-beta/genetics , Interleukin-12 , Mice , Mice, Knockout , Organ Specificity/genetics , Phosphorylation , Reproducibility of Results , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Transcriptome
10.
PLoS One ; 10(3): e0118401, 2015.
Article in English | MEDLINE | ID: mdl-25799189

ABSTRACT

Gastrointestinal infections with EHEC and EPEC are responsible for outbreaks of diarrheal diseases and represent a global health problem. Innate first-line-defense mechanisms such as production of mucus and antimicrobial peptides by intestinal epithelial cells are of utmost importance for host control of gastrointestinal infections. For the first time, we directly demonstrate a critical role for Stat3 activation in intestinal epithelial cells upon infection of mice with Citrobacter rodentium - a murine pathogen that mimics human infections with attaching and effacing Escherichia coli. C. rodentium induced transcription of IL-6 and IL-22 in gut samples of mice and was associated with activation of the transcription factor Stat3 in intestinal epithelial cells. C. rodentium infection induced expression of several antimicrobial peptides such as RegIIIγ and Pla2g2a in the intestine which was critically dependent on Stat3 activation. Consequently, mice with specific deletion of Stat3 in intestinal epithelial cells showed increased susceptibility to C. rodentium infection as indicated by high bacterial load, severe gut inflammation, pronounced intestinal epithelial cell death and dissemination of bacteria to distant organs. Together, our data implicate an essential role for Stat3 activation in intestinal epithelial cells during C. rodentium infection. Stat3 concerts the host response to bacterial infection by controlling bacterial growth and suppression of apoptosis to maintain intestinal epithelial barrier function.


Subject(s)
Citrobacter rodentium/immunology , Colitis/prevention & control , Colon/immunology , Enterobacteriaceae Infections/prevention & control , Epithelial Cells/immunology , Intestines/immunology , STAT3 Transcription Factor/physiology , Animals , Colitis/etiology , Colon/microbiology , Colon/pathology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Humans , Intestinal Mucosa/metabolism , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Arch Toxicol ; 89(4): 541-54, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25632846

ABSTRACT

Since its discovery in 1986, originally as B cell stimulating factor 2, the knowledge on IL-6 for immune homeostasis and its pathophysiological implications has rapidly increased. It is now clear that IL-6, alone or in combination with other cytokines, is an architect for shaping and generating immune responses which exerts profound activities on the induction of acute-phase reactions, the differentiation of B lymphocytes, the modulation of T cell apoptosis, the activation of T helper cells and the balance between regulatory T cells and Th17 cells. In parallel to the identification of these physiologic functions, IL-6 has emerged as a critical mediator for perpetuating chronic inflammation and autoimmunity and is increasingly recognized as a key cytokine for linking chronic inflammation to cancer development. In this review, we begin by briefly summarizing the molecular events of IL-6 regulation and signaling and then describe the role of IL-6 in orchestrating innate and adaptive immune responses and its immunopathological relevance for chronic inflammatory diseases. We further outline how IL-6 links chronic inflammation and cancer development and finally provide an outlook on novel therapeutic strategies targeting IL-6 signaling for the treatment of chronic inflammatory diseases and cancer.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Inflammatory Bowel Diseases/immunology , Interleukin-6/metabolism , Molecular Targeted Therapy , Neoplasms/immunology , Adaptive Immunity/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , Humans , Immunity, Innate/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Interleukin-6/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology , Signal Transduction
12.
J Immunol ; 193(7): 3779-91, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187663

ABSTRACT

The Citrobacter rodentium model mimics the pathogenesis of infectious colitis and requires sequential contributions from different immune cell populations, including innate lymphoid cells (ILCs) and CD4(+) lymphocytes. In this study, we addressed the role of STAT3 activation in CD4(+) cells during host defense in mice against C. rodentium. In mice with defective STAT3 in CD4(+) cells (Stat3(ΔCD4)), the course of infection was unchanged during the innate lymphoid cell-dependent early phase, but significantly altered during the lymphocyte-dependent later phase. Stat3(ΔCD4) mice exhibited intestinal epithelial barrier defects, including downregulation of antimicrobial peptides, increased systemic distribution of bacteria, and prolonged reduction in the overall burden of C. rodentium infection. Immunomonitoring of lamina propria cells revealed loss of virtually all IL-22-producing CD4(+) lymphocytes, suggesting that STAT3 activation was required for IL-22 production not only in Th17 cells, but also in Th22 cells. Notably, the defective host defense against C. rodentium in Stat3(∆CD4) mice could be fully restored by specific overexpression of IL-22 through a minicircle vector-based technology. Moreover, expression of a constitutive active STAT3 in CD4(+) cells shaped strong intestinal epithelial barrier function in vitro and in vivo through IL-22, and it promoted protection from enteropathogenic bacteria. Thus, our work indicates a critical role of STAT3 activation in Th17 and Th22 cells for control of the IL-22-mediated host defense, and strategies expanding STAT3-activated CD4(+) lymphocytes may be considered as future therapeutic options for improving intestinal barrier function in infectious colitis.


Subject(s)
Citrobacter rodentium/immunology , Colitis/immunology , Enterobacteriaceae Infections/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , STAT3 Transcription Factor/immunology , Th17 Cells/immunology , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/pathology , Interleukins/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Mice, Transgenic , Th17 Cells/pathology , Interleukin-22
13.
Nat Med ; 20(3): 313-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24562382

ABSTRACT

As antibodies to tumor necrosis factor (TNF) suppress immune responses in Crohn's disease by binding to membrane-bound TNF (mTNF), we created a fluorescent antibody for molecular mTNF imaging in this disease. Topical antibody administration in 25 patients with Crohn's disease led to detection of intestinal mTNF(+) immune cells during confocal laser endomicroscopy. Patients with high numbers of mTNF(+) cells showed significantly higher short-term response rates (92%) at week 12 upon subsequent anti-TNF therapy as compared to patients with low amounts of mTNF(+) cells (15%). This clinical response in the former patients was sustained over a follow-up period of 1 year and was associated with mucosal healing observed in follow-up endoscopy. These data indicate that molecular imaging with fluorescent antibodies has the potential to predict therapeutic responses to biological treatment and can be used for personalized medicine in Crohn's disease and autoimmune or inflammatory disorders.


Subject(s)
Antibodies, Monoclonal/chemistry , Crohn Disease/metabolism , Crohn Disease/therapy , Fluorescent Dyes/chemistry , Immunotherapy/methods , Adalimumab , Adult , Antibodies, Monoclonal, Humanized/chemistry , Endoscopy, Gastrointestinal , Female , Fluorescent Antibody Technique , Humans , Immunoglobulin G/chemistry , Inflammation , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestines/pathology , Male , Microscopy, Confocal , Middle Aged , Prospective Studies , Treatment Outcome , Tumor Necrosis Factor-alpha/chemistry
14.
Arthritis Rheum ; 65(9): 2310-22, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23784913

ABSTRACT

OBJECTIVE: Wnt signaling plays a pivotal role in skeletal development and in the control of cartilage and bone turnover. We have recently shown that the secreted Wnt antagonist Wnt inhibitory factor 1 (WIF-1) is mainly expressed in the upper layers of epiphyseal and articular cartilage and, to a lesser extent, in bone. Nevertheless, WIF-1(-/-) mice develop normally. In light of these findings, we undertook this study to analyze the role of WIF-1 in arthritis. METHODS: Expression analyses for WIF-1 were performed by real-time reverse transcription-polymerase chain reaction (RT-PCR). WIF-1(-/-) and tumor necrosis factor (TNF)-transgenic mice were crossbred, and the progression of arthritis in TNF-transgenic WIF-1(-/-) mice and littermate controls was evaluated. Structural joint damage was analyzed by histologic staining, histomorphometry, and micro-computed tomography. Wnt/ß-catenin signaling was investigated by real-time RT-PCR and immunofluorescence on primary chondrocytes. RESULTS: WIF-1 expression was repressed by TNFα in chondrocytes and osteoblasts and down-regulated in experimental arthritis and in articular cartilage from patients with rheumatoid arthritis. WIF-1 deficiency partially protected TNF-transgenic mice against bone erosion and loss of trabecular bone, probably as a result of less osteoclast activity. In contrast, arthritis-related cartilage damage was aggravated by WIF-1 deficiency, while overexpression of WIF-1 attenuated cartilage degradation in TNF-transgenic mice. In chondrocytes, TNFα stimulated canonical Wnt signaling, which could be blocked by WIF-1, indicating a direct effect of TNFα and WIF-1 on Wnt signaling in this system. CONCLUSION: These data suggest that WIF-1 may take part in the fine-tuning of cartilage and bone turnover, promoting the balance of cartilage versus bone anabolism.


Subject(s)
Arthritis, Experimental/metabolism , Bone and Bones/metabolism , Cartilage/metabolism , Extracellular Matrix Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adaptor Proteins, Signal Transducing , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone and Bones/pathology , Cartilage/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Progression , Down-Regulation , Extracellular Matrix Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Tumor Necrosis Factor-alpha/genetics
15.
Nat Med ; 19(3): 358-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23396210

ABSTRACT

Hepatic insulin resistance is a driving force in the pathogenesis of type 2 diabetes mellitus (T2DM) and is tightly coupled with excessive storage of fat and the ensuing inflammation within the liver. There is compelling evidence that activation of the transcription factor nuclear factor-κB (NF-κB) and downstream inflammatory signaling pathways systemically and in the liver are key events in the etiology of hepatic insulin resistance and ß-cell dysfunction, although the molecular mechanisms involved are incompletely understood. We here test the hypothesis that receptor activator of NF-κB ligand (RANKL), a prototypic activator of NF-κB, contributes to this process using both an epidemiological and experimental approach. In the prospective population-based Bruneck Study, a high serum concentration of soluble RANKL emerged as a significant (P<0.001) and independent risk predictor of T2DM manifestation. In close agreement, systemic or hepatic blockage of RANKL signaling in genetic and nutritional mouse models of T2DM resulted in a marked improvement of hepatic insulin sensitivity and amelioration or even normalization of plasma glucose concentrations and glucose tolerance. Overall, this study provides evidence for a role of RANKL signaling in the pathogenesis of T2DM. If so, translation to the clinic may be feasible given current pharmacological strategies to lower RANKL activity to treat osteoporosis.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Insulin Resistance/physiology , Liver/metabolism , RANK Ligand/metabolism , Adult , Aged , Animals , Cell Line , Enzyme Activation , Female , HEK293 Cells , Humans , Male , Mice , Middle Aged , NF-kappa B/metabolism , Prospective Studies , RANK Ligand/antagonists & inhibitors
16.
Gastroenterology ; 141(5): 1875-86, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21820391

ABSTRACT

BACKGROUND & AIMS: The soluble hematopoietin receptor Epstein-Barr virus-induced protein (EBI)-3 is an immune regulator that has been associated with the pathogenesis of inflammatory bowel disease. However, the concept that EBI3 is part of an interleukin (IL)-27 heterodimer that mediates chronic inflammatory and autoimmune diseases has been challenged by the description of IL-35, a bioactive cytokine comprising EBI3 and IL-12 p35. We investigated the roles of IL-27 and IL-35 in chronic inflammation of the intestine. METHODS: We analyzed EBI3-deficient mice and IL-27p28-deficient mice with spontaneous or T-cell transfer-induced colitis and compared outcomes with wild-type mice (controls). We constructed vectors that express EBI3 covalently linked to the IL-12p35 chain (recombinant [r]IL-35). RESULTS: Intestines of EBI3-deficient mice had increased pathologic features of colitis, compared with IL-27p28-deficient or control mice; they also had shorter survival times, indicating that IL-35, rather than IL-27, protects the intestine from immune responses in mice. The mucosa of EBI3-deficient mice accumulated subsets of activated CD4+ T cells that produced T-helper (Th)1 and Th17 cytokines. Adoptive transfer of these T cells induced colitis in RAG-deficient mice. The rIL-35 significantly reduced the development of several forms of experimental colitis and reduced levels of markers of Th1 and Th17 cells. CONCLUSIONS: IL-35 controls the development of T-cell-dependent colitis in mice. It might be developed as a therapeutic target for patients with chronic intestinal inflammation.


Subject(s)
Colitis/immunology , Colitis/metabolism , Immunity, Mucosal/physiology , Interleukins/metabolism , Intestinal Mucosa/metabolism , T-Lymphocytes/physiology , Animals , Colitis/pathology , Disease Models, Animal , Female , Immunity, Mucosal/drug effects , Interleukin-12 Subunit p35/deficiency , Interleukin-12 Subunit p35/genetics , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens , Phenotype , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , T-Lymphocytes/pathology , Th1 Cells/pathology , Th1 Cells/physiology , Th17 Cells/pathology , Th17 Cells/physiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL