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










Publication year range
1.
Eur J Immunol ; 38(12): 3411-25, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19009526

ABSTRACT

Since enteric microbial composition is a distinctive and stable individual trait, microbial heterogeneity may confer lifelong, non-genetic differences between individuals. Here we report that C57BL/6 mice bearing restricted flora microbiota, a distinct but diverse resident enteric microbial community, are numerically and functionally deficient in marginal zone (MZ) B cells. Surprisingly, MZ B-cell levels are minimally affected by germ-free conditions or null mutations of various TLR signaling molecules. In contrast, MZ B-cell depletion is exquisitely dependent on cytolytic CD8(+) T cells, and includes targeting of a cross-reactive microbial/endogenous MHC class 1B antigen. Thus, members of certain enteric microbial communities link with CD8(+) T cells as a previously unappreciated mechanism that shapes innate immunity dependent on innate-like B cells.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cell Shape , Animals , B-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Movement , Mice , Mice, Inbred C57BL , Proteins/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology
2.
Clin Cancer Res ; 11(21): 7674-82, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278387

ABSTRACT

PURPOSE: Cyclooxygenase-2 (COX-2), phosphorylated signal transducers and activators of transcription 3 (STAT3), and interleukin-6 (IL-6) are elevated in non-small cell lung cancer (NSCLC). These molecules affect numerous cellular pathways, including angiogenesis and apoptosis resistance, and, therefore, may act in concert in NSCLC. EXPERIMENTAL DESIGN: We examined IL-6 and phosphorylated STAT3 in COX-2-overexpressing [COX-2 sense-oriented (COX-2-S)] NSCLC cells and control cells. The effect of IL-6, STAT3, phosphatidylinositol 3-kinase, and mitogen-activated protein/extracellular signal-regulated kinase kinase on vascular endothelial growth factor (VEGF) production and apoptosis resistance was assessed in COX-2-overexpresing cells. RESULTS: We report that NSCLC cells overexpressing COX-2 (COX-2-S) have increased IL-6 and phosphorylated STAT3 expression compared with control cells. IL-6 induced expression of VEGF in NSCLC cells. Moreover, blocking IL-6, mitogen-activated protein/extracellular signal-regulated kinase kinase, or phosphatidylinositol 3-kinase decreased VEGF production in COX-2-S cells. The addition of IL-6 to NSCLC cells resulted in increased apoptosis resistance. Furthermore, the inhibition of STAT3 or IL-6 induced apoptosis and reduced survivin expression, a member of the inhibitor of apoptosis protein family in COX-2-S cells. CONCLUSIONS: Overall, these findings suggest a novel pathway in which COX-2 activates STAT3 by inducing IL-6 expression. This pathway could contribute to tumor formation by promoting survivin-dependent apoptosis resistance and VEGF production. These findings provide a rationale for the future development of STAT3, IL-6, and/or COX-2-targeted therapies for the treatment of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/physiology , Gene Expression Regulation, Neoplastic , Interleukin-6/metabolism , Lung Neoplasms/metabolism , STAT3 Transcription Factor/metabolism , Amino Acid Motifs , Apoptosis , Blotting, Western , Cell Differentiation , Cell Line, Tumor , Cell Survival , Cytokines/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
3.
Clin Lung Cancer ; 7(1): 30-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16098242

ABSTRACT

Clinical lung cancer is the ultimate event resulting from a series of genetic and epigenetic alterations in the respiratory epithelium at risk. According to the "field carcinogenesis" theory, these alterations can occur throughout the entire lung. In individuals with a genetic predisposition combined with a sufficient amount of procarcinogenic environmental influences, a few of these sites may eventually progress to malignancies. Recent advances in the understanding of tumor biology have identified new therapeutic targets for lung cancer chemoprevention, among which is cyclooxgygenase (COX)-2. Ample preclinical data suggest that the COX-2/prostaglandin E2 (PGE2) signaling pathway plays a pivotal role in conferring the malignant phenotype. Produced primarily by the action of COX on the free arachidonic acid liberated from membrane phospholipids, overproduction of PGE2, which is predominantly generated by upregulation of COX-2, is associated with a variety of mechanisms known to facilitate tumorigenesis. These mechanisms include abnormal expression of epithelial growth factors, epithelial and microvascular proliferation, resistance to apoptosis, and suppression of antitumor immunity. The lung is one of the major sites of PGE2 production, and previous studies have shown elevated PGE2 levels in bronchoalveolar lavage fluid of patients with bronchogenic carcinoma. In animal models, inhibition of COX-2 and PGE2 synthesis suppresses lung tumorigenesis. These preclinical data suggesting the antineoplastic effect of COX-2 inhibitors provide the basis for several ongoing pilot clinical trials to determine the feasibility of COX-2 inhibition in chemoprevention of bronchogenic carcinoma.


Subject(s)
Chemoprevention , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Dinoprostone/physiology , Lung Neoplasms/prevention & control , Apoptosis , Cell Transformation, Neoplastic , Clinical Trials as Topic , Dinoprostone/biosynthesis , Humans , Lung Neoplasms/physiopathology , Neovascularization, Pathologic , Signal Transduction , Up-Regulation
4.
Cancer Res ; 65(14): 6275-81, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16024629

ABSTRACT

Cyclooxygenase 2 (COX-2) overexpression is found in a wide variety of human cancers and is linked to all stages of tumorigenesis. Elevated tumor COX-2 expression is associated with increased angiogenesis, tumor invasion, suppression of host immunity and promotes tumor cell resistance to apoptosis. Previous reports have linked the COX-2 product prostaglandin E2 (PGE2) to the abnormal activation of the mitogen-activated protein kinase/Erk kinase pathway. Here we show that PGE2 is able to rapidly stimulate Erk phosphorylation in a subset of non-small cell lung cancer (NSCLC) cell lines. This effect is not evident in bronchial epithelial cells. In contrast to previous reports in colon cancer, we found that Erk activation as well as cellular proliferation induced by PGE2 was not inhibited by pretreatment of the cells with epidermal growth factor receptor (EGFR) inhibitors. Activation of the Erk pathway by PGE2 was also resistant to src kinase inhibitors but sensitive to the protein kinase C inhibition. PGE2 effects are mediated through four G protein-coupled receptors. Selective inhibition of EP receptors revealed the possible involvement of Ca2+-dependent signaling in PGE2-mediated activation of Erk. Our data indicate the presence of an EGFR-independent activation of the mitogen-activated protein kinase/Erk pathway by PGE2 in NSCLC cells. These findings provide evidence for the possible link between tumor COX-2 overexpression and elevated Erk-mediated cancer cell proliferation and migration. Importantly, these findings suggest that COX-2 overexpression may contribute to EGFR inhibitor resistance in NSCLC.


Subject(s)
Dinoprostone/pharmacology , ErbB Receptors/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Squamous Cell/enzymology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclooxygenase 2 , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , Humans , Lung Neoplasms/enzymology , MAP Kinase Signaling System/physiology , Membrane Proteins , Phosphorylation/drug effects , Prostaglandin-Endoperoxide Synthases/biosynthesis , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Signal Transduction/drug effects , src-Family Kinases/antagonists & inhibitors
5.
Biochem Biophys Res Commun ; 333(2): 470-5, 2005 Jul 29.
Article in English | MEDLINE | ID: mdl-15950941

ABSTRACT

COX-2 overexpression and subsequent PGE(2) production are frequently associated with non-small cell lung cancer and are implicated in tumor-mediated angiogenesis. Here, we report for the first time that IL-20 downregulates COX-2 and PGE(2) in human bronchial epithelial and endothelial cells. Flow cytometry analysis suggests that IL-20-dependent inhibition of COX-2/PGE(2) occurs through the IL-22R1/IL-20R2 dimers. In addition, we report that IL-20 exerts anti-angiogenic effects, inhibiting experimental angiogenesis. IL-20-mediated inhibition of PMA-induced angiogenesis occurs through the COX-2 regulatory pathway. Altogether our findings revealed that IL-20 is a negative modulator of COX-2/PGE(2) and inhibits angiogenesis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Carcinoma, Non-Small-Cell Lung/metabolism , Dinoprostone/metabolism , Interleukins/administration & dosage , Neovascularization, Pathologic/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Respiratory Mucosa/metabolism , Cell Line, Tumor , Cells, Cultured , Cyclooxygenase 2 , Cytokines/administration & dosage , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Membrane Proteins , Neovascularization, Pathologic/pathology , Respiratory Mucosa/drug effects
6.
Cancer Res ; 64(18): 6359-62, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15374938

ABSTRACT

Elevated tumor cyclooxygenase 2 (COX-2) expression is associated with increased angiogenesis, tumor invasion, and promotion of tumor cell resistance to apoptosis. In our previous studies using non-small cell lung cancer (NSCLC) cell lines constitutively expressing COX-2 cDNA in sense and antisense orientations, we demonstrated that constitutive overexpression of COX-2 leads to stabilization of the inhibitor of apoptosis protein survivin resulting in the elevated apoptosis resistance of COX-2-overexpressing cells. Genetic or pharmacologic suppression of COX-2 activity increased proteasomal degradation of survivin and cellular response to apoptosis induction. Our data show that expression of survivin in non-small cell lung cancer cells can be significantly down-regulated by RNA interference. Whereas COX-2-overexpressing NSCLC cells have significantly higher apoptosis resistance than the parental cells, inhibition of survivin expression by small interfering RNA decreases apoptosis resistance to the level of the parental non-small cell lung cancer. We conclude that COX-2-dependent expression of survivin is critical for apoptosis resistance in non-small cell lung cancer.


Subject(s)
Apoptosis/physiology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Isoenzymes/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/biosynthesis , Prostaglandin-Endoperoxide Synthases/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cyclooxygenase 2 , Humans , Inhibitor of Apoptosis Proteins , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Membrane Proteins , Microtubule-Associated Proteins/genetics , Neoplasm Proteins , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Survivin , Transfection
7.
Drug Resist Updat ; 7(3): 169-84, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15296859

ABSTRACT

Lung cancer is the leading cause of cancer death in the United States. Although the low 5-year survival rate (under 15%) has changed minimally in the last 25 years, new agents and combinations of agents that target tumor proliferation, invasion, and survival may lead to improvement in patient outcomes. There is evidence that cyclooxygenase-2 (COX-2) is overexpressed in lung cancer and promotes tumor proliferation, invasion, angiogenesis, and resistance to apoptosis. COX-2 inhibitors have been found to inhibit tumor growth in animal models and have demonstrated responses when combined with conventional therapy in phase II clinical trials. Further understanding of the mechanisms involved in COX-2-mediated tumorigenesis and its interaction with other molecules in lung cancer may lead to improved therapeutic strategies for this disease. In addition, delineation of how COX-2-dependent genes modulate the malignant phenotype will provide novel insights in lung cancer pathogenesis.


Subject(s)
Isoenzymes/physiology , Lung Neoplasms/etiology , Prostaglandin-Endoperoxide Synthases/physiology , Antigen-Presenting Cells/physiology , Apoptosis , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/therapeutic use , Cytokines/physiology , ErbB Receptors/antagonists & inhibitors , Humans , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Membrane Proteins , Neovascularization, Pathologic/etiology
8.
Int Immunol ; 15(11): 1359-67, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14565934

ABSTRACT

Heterotrimeric Gi signaling regulates immune homeostasis, since autoimmunity occurs upon disruption of this pathway. However, the role of the lymphocyte-expressed Galphai subunits (Galphai2 and 3) on T cell activation and cytokine production is poorly understood. To examine this role, we studied T lymphocytes from mice deficient in the Galphai2 or Galphai3 subunits. Galphai2(-/-) but not Galphai3(-/-) splenocytes were hyper-responsive for IFN-gamma and IL-4 production following activation through the TCR. Galphai2(-/-) T cells had a relaxed costimulatory requirement for IL-2 secretion and proliferation compared to wild-type cells. Purified naïve Galphai2(-/-) T cells produced more IL-2 than naïve wild-type T cells following TCR activation, indicating that the hyper-responsive cytokine profile was not due to the expanded Galphai2(-/-) memory T cells, but involved an intrinsic T cell alteration. Cytokine hyper-responsiveness was not seen when purified Galphai2(-/-) T cells were stimulated with phorbol myristic acetate/ionomycin, localizing the alteration to a proximal TCR-specific signaling pathway. Galphai2(-/-) CD4(+) T cells were distinguished from wild-type or Galphai3(-/-) T cells by a globally augmented TCR-induced calcium response. These findings indicate that Galphai2(-/-) mice have an intrinsic CD4(+) T cell abnormality in TCR signaling which may be one cause of augmented T cell effector function and Galphai2(-/-) autoimmune susceptibility.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Proto-Oncogene Proteins/physiology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Differentiation , Cytokines/immunology , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Interleukin-2/immunology , Lymphocyte Activation , Mice , Proto-Oncogene Proteins/genetics , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
9.
J Immunol ; 170(4): 1707-15, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574334

ABSTRACT

Null mutation of the Galphai2 trimeric G protein results in a discrete and profound mucosal disorder, including inflammatory bowel disease (IBD), attenuation of IL-10 expression, and immune function polarized to Th1 activity. Genetic and adoptive transfer experiments have established a role for B cells and IL-10 in mucosal immunologic homeostasis and IBD resistance. In this study, we addressed the hypothesis that Galphai2 is required for the development of IL-10-producing B cells. Galphai2(-/-) mice were reduced in the relative abundance of marginal zone (MZ), transitional type 2 (T2), and B-1a B cells and significantly increased in follicular mature and B-1b B cells. Reconstitution of RAG2(-/-) mice with Galphai2(-/-) bone marrow induced an IBD-like colitis and a deficiency in absolute numbers of MZ, T2, and B-1 B cells. Thus, the Galphai2(-/-) genotype in colitis susceptibility and B cell development involved a cis effect within the hemopoietic compartment. In vitro, the B cell population of Galphai2(-/-) mice was functionally deficient in LPS-induced proliferation and IL-10 production, consistent with the exclusive capacity of T2 and MZ cell subpopulations for LPS responsiveness. In vivo, Galphai2(-/-) mice were selectively impaired for the IgM response to T-independent type II, consistent with the relative depletion of MZ and peritoneal B-1 subpopulations. Collectively, these results reveal a selective role for Galphai2 in MZ and B-1 B cell development. Disorders of this Galphai2-dependent process in B cell development may represent a mechanism for IBD susceptibility.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Genes/physiology , Proto-Oncogene Proteins/genetics , Animals , Antigens, T-Independent/administration & dosage , Antigens, T-Independent/immunology , B-Lymphocyte Subsets/pathology , Bone Marrow Transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Colitis/genetics , Colitis/immunology , Colitis/pathology , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Genes/immunology , Genetic Predisposition to Disease , Immunophenotyping , Injections, Intraperitoneal , Lymphocyte Count , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/pathology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/physiology , Receptors, Antigen, B-Cell/physiology , Spleen/immunology , Spleen/pathology
10.
Infect Immun ; 70(12): 6567-75, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12438326

ABSTRACT

Commensal bacteria have emerged as an important disease factor in human Crohn's disease (CD) and murine inflammatory bowel disease (IBD) models. We recently isolated I2, a novel gene segment of microbial origin that is associated with human CD and that encodes a T-cell superantigen. To identify the I2 microorganism, BLAST analysis was used to identify a microbial homologue, PA2885, a novel open reading frame (ORF) in the Pseudomonas aeruginosa genome. PCR and Southern analysis identified Pseudomonas fluorescens as the originating species of I2, with homologues detectable in 3 of 13 other Pseudomonas species. Genomic cloning disclosed a locus containing the full-length I2 gene (pfiT) and three other orthologous genes, including a homologue of the pbrA/pvdS iron response gene. CD4(+) T-cell responses to recombinant proteins were potent for I2 and pfiT, but modest for PA2885. pfiT has several features of a virulence factor: association with an iron-response locus, restricted species distribution, and T-cell superantigen bioactivity. These findings suggest roles for pfiT and P. fluorescens in the pathogenesis of Crohn's disease.


Subject(s)
Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , Crohn Disease/immunology , Pseudomonas fluorescens/immunology , Superantigens/genetics , Amino Acid Sequence , Animals , Bacterial Proteins/immunology , Cloning, Molecular , Crohn Disease/physiopathology , Feces/microbiology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pseudomonas fluorescens/genetics , Pseudomonas fluorescens/metabolism , Sequence Alignment , Sequence Analysis, DNA , Superantigens/chemistry , Superantigens/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...