Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 178(2): 918-25, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202353

ABSTRACT

We previously reported the protection from myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) by the adoptive transfer of genetically modified embryonic stem cell-derived dendritic cells (ES-DC) presenting MOG peptide in the context of MHC class II molecules and simultaneously expressing TRAIL (ES-DC-TRAIL/MOG). In the present study, we found the severity of EAE induced by another myelin autoantigen, myelin basic protein, was also decreased after treatment with ES-DC-TRAIL/MOG. This preventive effect diminished, if the function of CD4(+)CD25(+) regulatory T cells (Treg) was abrogated by the injection of anti-CD25 mAb into mice before treatment with ES-DC-TRAIL/MOG. The adoptive transfer of CD4(+)CD25(+) T cells from ES-DC-TRAIL/MOG-treated mice protected the recipient mice from MOG- or myelin basic protein-induced EAE. The number of Foxp3(+) cells increased in the spinal cords of mice treated with ES-DC-TRAIL/MOG. In vitro experiments showed that TRAIL expressed in genetically modified ES-DC and also in LPS-stimulated splenic macrophages had a capacity to augment the proliferation of CD4(+)CD25(+) T cells. These results suggest that the prevention of EAE by treatment with ES-DC-TRAIL/MOG is mediated, at least in part, by MOG-reactive CD4(+)CD25(+) Treg propagated by ES-DC-TRAIL/MOG. For the treatment of organ-specific autoimmune diseases, induction of Treg reactive to the organ-specific autoantigens by the transfer of DC-presenting Ags and simultaneously overexpressing TRAIL therefore appears to be a promising strategy.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Associated Glycoprotein/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Cell Movement , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/metabolism , Genetic Engineering , Interleukin-2 Receptor alpha Subunit/metabolism , Lipopolysaccharides/pharmacology , Mice , Myelin Basic Protein/pharmacology , Myelin Proteins , Myelin-Associated Glycoprotein/genetics , Myelin-Associated Glycoprotein/therapeutic use , Myelin-Oligodendrocyte Glycoprotein , Spinal Cord/pathology , T-Lymphocytes, Regulatory/cytology , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/therapeutic use
2.
Cancer Sci ; 96(12): 889-96, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16367909

ABSTRACT

The close cooperation of both innate and acquired immunity is essential for the induction of truly effective antitumor immunity. We tested a strategy to enhance the cross-talk between NKT cells and conventional antigen-specific T cells with the use of alpha GalCer-loaded dendritic cells genetically engineered to express antigen plus chemokine, attracting both conventional T cells and NKT cells. DC genetically engineered to express a model antigen, OVA, along with SLC/CCL21 or monokine induced by IFN-gamma/CXCL9, had been generated using a method based on in vitro differentiation of DC from mouse ES cells. The ES-DC were loaded with alpha-GalCer and transferred to mice bearing MO4, an OVA-expressing melanoma, and their capacity to evoke antitumor immunity was evaluated. In vivo transfer of either OVA-expressing ES-DC, stimulating OVA-reactive T cells, or alpha-GalCer-loaded non-transfectant ES-DC, stimulating NKT cells, elicited a significant but limited degree of protection against the i.p. disseminated MO4. A more potent antitumor effect was observed when alpha-GalCer was loaded to ES-DC expressing OVA before in vivo transfer, and the effect was abrogated by the administration of anti-CD8, anti-NK1.1 or anti-asialo GM1 antibody. alpha-GalCer-loaded double transfectant ES-DC expressing SLC along with OVA induced the most potent antitumor immunity. Thus, alpha-GalCer-loaded ES-DC expressing tumor-associated antigen along with SLC can stimulate multiple subsets of effector cells to induce a potent therapeutic effect against peritoneally disseminated tumor cells. The present study suggests a novel way to use alpha-GalCer in immunotherapy for peritoneally


Subject(s)
Antineoplastic Agents/therapeutic use , Chemokines, CC/genetics , Dendritic Cells/immunology , Galactosylceramides/therapeutic use , Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , Cell Communication , Cell Line, Tumor , Cell Survival , Chemokine CCL21 , Dendritic Cells/drug effects , Dendritic Cells/physiology , Genetic Engineering , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Neoplasms/pathology , Ovalbumin/immunology , Recombinant Proteins , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
3.
Biochem Biophys Res Commun ; 335(1): 5-13, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16026756

ABSTRACT

Dendritic cells (DC) genetically modified to present tumor-associated antigen are a promising means for anti-cancer immunotherapy. By introducing expression vectors into ES cells and subsequently inducing differentiation to DC (ES-DC), we can generate transfectant DC expressing the transgenes. In the future clinical application of this technology, the unavailability of human ES cells genetically identical to the patients will be a problem. However, in most cases, semi-allogeneic ES cells sharing some of HLA alleles with recipients are expected to be available. In the present study, we observed that model tumor antigen (OVA)-expressing mouse ES-DC transferred into semi-allogeneic mice potently primed OVA-reactive CTL and elicited a significant protection against challenge with OVA-expressing tumor. Genetic modification of ES-DC to overexpress SPI-6, the specific inhibitor of granzyme B, further enhanced their capacity to prime antigen-specific CTL in semi-allogeneic recipient mice. These results suggest the potential of ES-DC as a novel means for anti-cancer immunotherapy.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Neoplasms/prevention & control , Stem Cells/cytology , Animals , Antigens/genetics , Antigens/immunology , Cell Line , Dendritic Cells/transplantation , Immunotherapy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Ovalbumin/genetics , Ovalbumin/immunology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Serpins/genetics , Serpins/metabolism , Stem Cell Transplantation , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...