Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 16(3): e1008340, 2020 03.
Article in English | MEDLINE | ID: mdl-32226027

ABSTRACT

Combination immunotherapy (CIT) is currently applied as a treatment for different cancers and is proposed as a cure strategy for chronic viral infections. Whether such therapies are efficient during an acute infection remains elusive. To address this, inhibitory receptors were blocked and regulatory T cells depleted in acutely Friend retrovirus-infected mice. CIT resulted in a dramatic expansion of cytotoxic CD4+ and CD8+ T cells and a subsequent reduction in viral loads. Despite limited viral replication, mice developed fatal immunopathology after CIT. The pathology was most severe in the gastrointestinal tract and was mediated by granzyme B producing CD4+ and CD8+ T cells. A similar post-CIT pathology during acute Influenza virus infection of mice was observed, which could be prevented by vaccination. Melanoma patients who developed immune-related adverse events under immune checkpoint CIT also presented with expanded granzyme-expressing CD4+ and CD8+ T cell populations. Our data suggest that acute infections may induce immunopathology in patients treated with CIT, and that effective measures for infection prevention should be applied.


Subject(s)
Antibodies/administration & dosage , Melanoma/immunology , Melanoma/therapy , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology , Animals , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Friend murine leukemia virus/physiology , Humans , Immunotherapy/adverse effects , Melanoma/pathology , Mice , Mice, Inbred C57BL , Retroviridae Infections/pathology , Retroviridae Infections/virology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
2.
J Immunol ; 203(6): 1636-1649, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31383741

ABSTRACT

Balanced control of innate immune signaling in the intestine represents an important host defense mechanism to avoid inappropriate responses that may exacerbate mucosal injury in acute inflammation. In this study, we report that TRIM58, a RING E3-ubiquitin ligase, associates with TLR2. The interaction was found in a yeast two-hybrid screen (human leukocyte and mononuclear library) and confirmed by coimmunoprecipitation of tagged and endogenous proteins. TRIM58 was predominantly expressed by murine and human myeloid-derived cells. Stimulation with a TLR2 ligand modulated TRIM58 synthesis in myeloid cells. Overexpression of TRIM58, but only in presence of the RING domain, promoted proteasome-dependent degradation of TLR2, inhibiting its signaling activity. Genetic deletion of Trim58 in mice (Trim58 -/-) led to impaired resolution of acute dextran sodium sulfate-induced colitis, which was characterized by delayed recovery from colonic injury and associated with enhanced expression of TLR2 protein and proinflammatory cyto/chemokine production in inflamed colons. Using myeloid cell-specific deletion of Trim58 in mice, we demonstrated that the myeloid cell compartment was responsible for early colitis acceleration in Trim58 deficiency. In vitro studies revealed that Trim58 -/- myeloid cells, which showed constitutive upregulation of TLR2 protein, overreacted to a proinflammatory milieu (TNF-α and IFN-γ) with increased IL-1ß protein production, which mechanistically depended on Tlr2 Finally, we found that TRIM58 mRNA and protein expression levels were reduced in colonic specimens from patients with ulcerative colitis. In conclusion, we identify TRIM58 as a novel negative mediator of innate immune control and mucosal homeostasis via TLR2 signaling. Dysfunction of TRIM58 in myeloid cells may contribute to ulcerative colitis pathogenesis.


Subject(s)
Inflammation/metabolism , Intestinal Mucosa/metabolism , Myeloid Cells/metabolism , Toll-Like Receptor 2/metabolism , Tripartite Motif Proteins/metabolism , Animals , Caco-2 Cells , Cell Line , Cell Line, Tumor , Colitis, Ulcerative/metabolism , Colon/metabolism , Disease Models, Animal , Female , HEK293 Cells , HL-60 Cells , Humans , Interferon-gamma/metabolism , Intestines/pathology , Male , Mice , Mucositis/metabolism , Signal Transduction/physiology , THP-1 Cells
3.
Sci Rep ; 7(1): 7629, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28794502

ABSTRACT

Remote ischemic preconditioning (RIPC) by repeated brief cycles of limb ischemia/reperfusion reduces myocardial ischemia/reperfusion injury. In left ventricular (LV) biopsies from patients undergoing coronary artery bypass grafting (CABG), only the activation of signal transducer and activator of transcription 5 was associated with RIPC's cardioprotection. We have now used an unbiased, non-hypothesis-driven proteomics and phosphoproteomics approach to analyze LV biopsies from patients undergoing CABG and from pigs undergoing coronary occlusion/reperfusion without (sham) and with RIPC. False discovery rate-based statistics identified a higher prostaglandin reductase 2 expression at early reperfusion with RIPC than with sham in patients. In pigs, the phosphorylation of 116 proteins was different between baseline and early reperfusion with RIPC and/or with sham. The identified proteins were not identical for patients and pigs, but in-silico pathway analysis of proteins with ≥2-fold higher expression/phosphorylation at early reperfusion with RIPC in comparison to sham revealed a relation to mitochondria and cytoskeleton in both species. Apart from limitations of the proteomics analysis per se, the small cohorts, the sampling/sample processing and the number of uncharacterized/unverifiable porcine proteins may have contributed to this largely unsatisfactory result.


Subject(s)
Coronary Artery Bypass , Coronary Occlusion/pathology , Heart Ventricles/pathology , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion , Phosphoproteins/analysis , Proteome/analysis , Aged , Animals , Biopsy , Computational Biology , Female , Humans , Male , Models, Animal , Proteomics , Swine
4.
PLoS One ; 12(7): e0180834, 2017.
Article in English | MEDLINE | ID: mdl-28686677

ABSTRACT

Patients with Ulcerative Colitis (UC) have an increased risk to develop colitis-associated colorectal cancer (CAC). Here, we found that protein expression of ABCB1 (ATP Binding Cassette Subfamily B Member 1) / MDR1 (multidrug resistance 1) was diminished in the intestinal mucosa of patients with active UC with or without CAC, but not in non-UC patients with sporadic colon cancer. We investigated the consequences of ABCB1/MDR1 loss-of-function in a common murine model for CAC (AOM/DSS). Mice deficient in MDR1A (MDR1A KO) showed enhanced intratumoral inflammation and cellular damage, which were associated with reduced colonic tumor size and decreased degree of dysplasia, when compared to wild-type (WT). Increased cell injury correlated with reduced capacity for growth of MDR1A KO tumor spheroids cultured ex-vivo. Gene expression analysis by microarray demonstrated that MDR1A deficiency shaped the inflammatory response towards an anti-tumorigenic microenvironment by downregulating genes known to be important mediators of cancer progression (PTGS2 (COX2), EREG, IL-11). MDR1A KO tumors showed increased gene expression of TNFSF10 (TRAIL), a known inducer of cancer cell death, and CCL12, a strong trigger of B cell chemotaxis. Abundant B220+ B lymphocyte infiltrates with interspersed CD138+ plasma cells were recruited to the MDR1A KO tumor microenvironment, concomitant with high levels of immunoglobulin light chain genes. In contrast, MDR1A deficiency in RAG2 KO mice that lack both B and T cells aggravated colonic tumor progression. MDR1A KO CD19+ B cells, but not WT CD19+ B cells, suppressed growth of colonic tumor-derived spheroids from AOM/DSS-WT mice in an ex-vivo co-culture system, implying that B-cell regulated immune responses contributed to delayed tumor development in MDR1A deficiency. In conclusion, we provide first evidence that loss of ABCB1/MDR1 function may represent an essential tumor-suppressive host defense mechanism in CAC.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/immunology , B-Lymphocytes/immunology , Colitis, Ulcerative/immunology , Colorectal Neoplasms/immunology , Gene Expression Regulation, Neoplastic , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , B-Lymphocytes/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Chemotaxis , Colitis, Ulcerative/complications , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colorectal Neoplasms/complications , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Disease Models, Animal , Epiregulin/genetics , Epiregulin/immunology , Genes, Immunoglobulin Light Chain/genetics , Humans , Interleukin-11/genetics , Interleukin-11/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Male , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/immunology , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology
5.
World J Gastroenterol ; 23(9): 1513-1520, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28321153

ABSTRACT

The gastrointestinal barrier is constantly exposed to numerous environmental substrates that are foreign and potentially harmful. These xenobiotics can cause shifts in the intestinal microbiota composition, affect mucosal immune responses, disturb tissue integrity and impair regeneration. The multidrug transporter ABCB1/MDR1 p-glycoprotein (p-gp) plays a key role at the front line of host defence by efficiently protecting the gastrointestinal barrier from xenobiotic accumulation. This Editorial discusses how altered expression and function of ABCB1/MDR1 p-gp may contribute to the development and persistence of chronic intestinal inflammation in inflammatory bowel diseases (IBD). Recent evidence implies multiple interactions between intestinal microbiota, innate immunity and xenobiotic metabolism via p-gp. While decreased efflux activity may promote disease susceptibility and drug toxicity, increased efflux activity may confer resistance to therapeutic drugs in IBD. Mice deficient in MDR1A develop spontaneously chronic colitis, providing a highly valuable murine IBD model for the study of intestinal epithelial barrier function, immunoregulation, infectious co-triggers and novel therapeutic approaches. Possible associations of human ABCB1 gene polymorphisms with IBD susceptibility have been evaluated, but results are inconsistent. Future studies must focus on further elucidation of the pathophysiological relevance and immunological functions of p-gp and how its ambiguous effects could be therapeutically targeted in IBD.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Inflammatory Bowel Diseases/immunology , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Chronic Disease , Colitis/genetics , Colitis/metabolism , Drug Resistance, Multiple , Gastrointestinal Microbiome , Gene Expression Regulation , Homeostasis , Humans , Immunity, Innate , Inflammation , Inflammatory Bowel Diseases/metabolism , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Xenobiotics/metabolism
6.
Oncotarget ; 7(46): 74415-74426, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27769070

ABSTRACT

Amplification or overexpression of MYCN is involved in development and maintenance of multiple malignancies. A subset of these tumors originates from neural precursors, including the most aggressive forms of the childhood tumors, neuroblastoma and medulloblastoma. In order to model the spectrum of MYCN-driven neoplasms in mice, we transgenically overexpressed MYCN under the control of the human GFAP-promoter that, among other targets, drives expression in neural progenitor cells. However, LSL-MYCN;hGFAP-Cre double transgenic mice did neither develop neural crest tumors nor tumors of the central nervous system, but presented with neuroendocrine tumors of the pancreas and, less frequently, the pituitary gland. Pituitary tumors expressed chromogranin A and closely resembled human pituitary adenomas. Pancreatic tumors strongly produced and secreted glucagon, suggesting that they derived from glucagon- and GFAP-positive islet cells. Interestingly, 3 out of 9 human pancreatic neuroendocrine tumors expressed MYCN, supporting the similarity of the mouse tumors to the human system. Serial transplantations of mouse tumor cells into immunocompromised mice confirmed their fully transformed phenotype. MYCN-directed treatment by AuroraA- or Brd4-inhibitors resulted in significantly decreased cell proliferation in vitro and reduced tumor growth in vivo. In summary, we provide a novel mouse model for neuroendocrine tumors of the pancreas and pituitary gland that is dependent on MYCN expression and that may help to evaluate MYCN-directed therapies.


Subject(s)
Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glucagon/biosynthesis , N-Myc Proto-Oncogene Protein/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Glial Fibrillary Acidic Protein/metabolism , Glucagonoma/genetics , Glucagonoma/metabolism , Glucagonoma/pathology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , N-Myc Proto-Oncogene Protein/metabolism , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Pituitary Neoplasms/pathology , Transcriptome
7.
PLoS One ; 11(6): e0156871, 2016.
Article in English | MEDLINE | ID: mdl-27271572

ABSTRACT

Mucinous adenocarcinoma (MAC) represents a distinct histopathological entity of colorectal cancer (CRC), which is associated with disease progression and poor prognosis. Here, we found that expression levels of miR-205 and miR-373 were specifically upregulated only in patients with mucinous colon cancers, but not in CRC that lack mucinous components. To investigate the effects of miR-205 and miR-373 on intestinal epithelial cell (IEC) biology by gain- and loss-of-function experiments in a proof-of-concept approach, we chose previously established in-vitro human Caco-2-based models of differentiated, non-invasive (expressing TLR4 wild-type; termed Caco-2[WT]) versus undifferentiated, invasive (expressing TLR4 mutant D299G; termed Caco-2[D299G]) IEC. Enterocyte-like Caco-2[WT] showed low levels of miR-205 and miR-373 expression, while both miRNAs were significantly upregulated in colorectal carcinoma-like Caco-2[D299G], thus resembling the miRNA expression pattern of paired normal versus tumor samples from MAC patients. Using stable transfection, we generated miR-205- or miR-373-expressing and miR-205- or miR-373-inhibiting subclones of these IEC lines. We found that introduction of miR-205 into Caco-2[WT] led to expansion of mucus-secreting goblet cell-like cells, which was associated with induction of KLF4, MUC2 and TGFß1 expression. Activation of miR-205 in Caco-2[WT] induced chemoresistance, while inhibition of miR-205 in Caco-2[D299G] promoted chemosensitivity. Caco-2[WT] overexpressing miR-373 showed mitotic abnormalities and underwent morphologic changes (loss of epithelial polarity, cytoskeletal reorganization, and junctional disruption) associated with epithelial-mesenchymal transition and progression to inflammation-associated colonic carcinoma, which correlated with induction of phosphorylated STAT3 and N-CADHERIN expression. Functionally, introduction of miR-373 into Caco-2[WT] mediated loss of cell-cell adhesion and increased proliferation and invasion. Reversely, inhibition of miR-373 allowed mesenchymal IEC to regain epithelial properties, which correlated with absence of neoplastic progression. Using xenografts in mice demonstrated miR-373-mediated acceleration of malignant intestinal tumor growth. In conclusion, our results provide first evidence that miR-205 and miR-373 may differentially contribute to the aggressive phenotype of MAC in CRC.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , MicroRNAs/physiology , Animals , Caco-2 Cells , Cells, Cultured , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Kruppel-Like Factor 4 , Mice , Mice, Nude , Neoplasm Invasiveness
8.
Curr Opin Support Palliat Care ; 10(2): 157-64, 2016 06.
Article in English | MEDLINE | ID: mdl-26986508

ABSTRACT

PURPOSE OF REVIEW: Intestinal mucositis represents a common complication and dose-limiting toxicity of cancer chemotherapy. So far chemotherapy-induced intestinal mucositis remains poorly treatable resulting in significant morbidity and reduced quality of life in cancer patients. This review discusses recent insights into the pathophysiology of chemotherapy-induced intestinal mucositis. Novel mechanisms linking gut microbiota, host innate immunity and anticancer drug metabolism are highlighted. RECENT FINDINGS: Gut microbiota may affect xenobiotic metabolism by direct and indirect mechanisms, critically modulating gut toxicity of chemotherapy drugs. Composition and metabolic function of the gut microbiome as well as innate immune responses of the intestinal mucosa are severely altered during chemotherapy. Commensal-mediated innate immune signaling via Toll-like receptors (TLRs) ambiguously shapes chemotherapy-induced genotoxic damage in the gastrointestinal tract. TLR2 may accelerate host detoxification by activating the multidrug transporter ATP-binding cassette 1 (ABCB1)/MDR1 P-glycoprotein to efflux harmful drugs, thus controlling the severity of cancer therapy-induced mucosal damage in the gastrointestinal tract. In contrast, selective chemotherapy drugs may drive LPS hyperresponsiveness via TLR4, which exacerbates mucosal injury through aberrant cytokine storms. Broad-spectrum antibiotic treatment does not seem to represent a valid therapeutic option, as drastic reduction in global gut microbiota may enhance risk of gastrointestinal toxicity and reduce efficacy of some chemotherapy drugs, at least in murine models. SUMMARY: Several variables (environment, metabolism, dysbiosis, infections and/or genetics) influence the outcome of mucosal TLR signaling during cancer treatment. Differences in innate immune responses also reflect chemotherapy drug-specific effects. Future studies must investigate in more detail whether manipulating the delicate balance between gut microbiota and host immune responses by either monotherapy or combinations of different TLR agonists and antagonists may be indeed useful to limit the toxic side-effects of complex chemotherapy regimens, accelerate mucosal tissue regeneration and improve the anticancer treatment response.


Subject(s)
Antineoplastic Agents/adverse effects , Intestinal Mucosa/physiopathology , Mucositis/chemically induced , Mucositis/physiopathology , Toll-Like Receptors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Dysbiosis/metabolism , Environment , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Immunity, Innate/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Xenobiotics/metabolism
9.
J Immunol ; 194(4): 1983-95, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25589072

ABSTRACT

Intestinal mucositis represents the most common complication of intensive chemotherapy, which has a severe adverse impact on quality of life of cancer patients. However, the precise pathophysiology remains to be clarified, and there is so far no successful therapeutic intervention. In this study, we investigated the role of innate immunity through TLR signaling in modulating genotoxic chemotherapy-induced small intestinal injury in vitro and in vivo. Genetic deletion of TLR2, but not MD-2, in mice resulted in severe chemotherapy-induced intestinal mucositis in the proximal jejunum with villous atrophy, accumulation of damaged DNA, CD11b(+)-myeloid cell infiltration, and significant gene alterations in xenobiotic metabolism, including a decrease in ABCB1/multidrug resistance (MDR)1 p-glycoprotein (p-gp) expression. Functionally, stimulation of TLR2 induced synthesis and drug efflux activity of ABCB1/MDR1 p-gp in murine and human CD11b(+)-myeloid cells, thus inhibiting chemotherapy-mediated cytotoxicity. Conversely, TLR2 activation failed to protect small intestinal tissues genetically deficient in MDR1A against DNA-damaging drug-induced apoptosis. Gut microbiota depletion by antibiotics led to increased susceptibility to chemotherapy-induced mucosal injury in wild-type mice, which was suppressed by administration of a TLR2 ligand, preserving ABCB1/MDR1 p-gp expression. Findings were confirmed in a preclinical model of human chemotherapy-induced intestinal mucositis using duodenal biopsies by demonstrating that TLR2 activation limited the toxic-inflammatory reaction and maintained assembly of the drug transporter p-gp. In conclusion, this study identifies a novel molecular link between innate immunity and xenobiotic metabolism. TLR2 acts as a central regulator of xenobiotic defense via the multidrug transporter ABCB1/MDR1 p-gp. Targeting TLR2 may represent a novel therapeutic approach in chemotherapy-induced intestinal mucositis.


Subject(s)
Antineoplastic Agents/adverse effects , Mucositis/immunology , Mucositis/microbiology , Signal Transduction/immunology , Toll-Like Receptor 2/metabolism , ATP Binding Cassette Transporter, Subfamily B/immunology , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoblotting , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Mucositis/chemically induced , Myeloid Cells/immunology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 2/immunology
10.
Pflugers Arch ; 466(10): 1953-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24390754

ABSTRACT

Ischemic postconditioning (PoCo) reduces infarct size following myocardial ischemia/reperfusion. To protect, PoCo must be performed early during reperfusion, and causal cardioprotective signaling must occur then. The role of microRNA (miRNA) in PoCo is unclear. Anesthetized pigs were subjected to 60 min left anterior descending coronary artery (LAD) occlusion and 180 min reperfusion. Immediate full reperfusion (IFR, n = 5) was compared to PoCo (four cycles of 60 s/60 s reperfusion/reocclusion, n = 5). Transmural myocardial biopsies from the LAD territory were sampled at baseline, 60 min ischemia, 10 and 180 min reperfusion. RNA was isolated. The expression of 11 miRNAs, including muscle-specific (miRNA-1, -133a, -206, -208b, -214, and -499), fibrosis- (miRNA-21, -24, and -29b), neovascularization- (miRNA-92a), and inflammation-associated (miRNA-146b) candidates, was quantified using real-time PCR (RT-PCR). mRNA expression at baseline and 180 min reperfusion was quantified and validated (microarray and RT-PCR). PoCo reduced infarct size from 44.9 ± 7.7 to 34.8 ± 5.3% of the area at risk. The expression of miRNA-1, -24, -29b, -133a, -146b, -208b, and -499 was increased at 10 min reperfusion with PoCo vs. IFR; however, that of miRNA-1, -24, -208b, and -499 was already increased at 60 min ischemia and probably reflects falsely positive results. Five mRNAs were different with PoCo vs. IFR. In silico analysis identified a tentative connection between three miRNAs and five mRNAs with the biological functions "cell death", "inflammatory response" and/or "glucose metabolism". If at all, only miRNA-29b, -133a, and -146b fulfill the minimal temporal requirements for a potential causal involvement in cardioprotection by PoCo.


Subject(s)
Ischemic Postconditioning , MicroRNAs/metabolism , Myocardial Reperfusion Injury/metabolism , Animals , MicroRNAs/genetics , Myocardial Reperfusion Injury/therapy , Swine , Swine, Miniature
11.
Oncoimmunology ; 2(7): e24890, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-24073372

ABSTRACT

Homeostatic TLR4 signaling protects the intestinal epithelium in health. Evidence suggests that perturbed TLR4 signaling is linked to carcinogenesis. We have recently demonstrated that the common human TLR4 variant D299G exerts pro-inflammatory effects and drives malignant tumor progression in human colon cancer.

12.
J Immunol ; 190(11): 5676-88, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23636052

ABSTRACT

Variants of the multidrug resistance gene (MDR1/ABCB1) have been associated with increased susceptibility to severe ulcerative colitis (UC). In this study, we investigated the role of TLR/IL-1R signaling pathways including the common adaptor MyD88 in the pathogenesis of chronic colonic inflammation in MDR1A deficiency. Double- or triple-null mice lacking TLR2, MD-2, MyD88, and MDR1A were generated in the FVB/N background. Deletion of TLR2 in MDR1A deficiency resulted in fulminant pancolitis with early expansion of CD11b(+) myeloid cells and rapid shift toward TH1-dominant immune responses in the lamina propria. Colitis exacerbation in TLR2/MDR1A double-knockout mice required the unaltered commensal microbiota and the LPS coreceptor MD-2. Blockade of IL-1ß activity by treatment with IL-1R antagonist (IL-1Ra; Anakinra) inhibited colitis acceleration in TLR2/MDR1A double deficiency; intestinal CD11b(+)Ly6C(+)-derived IL-1ß production and inflammation entirely depended on MyD88. TLR2/MDR1A double-knockout CD11b(+) myeloid cells expressed MD-2/TLR4 and hyperresponded to nonpathogenic Escherichia coli or LPS with reactive oxygen species production and caspase-1 activation, leading to excessive cell death and release of proinflammatory IL-1ß, consistent with pyroptosis. Inhibition of reactive oxygen species-mediated lysosome degradation suppressed LPS hyperresponsiveness. Finally, active UC in patients carrying the TLR2-R753Q and MDR1-C3435T polymorphisms was associated with increased nuclear expression of caspase-1 protein and cell death in areas of acute inflammation, compared with active UC patients without these variants. In conclusion, we show that the combined defect of two UC susceptibility genes, MDR1A and TLR2, sets the stage for spontaneous and uncontrolled colitis progression through MD-2 and IL-1R signaling via MyD88, and we identify commensally induced pyroptosis as a potential innate immune effector in severe UC pathogenesis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Toll-Like Receptor 2/genetics , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD11b Antigen , Caspase 1/metabolism , Cell Death/genetics , Cell Death/immunology , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Progression , Gene Deletion , Humans , Interleukin-1beta/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Antigen 96/metabolism , Lysosomes/metabolism , Male , Mice , Mice, Knockout , Mutation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 2/deficiency
13.
Curr Opin Gastroenterol ; 29(1): 85-91, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23207600

ABSTRACT

PURPOSE OF REVIEW: This review focuses on recent advances and novel insights into the mechanistic events that may link commensal microbiota and host innate immunity in the pathophysiology of intestinal inflammation and neoplasia. Unanswered questions are discussed and future perspectives in the field are highlighted. RECENT FINDINGS: Commensal microbiota, host innate immunity, and genetics form a multidimensional network that controls homeostasis of the mucosal barrier in the intestine. Large-scale sequencing projects have begun to catalog the healthy human microbiome. Converging evidence suggests that alterations in the regulation of the complex host environment [e.g., dysbiosis and overgrowth of select commensal bacterial species, dietary factors, copresence of facultative pathogens (including viruses), and changes in mucus characteristics] may trigger aberrant innate immune signaling, thereby contributing to the development of intestinal inflammation and associated colon cancer in the susceptible individual. Genetically determined innate immune malfunction may create an inflammatory environment that promotes tumor progression (such as the TLR4-D299G mutation). SUMMARY: The next challenging steps to be taken are to decipher changes in the human microbiome (and virome) during well defined diseased states, and relate them to intestinal mucosal immune functions and host genotypes.


Subject(s)
Enterocolitis/immunology , Immunity, Innate , Intestinal Neoplasms/immunology , Metagenome/immunology , Microbial Interactions , Enterocolitis/microbiology , Humans , Intestinal Neoplasms/microbiology , Signal Transduction
14.
Dig Dis ; 30(4): 334-40, 2012.
Article in English | MEDLINE | ID: mdl-22796793

ABSTRACT

Commensal microbiota plays a key role in the health and disease of the host. The innate immune system comprises an essential functional component of the intestinal mucosal barrier, maintaining hyporesponsiveness to omnipresent harmless commensals in the lumen, but rapidly recognizing and combating invading bacteria through diverse antimicrobial mechanisms. Interactions between commensals and innate immune cells are constant, multidimensional and entirely context-dependent. Environment, genetics and host defense differentially modulate commensal-innate immune effects and functions in the intestinal mucosa. In IBD, dysbiosis, mucus layer disruption, impairment in bacterial clearance, intestinal epithelial cell barrier dysfunction and/or immune cell deregulation may lead to commensal-innate immune miscommunication, which critically drives mucosal inflammation and associated cancer.


Subject(s)
Bacteria/immunology , Immunity/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , Epithelium/immunology , Epithelium/microbiology , Epithelium/pathology , Humans , Mucus/immunology , Mucus/microbiology
15.
Gastroenterology ; 141(6): 2154-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21920464

ABSTRACT

BACKGROUND & AIMS: The Toll-like receptor (TLR) 4 mediates homeostasis of the intestinal epithelial cell (IEC) barrier. We investigated the effects of TLR4-D299G on IEC functions. METHODS: We engineered IECs (Caco-2) to stably overexpress hemagglutinin-tagged wild-type TLR4, TLR4-D299G, or TLR4-T399I. We performed gene expression profiling using DNA microarray analysis. Findings were confirmed by real-time, quantitative, reverse-transcriptase polymerase chain reaction, immunoblot, enzyme-linked immunosorbent assay, confocal immunofluorescence, and functional analyses. Tumorigenicity was tested using the CD1 nu/nu mice xenograft model. Human colon cancer specimens (N = 214) were genotyped and assessed for disease stage. RESULTS: Caco-2 cells that expressed TLR4-D299G underwent the epithelial-mesenchymal transition and morphologic changes associated with tumor progression, whereas cells that expressed wild-type TLR4 or TLR4-T399I did not. Caco-2 cells that expressed TLR4-D299G had significant increases in expression levels of genes and proteins associated with inflammation and/or tumorigenesis compared with cells that expressed other forms of TLR4. The invasive activity of TLR4-D299G Caco-2 cells required Wnt-dependent activation of STAT3. In mice, intestinal xenograft tumors grew from Caco-2 cells that expressed TLR4-D299G, but not cells that expressed other forms of TLR4; tumor growth was blocked by a specific inhibitor of STAT3. Human colon adenocarcinomas from patients with TLR4-D299G were more frequently of an advanced stage (International Union Against Cancer [UICC] ≥III, 70% vs 46%; P = .0142) with metastasis (UICC IV, 42% vs 19%; P = .0065) than those with wild-type TLR4. Expression of STAT3 messenger RNA was higher among colonic adenocarcinomas with TLR4-D299G than those with wild-type TLR4. CONCLUSIONS: TLR4-D299G induces features of neoplastic progression in intestinal epithelial Caco-2 cells and associates with aggressive colon cancer in humans, implying a novel link between aberrant innate immunity and colonic cancerogenesis.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Disease Progression , Intestinal Mucosa/drug effects , Toll-Like Receptor 4/physiology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Animals , Caco-2 Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial-Mesenchymal Transition , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Inflammation/immunology , Male , Mice , Microscopy, Fluorescence , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Wnt Signaling Pathway
16.
Invest Ophthalmol Vis Sci ; 52(1): 256-63, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20847111

ABSTRACT

PURPOSE: CD36 is a Class B scavenger receptor that is constitutively expressed in the corneal epithelium and has been implicated in many homeostatic functions, including the homeostasis of the epidermal barrier. The aim of this study is to determine (1) whether CD36 is required for the maintenance of the corneal epithelial barrier to infection, and (2) whether CD36-deficient mice present with an increased susceptibility to bacterial keratitis. METHODS: The corneas of CD36(-/-), TSP1(-/-), TLR2(-/-), and C57BL/6 WT mice were screened via slit lamp microscopy or ex vivo analysis. The epithelial tight junctions and mucin layer were assessed via LC-biotin and Rose Bengal staining, respectively. Bacterial quantification was performed on corneal buttons and GFP-expressing Staphylococcus aureus was used to study bacterial binding. RESULTS: CD36(-/-) mice develop spontaneous corneal defects that increased in frequency and severity with age. The mild corneal defects were characterized by a disruption in epithelial tight junctions and the mucin layer, an infiltrate of macrophages, and increased bacterial binding. Bacterial quantification revealed high levels of Staphylococcus xylosus in the corneas of CD36(-/-) mice with severe defects, but not in wild-type controls. CONCLUSIONS: CD36(-/-) mice develop spontaneous bacterial keratitis independent of TLR2 and TSP1. The authors conclude that CD36 is a critical component of the corneal epithelial barrier, and in the absence of CD36 the barrier breaks down, allowing bacteria to bind to the corneal epithelium and resulting in spontaneous keratitis. This is the first report of spontaneous bacterial keratitis in mice.


Subject(s)
CD36 Antigens/physiology , Corneal Ulcer/microbiology , Eye Infections, Bacterial/microbiology , Staphylococcal Infections/microbiology , Animals , Corneal Ulcer/pathology , Epithelium, Corneal/metabolism , Eye Infections, Bacterial/pathology , Female , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/metabolism , Polymerase Chain Reaction , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Staphylococcal Infections/pathology , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Thrombospondin 1/physiology , Tight Junctions/metabolism , Toll-Like Receptor 2/physiology
17.
Inflamm Bowel Dis ; 16(9): 1583-97, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20803699

ABSTRACT

Differential alteration of Toll-like receptor (TLR) expression in inflammatory bowel disease (IBD) was first described 10 years ago. Since then, studies from many groups have led to the current concept that TLRs represent key mediators of innate host defense in the intestine, involved in maintaining mucosal as well as commensal homeostasis. Recent findings in diverse murine models of colitis have helped to reveal the mechanistic importance of TLR dysfunction in IBD pathogenesis. It has become evident that environment, genetics, and host immunity form a multidimensional and highly interactive regulatory triad that controls TLR function in the intestinal mucosa. Imbalanced relationships within this triad may promote aberrant TLR signaling, critically contributing to acute and chronic intestinal inflammatory processes in IBD colitis and associated cancer.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Toll-Like Receptors/metabolism , Animals , Humans , Inflammatory Bowel Diseases/pathology
18.
Curr Opin Gastroenterol ; 26(6): 583-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20664345

ABSTRACT

PURPOSE OF REVIEW: The intestinal epithelium serves as a highly dynamic immunologic frontier - exhibiting both innate and adaptive immune features. This review focuses on recent advances and novel insights into key intrinsic processes of the intestinal epithelium to closely monitor its intracellular and extracellular environment, communicate messages to neighbouring cells and rapidly initiate active defensive and repair measures, if necessary. RECENT FINDINGS: The intestinal epithelium is uniquely equipped with a vast array of features to control immune barrier homeostasis at the gates of the healthy intestinal mucosa. Deficient Toll-like receptor or NOD-like receptor signalling in the intestinal epithelium may imbalance commensal-dependent homeostasis, facilitating mucosal injury and leading to inflammatory disease. Dysfunction of the NLRP3 inflammasome may trigger aggravation of mucosal inflammation and cancer and has been associated with human inflammatory bowel diseases. Deregulated autophagy may alter inflammasome activity. SUMMARY: Exciting progress has been made in better understanding the complex diversity of physiological functions of innate immune responses in the intestinal epithelial barrier. Regulatory platforms of signalling mechanisms exist which are closely related and interact. However, many questions remain to be answered and more puzzles have arisen which are highlighted here.


Subject(s)
Immunity, Innate/physiology , Inflammasomes/immunology , Intestinal Mucosa/immunology , Animals , Cell Communication , Homeostasis/immunology , Humans , Immunity, Mucosal , Inflammasomes/physiology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/physiology , Signal Transduction , Toll-Like Receptors/immunology
19.
J Biol Chem ; 284(33): 22332-22343, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19528242

ABSTRACT

Gap junctional intercellular communication (GJIC) coordinates cellular functions essential for sustaining tissue homeostasis; yet its regulation in the intestine is not well understood. Here, we identify a novel physiological link between Toll-like receptor (TLR) 2 and GJIC through modulation of Connexin-43 (Cx43) during acute and chronic inflammatory injury of the intestinal epithelial cell (IEC) barrier. Data from in vitro studies reveal that TLR2 activation modulates Cx43 synthesis and increases GJIC via Cx43 during IEC injury. The ulcerative colitis-associated TLR2-R753Q mutant targets Cx43 for increased proteasomal degradation, impairing TLR2-mediated GJIC during intestinal epithelial wounding. In vivo studies using mucosal RNA interference show that TLR2-mediated mucosal healing depends functionally on intestinal epithelial Cx43 during acute inflammatory stress-induced damage. Mice deficient in TLR2 exhibit IEC-specific alterations in Cx43, whereas administration of a TLR2 agonist protects GJIC by blocking accumulation of Cx43 and its hyperphosphorylation at Ser368 to prevent spontaneous chronic colitis in MDR1alpha-deficient mice. Finally, adding the TLR2 agonist to three-dimensional intestinal mucosa-like cultures of human biopsies preserves intestinal epithelial Cx43 integrity and polarization ex vivo. In conclusion, Cx43 plays an important role in innate immune control of commensal-mediated intestinal epithelial wound repair.


Subject(s)
Connexin 43/physiology , Gene Expression Regulation , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/physiology , Animals , Caco-2 Cells , Connexin 43/metabolism , Epithelial Cells/metabolism , Gap Junctions , Humans , Immune System , Inflammation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Wound Healing
20.
Gastroenterology ; 137(1): 209-20, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19303021

ABSTRACT

BACKGROUND & AIMS: Goblet cells (GC) facilitate mucosal protection and epithelial barrier repair, yet the innate immune mechanisms that selectively drive GC functions have not been defined. The aim of this study was to determine whether Toll-like receptor (TLR) 2 and modulation of GC-derived trefoil factor (TFF) 3 are functionally linked in the intestine. METHODS: GC modulation was assessed using quantitative real-time polymerase chain reaction analysis (qRT-PCR), Western blotting, and confocal microscopy. Dextran sulfate sodium (DSS) colitis was induced in wild-type, TFF3(-/-), and TLR2(-/-) mice. Recombinant TLR2 ligand or TFF3 peptide were orally administered after DSS termination. Caco-2 cells overexpressing full-length TLR2 or mutant TLR2-R753Q were tested for TFF3 synthesis and functional-related effects in a wounding assay. RESULTS: Data from in vitro (Ls174T) and ex vivo models of murine and human GC reveal that TLR2 activation selectively induces synthesis of TFF3. In vivo studies using TFF3(-/-) or TLR2(-/-) mice demonstrate the ability for oral treatment with a TLR2 agonist to confer antiapoptotic protection of the intestinal mucosa against inflammatory stress-induced damage through TFF3. Recombinant TFF3 rescues TLR2-deficient mice from increased morbidity and mortality during acute colonic injury. Severe ulcerative colitis (UC) has recently been found to be associated with the R753Q polymorphism of the TLR2 gene. The relevance of the observed functional effect of TLR2 in regulating GC is confirmed by the finding that the UC-associated TLR2-R753Q variant is functionally deficient in the ability to induce TFF3 synthesis, thus leading to impaired wound healing. CONCLUSIONS: These data demonstrate a novel function of TLR2 in intestinal GC that links products of commensal bacteria to innate immune protection of the host via TFF3.


Subject(s)
Colitis/metabolism , Goblet Cells/metabolism , Intestinal Mucosa/metabolism , Mucins/metabolism , Toll-Like Receptor 2/metabolism , Wound Healing , Animals , Apoptosis , Caco-2 Cells , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colitis/prevention & control , Dextran Sulfate , Disease Models, Animal , Goblet Cells/immunology , Goblet Cells/pathology , Humans , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucins/deficiency , Mucins/genetics , Mutation , Organ Culture Techniques , Recombinant Proteins/metabolism , Time Factors , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Transfection , Trefoil Factor-3
SELECTION OF CITATIONS
SEARCH DETAIL
...