Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Gastroenterology ; 142(7): 1483-92.e6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22387394

ABSTRACT

BACKGROUND & AIMS: Polymorphisms that reduce the function of nucleotide-binding oligomerization domain (NOD)2, a bacterial sensor, have been associated with Crohn's disease (CD). No proteins that regulate NOD2 activity have been identified as selective pharmacologic targets. We sought to discover regulators of NOD2 that might be pharmacologic targets for CD therapies. METHODS: Carbamoyl phosphate synthetase/aspartate transcarbamylase/dihydroorotase (CAD) is an enzyme required for de novo pyrimidine nucleotide synthesis; it was identified as a NOD2-interacting protein by immunoprecipitation-coupled mass spectrometry. CAD expression was assessed in colon tissues from individuals with and without inflammatory bowel disease by immunohistochemistry. The interaction between CAD and NOD2 was assessed in human HCT116 intestinal epithelial cells by immunoprecipitation, immunoblot, reporter gene, and gentamicin protection assays. We also analyzed human cell lines that express variants of NOD2 and the effects of RNA interference, overexpression and CAD inhibitors. RESULTS: CAD was identified as a NOD2-interacting protein expressed at increased levels in the intestinal epithelium of patients with CD compared with controls. Overexpression of CAD inhibited NOD2-dependent activation of nuclear factor κB and p38 mitogen-activated protein kinase, as well as intracellular killing of Salmonella. Reduction of CAD expression or administration of CAD inhibitors increased NOD2-dependent signaling and antibacterial functions of NOD2 variants that are and are not associated with CD. CONCLUSIONS: The nucleotide synthesis enzyme CAD is a negative regulator of NOD2. The antibacterial function of NOD2 variants that have been associated with CD increased in response to pharmacologic inhibition of CAD. CAD is a potential therapeutic target for CD.


Subject(s)
Aspartate Carbamoyltransferase/physiology , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/physiology , Crohn Disease/immunology , Deoxyribonucleases/physiology , Dihydroorotase/physiology , Intestinal Mucosa/microbiology , Nod2 Signaling Adaptor Protein/immunology , Aspartate Carbamoyltransferase/antagonists & inhibitors , Aspartate Carbamoyltransferase/therapeutic use , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/antagonists & inhibitors , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/therapeutic use , Cell Line , Cells, Cultured , Crohn Disease/drug therapy , Crohn Disease/microbiology , Dihydroorotase/antagonists & inhibitors , Dihydroorotase/therapeutic use , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Immunoprecipitation , Intestinal Mucosa/immunology , Mass Spectrometry , NF-kappa B/physiology , Nod2 Signaling Adaptor Protein/physiology , Salmonella/growth & development , Salmonella/immunology , Signal Transduction
2.
Gastroenterology ; 139(5): 1630-41, 1641.e1-2, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20637199

ABSTRACT

BACKGROUND & AIMS: The identification of numerous genes that confer susceptibility to Crohn's disease (CD) indicates that this complex disease might arise from alterations in several genes with related functions. We examined the functional interaction between the CD risk genes ATG16L1 and NOD2 to identify an autophagy-dependent pathway that is altered by disease-associated variants. METHODS: We assessed Nod2 signaling and autophagy activation in response to muramyl dipeptide (MDP) by immunoblot, confocal microscopy, flow cytometry, reporter gene, and gentamicin protection assays in human epithelial cell lines and primary human macrophages and dendritic cells from healthy individuals. The requirement of Nod2 and ATG16L1 expression and the effects of CD-associated variants in MDP-stimulated autophagy and Nod2-dependent signaling were assessed in cell lines manipulated by RNA interference, inhibitors, or ATG16L1 or NOD2 variants and in primary macrophages and dendritic cells from healthy genotyped donors. RESULTS: MDP stimulation of epithelial cells, macrophages, and dendritic cells activated autophagy and nuclear factor κB and mitogen-activated protein kinase signaling; it also increased killing of Salmonella. These responses depended on ATG16L1 and Nod2 expression and were impaired by CD-associated NOD2 variants. Nod2-dependent signaling was not impaired in cells with the ATG16L1 T300A genotype, which is associated with CD. However, the ATG16L1 T300A variant blocked the increase in MDP-mediated killing of Salmonella only in epithelial cell lines and not primary macrophages or dendritic cells. CONCLUSIONS: ATG16L1 and NOD2 are components of an autophagy-mediated antibacterial pathway that is altered in a cell- and function-specific manner by CD-associated mutations.


Subject(s)
Autophagy , Carrier Proteins/genetics , Crohn Disease/genetics , Gene Expression , Genetic Predisposition to Disease , Nod2 Signaling Adaptor Protein/genetics , RNA/genetics , Autophagy-Related Proteins , Carrier Proteins/metabolism , Cell Line , Crohn Disease/metabolism , Crohn Disease/pathology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genotype , Humans , Immunoblotting , Microscopy, Confocal , Nod2 Signaling Adaptor Protein/metabolism , Phagocytosis
3.
Cancer Res ; 65(12): 5428-38, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958592

ABSTRACT

Here we report that glioblastoma multiforme (GBM) mediates immunosuppression by promoting T-cell death via tumor-associated CD70 and gangliosides that act through receptor-dependent and receptor-independent pathways, respectively. GBM lines cocultured with T cells induced lymphocyte death. The GBM lines were characterized for their expression of CD70, Fas ligand (FasL), and tumor necrosis factor-alpha (TNF-alpha), and the possible participation of those molecules in T-cell killing was assessed by doing GBM/T cell cocultures in the presence of anti-CD70 antibodies, Fas fusion proteins, or anti-TNF-alpha antibodies. CD70 but not TNF-alpha or FasL is responsible for initiating T-cell death via the receptor-dependent pathway. Of the four GBM cell lines that induced T-cell death, three highly expressed CD70. Two nonapoptogenic GBM lines (CCF3 and U138), on the other hand, had only minimally detectable CD70 expression. Blocking experiments with the anti-CD70 antibody confirmed that elevated CD70 levels were involved in the apoptogenicity of the three GBM lines expressing that molecule. Gangliosides were found to participate in the induction of T-cell apoptosis, because the glucosylceramide synthase inhibitor (PPPP) significantly reduced the abilities of all four apoptogenic lines to kill the lymphocytes. High-performance liquid chromatography (HPLC) and mass spectroscopy revealed that GM2, GM2-like gangliosides, and GD1a were synthesized in abundance by all four apoptogenic GBM lines but not by the two GBMs lacking activity. Furthermore, gangliosides isolated from GBM lines as well as HPLC fractions containing GM2 and GD1a were directly apoptogenic for T cells. Our results indicate that CD70 and gangliosides are both products synthesized by GBMs that may be key mediators of T-cell apoptosis and likely contribute to the T-cell dysfunction observed within the tumor microenvironment.


Subject(s)
Antigens, CD/immunology , Brain Neoplasms/immunology , Gangliosides/immunology , Glioblastoma/immunology , Membrane Proteins/immunology , T-Lymphocytes/immunology , Antigens, CD/biosynthesis , Apoptosis/immunology , Brain Neoplasms/pathology , CD27 Ligand , Cell Line, Tumor , Coculture Techniques , Fas Ligand Protein , Glioblastoma/pathology , Humans , Lymphocyte Activation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Membrane Proteins/biosynthesis , Receptors, Tumor Necrosis Factor/physiology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
4.
Clin Cancer Res ; 11(1): 226-31, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15671550

ABSTRACT

Whereas normally expressed at sites of cell-to-cell contact in adult epithelial tissues, recent studies have shown that the receptor tyrosine kinase EphA2 is overexpressed in numerous epithelial-type carcinomas, with the greatest level of EphA2 expression observed in metastatic lesions. In the current study, we have assessed EphA2 expression in archived renal cell carcinoma (RCC) tissues as it relates to patient disease course. Using specific anti-EphA2 monoclonal antibody 208 and immunohistochemistry, we evaluated EphA2 protein expression levels in RCC specimens surgically resected from 34 patients (including 30 conventional clear-cell RCC, 3 papillary, and 1 chromophobic RCC cases) resulting in clinical cures. Regardless of histopathologic subtype, RCC lesions expressing higher levels of EphA2 tended to be of a higher grade (P < 0.05) and larger (P = 0.093), more-highly-vascularized tumors (P = 0.005). Perhaps most notable, the degree of EphA2 overexpression (versus normal matched autologous kidney tissue) seemed predictive of short-term (<1 year) versus longer-term (> or =1 year) disease-free interval (P < 0.001) and of overall survival (P < 0.001) among the RCC patients evaluated. These data suggest that EphA2 expression level may serve as a useful prognostic tool in the clinical management of patients who have been successfully treated with surgery, but who are at greater risk for accelerated disease recurrence and who have a poorer prognosis.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/diagnosis , Kidney Neoplasms/metabolism , Receptor, EphA2/biosynthesis , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/chemistry , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/surgery , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kidney/metabolism , Kidney Neoplasms/mortality , Kidney Neoplasms/surgery , Male , Middle Aged , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Prognosis , Treatment Outcome
5.
Clin Cancer Res ; 10(18 Pt 2): 6360S-6S, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15448031

ABSTRACT

PURPOSE: We reported that in renal cell carcinoma patients with active disease, T-cell reactions to the tumor-associated antigens MAGE-6 and EphA2 are highly skewed toward TH2-type cytokine responses [interleukin (IL) 5]. Herein, we determined whether tumor-derived products, including gangliosides isolated from renal cell carcinoma patients, participate in the down-regulation of type 1 T-cell responses. EXPERIMENTAL DESIGN: T cells from healthy volunteers or renal cell carcinoma patients were cultured in the presence and absence of supernatants derived from renal cell carcinoma explants or with gangliosides isolated from those tumor supernatants. T cells were stimulated or not with either autologous dendritic cells pulsed with superantigen (Staphylococcus enterotoxin B) or with phorbol 12-myristate 13-acetate and ionomycin and then were assessed for type 1 or type 2 responses (cytokine production and gene expression) and apoptosis. RESULTS: Tumor supernatants efficiently inhibited the TH1-type responses [interferon (IFN) gamma] of T cells stimulated with either S. enterotoxin B or phorbol 12-myristate 13-acetate and ionomycin but had no inhibitory effect on activated T-cell production of type 2 cytokines (IL-4, IL-5, and IL-10). Likewise, IFN-gamma mRNA and protein production were inhibited when T cells were cocultured with either renal cell carcinoma supernatant-derived gangliosides or a commercial source of purified GD1a. It was also determined that gangliosides impair type 1 responses by inducing apoptosis of activated T cells. CONCLUSIONS: We propose that renal cell carcinoma-derived tumor products such as gangliosides can induce a type 2 bias in antitumor immunity by initiating apoptosis in the IFN-gamma-producing type 1 effector cells. This represents a relevant mechanism by which renal cell carcinoma can inhibit protective antitumor immunity.


Subject(s)
Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , T-Lymphocytes/immunology , CD4 Lymphocyte Count , Cytokines/immunology , Gangliosides/immunology , Gangliosides/isolation & purification , Humans , Th1 Cells/immunology , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...