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.
J Immunol ; 174(9): 5490-8, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843547

ABSTRACT

Breast and kidney-expressed chemokine (BRAK) CXCL14 is a new CXC chemokine with unknown function and receptor selectivity. The majority of head and neck squamous cell carcinoma (HNSCC) and some cervical squamous cell carcinoma do not express CXCL14 mRNA, as opposed to constitutive expression by normal oral squamous epithelium. In this study, we demonstrate that the loss of CXCL14 in HNSCC cells and at HNSCC primary tumor sites was correlated with low or no attraction of dendritic cell (DC) in vitro, and decreased infiltration of HNSCC mass by DC at the tumor site in vivo. Next, we found that recombinant human CXCL14 and CXCL14-positive HNSCC cell lines induced DC attraction in vitro, whereas CXCL14-negative HNSCC cells did not chemoattract DC. Transduction of CXCL14-negative HNSCC cell lines with the human CXCL14 gene resulted in stimulation of DC attraction in vitro and increased tumor infiltration by DC in vivo in chimeric animal models. Furthermore, evaluating the biologic effect of CXCL14 on DC, we demonstrated that the addition of recombinant human CXCL14 to DC cultures resulted in up-regulation of the expression of DC maturation markers, as well as enhanced proliferation of allogeneic T cells in MLR. Activation of DC with recombinant human CXCL14 was accompanied by up-regulation of NF-kappaB activity. These data suggest that CXCL14 is a potent chemoattractant and activator of DC and might be involved in DC homing in vivo.


Subject(s)
Chemokines, CXC/metabolism , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Down-Regulation/immunology , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Neoplasms, Squamous Cell/immunology , Neoplasms, Squamous Cell/pathology , Animals , Cell Line, Tumor , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Chemokines, CXC/pharmacology , Chemotaxis, Leukocyte/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dose-Response Relationship, Immunologic , Down-Regulation/genetics , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Male , Mice , Mice, SCID , Neoplasms, Squamous Cell/metabolism , Transduction, Genetic
2.
Diabetes ; 54(2): 424-33, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15677500

ABSTRACT

Immunosuppressive drugs are routinely used to provide tolerance after whole pancreas and islet cell transplantations. While they are essential in inhibiting graft rejection, little is known about their effect on islet function and beta-cell viability. In this study, we report that tacrolimus, sirolimus, and mycophenolic acid, when added to cultures of freshly isolated human islets, induce a downregulation of the synthesis and secretion of insulin. These functional changes are associated with decreased islet cell viability. All three agents induce a decrease of intracellular levels of Bcl-2 and Bcl-xL, with an increased level of Smac, indicating that they are capable of promoting a downregulation of anti-apoptotic factors and an accumulation of pro-apoptotic mediators. Transduction of islet cells with the anti-apoptotic gene XIAP prevents the negative effects of these drugs on the function and viability of islets. XIAP-infected cells show a higher expression of phospho-CREB (cAMP-responsive element binding protein) and a reduced level of Smac, resulting in a significant reduction of apoptotic cells and a preservation of the glucose-dependent secretion of insulin. In conclusion, the present study demonstrates that genetically modified human islets expressing XIAP are resistant to the negative effects of immunosuppressive drugs on insulin secretion and cell viability.


Subject(s)
Immunosuppressive Agents/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Proteins/genetics , Adenoviridae , Cell Culture Techniques/methods , Cell Death/drug effects , Cell Survival/drug effects , Gene Transfer Techniques , Humans , Immunosuppressive Agents/antagonists & inhibitors , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Proteins/metabolism , Recombinant Proteins/metabolism , Tacrolimus/pharmacology , Transfection , X-Linked Inhibitor of Apoptosis Protein
3.
J Gene Med ; 6(5): 537-44, 2004 May.
Article in English | MEDLINE | ID: mdl-15133764

ABSTRACT

BACKGROUND: Tumors develop mechanisms to escape recognition by the immune system. It has recently been demonstrated that tumors cause apoptotic death of key immune cells, including the major antigen-presenting cells, dendritic cells (DC). Elimination of DC from the tumor environment significantly diminishes development of specific immunologic responses. We have recently demonstrated that tumor-induced DC apoptosis could be prevented by overexpression of the anti-apoptotic molecule Bcl-x(L). The aim of this study was to identify extrinsic and intrinsic tumor-induced apoptotic pathways in DC by targeting different anti-apoptotic molecules, including FLIP, XIAP/hILP, dominant-negative procaspase-9 and HSP70. METHODS: Murine bone marrow derived DC were transduced with adenoviral vectors carrying different anti-apoptotic molecules and co-incubated with tumor cells in a Transwell system. Apoptosis of DC was assessed by Annexin V and PI staining. RESULTS: We have demonstrated that adenoviral infection of DC with genes encoding different anti-apoptotic molecules exhibits different degrees of resistance to melanoma-induced apoptosis. Furthermore, we have shown that anti-apoptotic molecules other than the Bcl-2 family of proteins are able to protect DC and prevent tumor-induced apoptosis in DC. CONCLUSIONS: The results show that tumor-induced apoptosis of DC is not limited to the mitochondrial pathway of cell death and open additional possibilities for targeted molecular protection of DC longevity in cancer. Therefore, effective protection of DC from tumor-induced apoptosis may significantly improve the efficacy of DC-based therapies for cancer.


Subject(s)
Apoptosis , Dendritic Cells/metabolism , Transduction, Genetic , Adenoviridae/genetics , Animals , Bone Marrow Cells/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 9 , Caspases/genetics , Caspases/metabolism , Cell Survival , Genetic Vectors , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Melanoma , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Tumor Cells, Cultured
4.
J Neuroimmunol ; 145(1-2): 55-67, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14644031

ABSTRACT

Development of tumors is regulated by tumor-derived neuroendocrine factors, including bombesin-like peptides (BLP). We have evaluated neuroendocrine regulation of dendritic cell (DC) maturation and function by both tumor-derived and purified bombesin (BOM), neuromedin B (NMB), gastrin-releasing peptide (GRP), and a BOM antagonist D-Phe-bombesin (DPB). BOM, NMB and GRP dose-dependently inhibited maturation of DC assessed as down-regulation of CD40, CD80 and CD86 expression on DC. BOM and GRP also inhibited interleukin-12 (IL-12) production by DC and their ability to activate T cells. DPB partly abrogated immunosuppressive effect of tumor cells on DC. These data are a first evidence for the role of BLP in the regulation of DC maturation and function, demonstrating that BLP inhibit DC maturation and longevity in the lung cancer microenvironment. This suggests a new mechanism of tumor escape and provides new targets for the immunopharmacological correction of immune effectors in cancer.


Subject(s)
Bombesin/pharmacology , Dendritic Cells/cytology , Dendritic Cells/immunology , Down-Regulation , Immunosuppressive Agents/pharmacology , Lung Neoplasms/metabolism , Neoplasm Proteins/pharmacology , Neurokinin B/analogs & derivatives , Antigen Presentation , Bombesin/metabolism , Cell Differentiation/immunology , Cell Line, Tumor , Cell Survival/immunology , Cells, Cultured , Dendritic Cells/metabolism , Dose-Response Relationship, Immunologic , Down-Regulation/immunology , Endocytosis/immunology , Gastrin-Releasing Peptide/metabolism , Humans , Interleukin-12/antagonists & inhibitors , Interleukin-12/biosynthesis , Lung Neoplasms/immunology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neurokinin B/biosynthesis , Neurokinin B/genetics , RNA, Messenger/biosynthesis , Receptors, Bombesin/biosynthesis , Receptors, Bombesin/genetics
5.
J Leukoc Biol ; 72(5): 1037-45, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429727

ABSTRACT

It has been recently demonstrated that dendritic cells (DC) coincubated with interleukin (IL)-15 express high levels of the Bcl-2 family of proteins and display an increased resistance to tumor-induced apoptotic death. Here, the phenotype, functions, and survival of human DC transduced with adenoviral vector encoding the human IL-15 gene were studied. The transduction of DC with the IL-15 gene resulted in a significant elevation of expression of CD83, CD86, and CD40 molecules, which was blocked by anti-IL-15 monoclonal antibodies. This effect was also accompanied by an increased production of IL-12 and stimulated ability of DC to induce T cell proliferation. Furthermore, transduction of DC with the IL-15 gene significantly increased their resistance to prostate cancer-induced apoptosis: Overexpression of IL-15 on DC blocked tumor-induced inhibition of Bcl-2 expression and prolonged DC survival after coincubation with tumor cells. Finally, overexpression of IL-15 in DC was associated with a higher level of expression of IL-15 receptor alpha chain mRNA. In summary, these results suggest that transduction of DC with the IL-15 gene markedly stimulates DC function and protects them from tumor-induced apoptosis.


Subject(s)
Dendritic Cells/immunology , Interleukin-15/genetics , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, Interleukin-2/biosynthesis , Up-Regulation , Antigens, CD/biosynthesis , Apoptosis , B7-2 Antigen , CD40 Antigens/biosynthesis , Cell Survival , Cells, Cultured , Dendritic Cells/cytology , Gene Expression Regulation , Humans , Immunoglobulins/biosynthesis , Interleukin-12/biosynthesis , Interleukin-15/physiology , Lymphocyte Activation , Male , Membrane Glycoproteins/biosynthesis , Prostatic Neoplasms/immunology , Proto-Oncogene Proteins c-bcl-2/physiology , RNA, Messenger/biosynthesis , Receptors, Interleukin-15 , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , T-Lymphocytes/immunology , Transduction, Genetic , Tumor Cells, Cultured , CD83 Antigen
6.
Int J Cancer ; 101(1): 61-8, 2002 Sep 01.
Article in English | MEDLINE | ID: mdl-12209589

ABSTRACT

As CD40 plays a key role in both antitumor immunity and DC maturation, we have studied the regulation of its expression during DC hematopoiesis (dendropoiesis) in vitro and in vivo in the tumor microenvironment. Using MC38 colon adenocarcinoma tumor models, we have demonstrated that DCs generated in vitro from bone marrow precursors obtained from tumor-bearers have significantly lower expression of CD40 molecules compared to DCs generated from tumor-free mice. Furthermore, CD40 expression on DCs isolated from the spleens of tumor-bearing mice was also significantly reduced, suggesting that tumor-derived factors inhibit CD40 expression on DCs during dendropoiesis both in vitro and in vivo. Interestingly, CD40 ligation on DCs generated from tumor-bearers did not result in inducible expression of IL-12 protein or IL-12 p40 mRNA. However, Staphylococcus aureus-induced IL-12 production by DCs was not altered in tumor-bearers, confirming that inhibition of IL-12 production by DCs generated in vitro from tumor-bearing mice was due to reduced expression of CD40 on DCs. We have also shown that MC38 tumor cells produce IL-10 and that exogenous IL-10 causes downregulation of CD40 expression on DCs. In addition, endogenous IL-10 produced by colon carcinoma cells inhibited CD40-dependent IL-12 production by DCs since tumor-induced inhibition of IL-12 production was abrogated by neutralizing anti-IL-10 antibody. Finally, systemic administration of FLT3L and/or CD40L reversed CD40 and IL-12 (p40) deficiency of DCs in tumor-bearing mice in vivo. These findings thus demonstrate that tumor-derived factors, including IL-10, inhibit CD40 expression on DCs and DC precursors and suppress their maturation and function.


Subject(s)
CD40 Antigens/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-10/pharmacology , Animals , CD40 Ligand/pharmacology , Cell Differentiation , Dendritic Cells/cytology , Down-Regulation/drug effects , Flow Cytometry , Interleukin-12/biosynthesis , Male , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...