Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Immunol ; 181(11): 7843-52, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017974

ABSTRACT

Many human solid tumors express MHC class II (MHC-II) molecules, and proteins normally localized to melanosomes give rise to MHC-II-restricted epitopes in melanoma. However, the pathways by which this response occurs have not been defined. We analyzed the processing of one such epitope, gp100(44-59), derived from gp100/Pmel17. In melanomas that have down-regulated components of the melanosomal pathway, but constitutively express HLA-DR*0401, the majority of gp100 is sorted to LAMP-1(high)/MHC-II(+) late endosomes. Using mutant gp100 molecules with altered intracellular trafficking, we demonstrate that endosomal localization is necessary for gp100(44-59) presentation. By depletion of the AP-2 adaptor protein using small interfering RNA, we demonstrate that gp100 protein internalized from the plasma membrane to such endosomes is a major source for gp100(44-59) epitope production. The gp100 trapped in early endosomes gives rise to epitopes that are indistinguishable from those produced in late endosomes but their production is less sensitive to inhibition of lysosomal proteases. In melanomas containing melanosomes, gp100 is underrepresented in late endosomes, and accumulates in stage II melanosomes devoid of MHC-II molecules. The gp100(44-59) presentation is dramatically reduced, and processing occurs entirely in early endosomes or stage I melanosomes. This occurrence suggests that melanosomes are inefficient Ag-processing compartments. Thus, melanoma de-differentiation may be accompanied by increased presentation of MHC-II restricted epitopes from gp100 and other melanosome-localized proteins, leading to enhanced immune recognition.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Endosomes/immunology , Epitopes, T-Lymphocyte/immunology , HLA-DR Antigens/immunology , Melanoma/immunology , Melanosomes/immunology , Membrane Glycoproteins/immunology , Adaptor Protein Complex 2/immunology , Cell Adhesion Molecules, Neuronal/immunology , Cell Dedifferentiation/immunology , Cell Line, Tumor , GPI-Linked Proteins , Humans , Protein Transport/immunology , RNA, Small Interfering/immunology , gp100 Melanoma Antigen
2.
J Immunol ; 177(8): 5440-50, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17015730

ABSTRACT

Although multiple components of the class I MHC processing pathway have been elucidated, the participation of nonproteasomal cytosolic enzymes has been largely unexplored. In this study, we provide evidence for multiple cytosolic mechanisms in the generation of an HLA-A*0201-associated epitope from tyrosinase. This epitope is presented in two isoforms containing either Asn or Asp, depending on the structure of the tyrosinase precursor. We show that deamidation of Asn to Asp is dependent on glycosylation in the endoplasmic reticulum (ER), and subsequent deglycosylation by peptide-N-glycanase in the cytosol. Epitope precursors with N-terminal extensions undergo a similar process. This is linked to an inability of ER aminopeptidase 1 to efficiently remove N-terminal residues, necessitating processing by nonproteasomal peptidases in the cytosol. Our work demonstrates that processing of this tyrosinase epitope involves recycling between the ER and cytosol, and an obligatory interplay between enzymes involved in proteolysis and glycosylation/deglycosylation located in both compartments.


Subject(s)
Aminopeptidases/metabolism , Antigen Presentation , Cytosol/enzymology , Endoplasmic Reticulum/metabolism , Epitopes/immunology , Histocompatibility Antigens Class I/immunology , Monophenol Monooxygenase/immunology , Peptide Hydrolases/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Endoplasmic Reticulum/enzymology , Glycosylation , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Minor Histocompatibility Antigens
3.
Cancer Immunol Immunother ; 53(12): 1068-84, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15696607

ABSTRACT

PURPOSE: CD227 (MUC1), a membrane-associated glycoprotein expressed by many types of ductal epithelia, including pancreas, breast, lung, and gastrointestinal tract, is overexpressed and aberrantly glycosylated by malignant cells. We sought to define epitopes on MUC1 recognized by the different cell-mediated immune responses by an in vivo assay. Epitopes identified by this assay were evaluated for efficacy to protect mice transgenic for human MUC1 (MUC1.Tg) against MUC1-expressing tumor growth. METHODS: We investigated contributions of the tandem repeat (TR) and the cytoplasmic tail (CT) of MUC1 to the MUC1-specific immunological rejection of tumor cells. MUC1 cDNA constructs, in which the TR region was deleted or the CT was truncated, were transfected into two different murine tumor cell lines (B16 and Panc02), which were used to challenge mice and evaluate immunological rejection of the tumors. We used tumor rejection in vivo to define epitopes on the TR and CT of MUC1 recognized by T cell-mediated immune responses in a preclinical murine model. RESULTS: Our findings demonstrated that the TR and a portion of the MUC1 CT contributed to CD4+ T cell rejection of MUC1-expressing B16 tumor cells, but not rejection of MUC1-expressing Panc02 tumor cells. A separate epitope in the CT of MUC1 was necessary for CD8+ T cell rejection of Panc02 tumor cells. Based on these studies, we sought to evaluate the efficacy of immunizing mice transgenic for (and immunologically tolerant to) human MUC1 with peptides derived from the amino acid sequence of the CT of MUC1. Results showed that survival can be significantly prolonged in vaccinated MUC1.Tg mice challenged with MUC1-expressing tumor cells, without induction of autoimmune responses. CONCLUSIONS: These studies demonstrated that MUC1 peptides may be utilized as an effective anticancer immunotherapeutic, and confirmed the importance of immunogenic epitopes outside of the TR.


Subject(s)
Cancer Vaccines/immunology , Melanoma, Experimental/therapy , Mucin-1/immunology , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , Humans , Immunization , Male , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Mucin-1/chemistry , Mucin-1/genetics , Tandem Repeat Sequences
4.
Cancer Immunol Immunother ; 51(6): 327-40, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12111121

ABSTRACT

Previous studies have indicated that different effector cells are required to eliminate MUC1-expressing tumors derived from different organ sites and that different vaccine strategies may be necessary to generate these two different MUC1-specific immune responses. In this study, we characterized molecular components that are required to produce immune responses that eliminate Panc02.MUC1 tumors in vivo by utilizing mice genetically deficient in molecules related to immunity. A parallel study has been reported for a B16.MUC1 tumor model. We confirmed that a CD8(+) effector cell was required to eliminate MUC1-expressing Panc02 tumors, and demonstrated that T cells expressing TCR-alpha/beta and co-stimulation through CD28 and CD40:CD40L interactions played critical roles during the initiation of the anti-Panc02.MUC1 immune response. TCR-alpha/beta(+) cells were required to eliminate Panc02.MUC1 tumors, while TCR-gamma/delta(+) cells played a suppressive non-MUC1-specific role in anti-Panc02 tumor immunity. Type 1 cytokine interferon-gamma (IFN-gamma), but not interleukin-12 (IL-12), was essential for eliminating MUC1-expressing tumors, while neither IL-4 nor IL-10 (type 2 cytokines) were required for tumor rejection. In vitro studies demonstrated that IFN-gamma upregulated MHC class I, but not MHC class II, on Panc02.MUC1 tumor cells. Surprisingly, both perforin and FasL played unique roles during the effector phase of immunity to Panc02.MUC1, while lymphotoxin-alpha, but not TNFR-1, was required for immunity against Panc02.MUC1 tumors. The findings presented here and in parallel studies of B16.MUC1 immunity clearly demonstrate that different effector cells and cytolytic mechanisms are required to eliminate MUC1-expressing tumors derived from different organ sites, and provide insight into the immune components required to eliminate tumors expressing the same antigen but derived from different tissues.


Subject(s)
Antigens, Neoplasm/analysis , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Carcinoma/immunology , Mucin-1/analysis , Pancreatic Neoplasms/immunology , Animals , Antigens, CD/genetics , Antigens, CD/physiology , CD28 Antigens/genetics , CD28 Antigens/immunology , CD4 Antigens/genetics , CD40 Antigens/genetics , CD40 Antigens/immunology , CD40 Ligand/genetics , CD40 Ligand/immunology , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/chemistry , Cytotoxicity, Immunologic , Fas Ligand Protein , Graft Rejection , Humans , Immune Tolerance , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukins/deficiency , Interleukins/genetics , Interleukins/physiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Mice, Transgenic , Mucin-1/genetics , Neoplasm Transplantation , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/physiology , Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology
5.
Int Immunol ; 14(8): 873-82, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12147624

ABSTRACT

MUC1 was first defined as a tumor antigen in the late 1980s, yet little is known about the types of immune responses that mediate rejection of MUC1(+) tumors in vivo. MUC1-specific antibodies, T(h) cells and cytotoxic T cells can be detected in patients with different adenocarcinomas, yet these tumors usually progress. Thus, there is a need to better understand the in vivo mechanisms of antigen-specific tumor rejection. To characterize the nature of MUC1-specific immune responses in vivo, rejection of a MUC1-expressing melanoma tumor line (B16.MUC1) was evaluated in mice lacking specific T cell subsets, cytokines, co-stimulatory molecules or molecular effectors of cytolytic pathways. Results demonstrated that rejection of the B16.MUC1 tumor cell line was primarily mediated by CD4(+) T cells, and required Fas ligand, lymphotoxin-alpha, CD40, CD40 ligand and CD28, but not perforin, gammadelta T cells, IL-4, IL-10, IL-12 or tumor necrosis factor receptor-1. Depletion of NK cells demonstrated that NK cells might also contribute to MUC1 immunity in the B16.MUC1 tumor model. These results demonstrated that the immune response generated against MUC1 does not fit the type 1 or 2 model described for many immune responses. Additionally, multiple cytolytic mechanisms are required for B16.MUC1 rejection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Melanoma, Experimental/immunology , Mucin-1/immunology , Animals , Cytokines/biosynthesis , Cytokines/deficiency , Cytokines/genetics , Cytotoxicity, Immunologic , Female , Graft Rejection , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Killer Cells, Natural/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Mucin-1/genetics , Neoplasm Transplantation , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...