Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
Oncogene ; 38(11): 1787-1801, 2019 03.
Article in English | MEDLINE | ID: mdl-30353167

ABSTRACT

While great advances have been achieved regarding the genetic basis of colorectal cancer, the complex role of cell-cell communication and cytokine-induced signaling during its pathogenesis remains less understood. Signal transducer and activator of transcription 6 (Stat6) is the main transcription factor of interleukin-4 (IL-4) signaling and its participation in the development of various tumor types has been already reported. Here we aimed to examine the contribution of Stat6 in intestinal epithelial cells (IEC) in mouse models of intestinal carcinogenesis. Wild-type (WT), Stat6 knockout (Stat6-/-), and intestinal epithelial cell-specific IL-4Rα knockout (Il-4rαΔIEC) mice were subjected to colitis-associated (AOM/DSS) and colitis-independent (sporadic) carcinogenesis. IEC proliferation, apoptosis and RNA expression were evaluated by immunohistochemical, immunoblot, and RT-PCR analysis. We found that Stat6-/- mice developed more tumors in the colitis-associated carcinogenesis model. This was accompanied by a more pronounced inflammatory response during colitis and an elevated Stat3-dependent proliferation of IEC. Increased sensitivity to DSS-induced colitis was caused by elevated cell death in response to the initial carcinogen exposure as Stat6 deficiency led to increased chromatin compaction affecting DNA damage response in IEC upon treatment with alkylating agents independently of IL-4Rα engagement. Thus, loss of Stat6 caused more severe colitis and increased tumor load, however loss-of-initiated Stat6-/- IEC prevented tumor formation in the absence of overt inflammation. Our data unravel unexpected IL-4-independent functions of Stat6 in chromatin compaction in intestinal epithelial cells ultimately providing both tumor suppressive as well as tumor promoting effects in different models of intestinal tumorigenesis.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/metabolism , Colitis/complications , Colonic Neoplasms , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , STAT6 Transcription Factor/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , DNA Packaging/genetics , Disease Models, Animal , Epithelial Cells/pathology , Female , Gene Deletion , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
Cell ; 174(1): 88-101.e16, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29909986

ABSTRACT

In colorectal cancer patients, a high density of cytotoxic CD8+ T cells in tumors is associated with better prognosis. Using a Stat3 loss-of-function approach in two wnt/ß-catenin-dependent autochthonous models of sporadic intestinal tumorigenesis, we unravel a complex intracellular process in intestinal epithelial cells (IECs) that controls the induction of a CD8+ T cell based adaptive immune response. Elevated mitophagy in IECs causes iron(II)-accumulation in epithelial lysosomes, in turn, triggering lysosomal membrane permeabilization. Subsequent release of proteases into the cytoplasm augments MHC class I presentation and activation of CD8+ T cells via cross-dressing of dendritic cells. Thus, our findings highlight a so-far-unrecognized link between mitochondrial function, lysosomal integrity, and MHC class I presentation in IECs and suggest that therapies triggering mitophagy or inducing LMP in IECs may prove successful in shifting the balance toward anti-tumor immunity in colorectal cancer.


Subject(s)
Adaptive Immunity , Mitophagy , Adaptive Immunity/drug effects , Animals , Azoxymethane/toxicity , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Membrane Permeability , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Ferrous Compounds/metabolism , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Mitophagy/drug effects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Survival Rate
4.
FEBS J ; 285(12): 2161-2181, 2018 06.
Article in English | MEDLINE | ID: mdl-29582565

ABSTRACT

Nuclear factor κB (NF-κB) transcription factors are the central hubs of signaling pathways connecting proinflammatory signals to cell survival, proliferation and cytokine production. In cancers, NF-κB signaling influences many aspects of tumor development, from initiation to metastasis. These functions are mediated by tumor-induced plasticity that allows tumor cells to adapt and survive in changing conditions within the tumor microenvironment. Tumor cell plasticity is shaped by the inflammatory microenvironment in tumors. This review focuses on inhibitor of NF-κB kinases, the direct upstream elements of NF-κB regulation, specifically on their conventional and non-conventional functions in animal models of tumorigenesis from the recent literature.


Subject(s)
Gene Expression Regulation, Neoplastic , I-kappa B Kinase/genetics , NF-kappa B/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Tumor Microenvironment/genetics , Animals , Carcinogenesis , Epithelial-Mesenchymal Transition/genetics , Gene Regulatory Networks , Humans , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , Protein Subunits/genetics , Protein Subunits/metabolism , Signal Transduction
5.
Gastroenterology ; 153(1): 178-190.e10, 2017 07.
Article in English | MEDLINE | ID: mdl-28400195

ABSTRACT

BACKGROUND & AIMS: Inhibitors of the epidermal growth factor receptor (EGFR) are the first-line therapy for patients with metastatic colorectal tumors without RAS mutations. However, EGFR inhibitors are ineffective in these patients, and tumor level of EGFR does not associate with response to therapy. We screened human colorectal tumors for EGFR-positive myeloid cells and investigated their association with patient outcome. We also performed studies in mice to evaluate how EGFR expression in tumor cells and myeloid cells contributes to development of colitis-associated cancer and ApcMin-dependent intestinal tumorigenesis. METHODS: We performed immunohistochemical and immunofluorescent analyses of 116 colorectal tumor biopsies to determine levels of EGFR in tumor and stroma; we also collected information on tumor stage and patient features and outcomes. We used the Mann-Whitney U and Kruskal-Wallis tests to correlate tumor levels of EGFR with tumor stage, and the Kaplan-Meier method to estimate patients' median survival time. We performed experiments in mice lacking EGFR in intestinal epithelial cells (Villin-Cre; Egfrf/f and Villin-CreERT2; Egfrf/f mice) or myeloid cells (LysM-Cre; Egfrf/f mice) on a mixed background. These mice were bred with ApcMin/+ mice; colitis-associated cancer and colitis were induced by administration of dextran sodium sulfate (DSS), with or without azoxymethane (AOM), respectively. Villin-CreERT2 was activated in developed tumors by administration of tamoxifen to mice. Littermates that expressed full-length EGFR were used as controls. Intestinal tissues were collected; severity of colitis, numbers and size of tumors, and intestinal barrier integrity were assessed by histologic, immunohistochemical, quantitative reverse transcription polymerase chain reaction, and flow cytometry analyses. RESULTS: We detected EGFR in myeloid cells in the stroma of human colorectal tumors; myeloid cell expression of EGFR associated with tumor metastasis and shorter patient survival time. Mice with deletion of EGFR from myeloid cells formed significantly fewer and smaller tumors than the respective EGFR-expressing controls in an ApcMin/+ background as well as after administration of AOM and DSS. Deletion of EGFR from intestinal epithelial cells did not affect tumor growth. Furthermore, tamoxifen-induced deletion of EGFR from epithelial cells of established intestinal tumors in mice given AOM and DSS did not reduce tumor size. EGFR signaling in myeloid cells promoted activation of STAT3 and expression of survivin in intestinal tumor cells. Mice with deletion of EGFR from myeloid cells developed more severe colitis after DSS administration, characterized by increased intestinal inflammation and intestinal barrier disruption, than control mice or mice with deletion of EGFR from intestinal epithelial cells. EGFR-deficient myeloid cells in the colon of DSS-treated LysM-Cre; Egfrf/f mice had reduced expression of interleukin 6 (IL6), and epithelial STAT3 activation was reduced compared with controls. Administration of recombinant IL6 to LysM-Cre; Egfrf/f mice given DSS protected them from weight loss and restored epithelial proliferation and STAT3 activation, compared with administration of DSS alone to these mice. CONCLUSIONS: Increased expression of EGFR in myeloid cells from the colorectal tumor stroma associates with tumor progression and reduced survival time of patients with metastatic colorectal cancer. Deletion of EGFR from myeloid cells, but not intestinal epithelial cells, protects mice from colitis-induced intestinal cancer and ApcMin-dependent intestinal tumorigenesis. Myeloid cell expression of EGFR increases activation of STAT3 and expression of survivin in intestinal epithelial cells and expression of IL6 in colon tissues. These findings indicate that expression of EGFR by myeloid cells of the colorectal tumor stroma, rather than the cancer cells themselves, contributes to tumor development.


Subject(s)
Colitis/complications , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/pathology , ErbB Receptors/analysis , ErbB Receptors/metabolism , Intestinal Mucosa/metabolism , Myeloid Cells/chemistry , STAT3 Transcription Factor/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Azoxymethane , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Dextran Sulfate , Epithelial Cells/metabolism , ErbB Receptors/genetics , Humans , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-6/metabolism , Interleukin-6/pharmacology , Intestinal Mucosa/pathology , Kaplan-Meier Estimate , Mice , Myeloid Cells/metabolism , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Repressor Proteins/metabolism , Signal Transduction , Survival Rate , Survivin , Tumor Burden
6.
J Exp Med ; 214(2): 423-437, 2017 02.
Article in English | MEDLINE | ID: mdl-28082356

ABSTRACT

Inhibition of the IκB kinase complex (IKK) has been implicated in the therapy of several chronic inflammatory diseases including inflammatory bowel diseases. In this study, using mice with an inactivatable IKKα kinase (IkkαAA/AA), we show that loss of IKKα function markedly impairs epithelial regeneration in a model of acute colitis. Mechanistically, this is caused by compromised secretion of cytoprotective IL-18 from IKKα-mutant intestinal epithelial cells because of elevated caspase 12 activation during an enhanced unfolded protein response (UPR). Induction of the UPR is linked to decreased ATG16L1 stabilization in IkkαAA/AA mice. We demonstrate that both TNF-R and nucleotide-binding oligomerization domain stimulation promote ATG16L1 stabilization via IKKα-dependent phosphorylation of ATG16L1 at Ser278. Thus, we propose IKKα as a central mediator sensing both cytokine and microbial stimulation to suppress endoplasmic reticulum stress, thereby assuring antiinflammatory function during acute intestinal inflammation.


Subject(s)
Carrier Proteins/metabolism , I-kappa B Kinase/physiology , Inflammation/metabolism , Animals , Autophagy-Related Proteins , Carrier Proteins/chemistry , Caspase 12/physiology , Colitis/prevention & control , Endoplasmic Reticulum Stress , Endoribonucleases/physiology , Interleukin-18/metabolism , Mice , NF-kappa B/physiology , Nod2 Signaling Adaptor Protein/physiology , Protein Serine-Threonine Kinases/physiology , Protein Stability , Unfolded Protein Response
7.
Oncotarget ; 6(30): 28833-50, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26353931

ABSTRACT

NF-κB activation depends on the IKK complex consisting of the catalytically active IKK1 and 2 subunits and the scaffold protein NEMO. Hitherto, IKK2 activation has always been associated with IκBα degradation, NF-κB activation, and cytokine production. In contrast, we found that in SCF-stimulated primary bone marrow-derived mast cells (BMMCs), IKK2 is alternatively activated. Mechanistically, activated TAK1 mediates the association between c-Kit and IKK2 and therefore facilitates the Lyn-dependent IKK2 activation which suffices to mediate mitogenic signaling but, surprisingly, does not result in NF-κB activation. Moreover, the c-Kit-mediated and Lyn-dependent IKK2 activation is targeted by MyD88-dependent pathways leading to enhanced IKK2 activation and therefore to potentiated effector functions. In neoplastic cells, expressing constitutively active c-Kit mutants, activated TAK1 and IKKs do also not induce NF-κB activation but mediate uncontrolled proliferation, resistance to apoptosis and enables IL-33 to mediate c-Kit-dependent signaling. Together, we identified the formation of the c-Kit-Lyn-TAK1 signalosome which mediates IKK2 activation. Unexpectedly, this IKK activation is uncoupled from the NF-κB-machinery but is critical to modulate functional cell responses in primary-, and mediates uncontrolled proliferation and survival of tumor-mast cells. Therefore, targeting TAK1 and IKKs might be a novel approach to treat c-Kit-driven diseases.


Subject(s)
I-kappa B Kinase/metabolism , MAP Kinase Kinase Kinases/metabolism , Mast Cells/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Stem Cell Factor/pharmacology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme Activation , Genotype , HEK293 Cells , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/deficiency , I-kappa B Kinase/genetics , Interleukin-33/metabolism , MAP Kinase Kinase Kinases/genetics , Mast Cells/enzymology , Mast Cells/pathology , Mice, Knockout , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Primary Cell Culture , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Time Factors , Transfection , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
Oncotarget ; 6(7): 5354-68, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25749030

ABSTRACT

Mast cell differentiation and proliferation depends on IL-3. IL-3 induces the activation of MAP-kinases and STATs and consequently induces proliferation and survival. Dysregulation of IL-3 signaling pathways also contribute to inflammation and tumorigenesis. We show here that IL-3 induces a SFK- and Ca²âº-dependent activation of the inhibitor of κB kinases 2 (IKK2) which results in mast cell proliferation and survival but does not induce IκBα-degradation and NFκB activation. Therefore we propose the term "subthreshold IKK activation".This subthreshold IKK activation also primes mast cells for enhanced responsiveness to IL-33R signaling. Consequently, co-stimulation with IL-3 and IL-33 increases IKK activation and massively enhances cytokine production induced by IL-33.We further reveal that in neoplastic mast cells expressing constitutively active Ras, subthreshold IKK activation is associated with uncontrolled proliferation. Consequently, pharmacological IKK inhibition reduces tumor growth selectively by inducing apoptosis in vivo.Together, subthreshold IKK activation is crucial to mediate the full IL-33-induced effector functions in primary mast cells and to mediate uncontrolled proliferation of neoplastic mast cells. Thus, IKK2 is a new molecularly defined target structure.


Subject(s)
Cell Transformation, Neoplastic/pathology , I-kappa B Kinase/metabolism , Interleukin-3/pharmacology , Mast Cells/pathology , NF-kappa B/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Homeodomain Proteins/physiology , Humans , I-kappa B Kinase/genetics , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred DBA , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , NF-kappa B/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Cell Rep ; 7(6): 1914-25, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24882009

ABSTRACT

The recruitment of immune cells into solid tumors is an essential prerequisite of tumor development. Depending on the prevailing polarization profile of these infiltrating leucocytes, tumorigenesis is either promoted or blocked. Here, we identify IκB kinase α (IKKα) as a central regulator of a tumoricidal microenvironment during intestinal carcinogenesis. Mice deficient in IKKα kinase activity are largely protected from intestinal tumor development that is dependent on the enhanced recruitment of interferon γ (IFNγ)-expressing M1-like myeloid cells. In IKKα mutant mice, M1-like polarization is not controlled in a cell-autonomous manner but, rather, depends on the interplay of both IKKα mutant tumor epithelia and immune cells. Because therapies aiming at the tumor microenvironment rather than directly at the mutated cancer cell may circumvent resistance development, we suggest IKKα as a promising target for colorectal cancer (CRC) therapy.


Subject(s)
Carcinogenesis/metabolism , I-kappa B Kinase/metabolism , Intestines/immunology , Killer Cells, Natural/pathology , Myeloid Cells/cytology , Myeloid Cells/enzymology , Animals , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/pathology , Carcinogenesis/pathology , Cell Polarity , Cell Transformation, Neoplastic , HEK293 Cells , Humans , Killer Cells, Natural/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Phosphorylation , Signal Transduction
10.
J Immunol ; 191(2): 837-47, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772033

ABSTRACT

The p53 protein has not only important tumor suppressor activity but also additional immunological and other functions, whose nature and extent are just beginning to be recognized. In this article, we show that p53 has a novel inflammation-promoting action in the intestinal tract, because loss of p53 or the upstream activating kinase, ATM, protects against acute intestinal inflammation in murine models. Mechanistically, deficiency in p53 leads to increased survival of epithelial cells and lamina propria macrophages, higher IL-6 expression owing to enhanced glucose-dependent NF-κB activation, and increased mucosal STAT3 activation. Blockade or loss of IL-6 signaling reverses the protective effects of p53 deficiency. Conversely, IL-6 treatment protects against acute colitis in a manner dependent on STAT3 signaling and induction of cytoprotective factors in epithelial cells. Together, these results indicate that p53 promotes inflammation in the intestinal tract through suppression of epithelium-protective factors, thus significantly expanding the spectrum of physiological and immunological p53 activities unrelated to cancer formation.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Inflammation/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Ataxia Telangiectasia Mutated Proteins , Bone Marrow Cells/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Colitis/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Enzyme Activation , Epithelial Cells/metabolism , Inflammation/prevention & control , Interleukin-6/biosynthesis , Interleukin-6/pharmacology , Interleukins/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL
...