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.
Breast Cancer Res ; 8(1): R5, 2006.
Article in English | MEDLINE | ID: mdl-16417648

ABSTRACT

INTRODUCTION: Dendritic cells (DCs) are key antigen-presenting cells that play an essential role in initiating and directing cellular and humoral immunity, including anti-tumor responses. Due to their critical role in cancer, induction of DC apoptosis may be one of the central mechanisms used by tumors to evade immune recognition. METHODS: Spontaneous apoptosis of blood DCs (lineage negative HLA-DR positive cells) was assessed in peripheral blood mononuclear cells (PBMCs) using Annexin-V and TUNEL assays immediately after blood collection. The role of tumor products was assessed by culturing cells with supernatants derived from breast cancer cell lines (TDSN) or PBMCs (PBMC-SN, as a control). The capacity of DC stimulation to prevent apoptosis was assessed by incubating DC with inflammatory cytokines, poly I:C, IL-12 or CD40 ligand (CD40L) prior to culture with TDSN. Apoptosis was determined by flow cytometry and microscopy, and Bcl-2 expression determined by intracellular staining. RESULTS: In this study we document the presence of a significantly higher proportion of apoptotic (Annexin-V+ and TUNEL+) blood DCs in patients with early stage breast cancer (stage I to II; n = 13) compared to healthy volunteers (n = 15). We examined the role of tumor products in this phenomenon and show that supernatants derived from breast cancer lines induce apoptosis of blood DCs in PBMC cultures. Aiming to identify factors that protect blood DC from apoptosis, we compared a range of clinically available maturation stimuli, including inflammatory cytokines (tumor necrosis factor-alpha, IL-1beta, IL-6 and prostaglandin (PG)E2 as a cytokine cocktail), synthetic double-stranded RNA (poly I:C) and soluble CD40 ligand. Although inflammatory cytokines and poly I:C induced robust phenotypic maturation, they failed to protect blood DCs from apoptosis. In contrast, CD40 stimulation induced strong antigen uptake, secretion of IL-12 and protected blood DCs from apoptosis through sustained expression of Bcl-2. Exogenous IL-12 provided similar Bcl-2 mediated protection, suggesting that CD40L effect is mediated, at least in part, through IL-12 secretion. CONCLUSION: Cumulatively, our results demonstrate spontaneous apoptosis of blood DCs in patients with breast cancer and confirm that ex vivo conditioning of blood DCs can protect them from tumor-induced apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/blood , Breast Neoplasms/physiopathology , Dendritic Cells , Adult , Aged , CD40 Antigens , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Interleukin-12/physiology , Middle Aged , Phenotype
2.
Neoplasia ; 7(12): 1112-22, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354594

ABSTRACT

Dendritic cell (DC) defects are an important component of immunosuppression in cancer. Here, we assessed whether cancer could affect circulating DC populations and its correlation with tumor progression. The blood DC compartment was evaluated in 136 patients with breast cancer, prostate cancer, and malignant glioma. Phenotypic, quantitative, and functional analyses were performed at various stages of disease. Patients had significantly fewer circulating myeloid (CD11c+) and plasmacytoid (CD123+) DC, and a concurrent accumulation of CD11c(-)CD123(-) immature cells that expressed high levels of HLA-DR+ immature cells (DR(+)IC). Although DR(+)IC exhibited a limited expression of markers ascribed to mature hematopoietic lineages, expression of HLA-DR, CD40, and CD86 suggested a role as antigen-presenting cells. Nevertheless, DR(+)IC had reduced capacity to capture antigens and elicited poor proliferation and interferon-gamma secretion by T-lymphocytes. Importantly, increased numbers of DR(+)IC correlated with disease status. Patients with metastatic breast cancer showed a larger number of DR(+)IC in the circulation than patients with local/nodal disease. Similarly, in patients with fully resected glioma, the proportion of DR(+)IC in the blood increased when evaluation indicated tumor recurrence. Reduction of blood DC correlating with accumulation of a population of immature cells with poor immunologic function may be associated with increased immunodeficiency observed in cancer.


Subject(s)
Breast Neoplasms/blood , Dendritic Cells/immunology , Glioma/blood , HLA-DR Antigens/metabolism , Prostatic Neoplasms/blood , Adenocarcinoma/blood , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antigen-Presenting Cells/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Glioma/pathology , Humans , Interferon-gamma/metabolism , Male , Middle Aged , Myeloid Cells/cytology , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes/metabolism
3.
Neoplasia ; 7(12): 1123-32, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354595

ABSTRACT

Dendritic cells (DC) have been implicated in the defective function of the immune system during cancer progression. We have demonstrated that patients with cancer have fewer myeloid (CD11c+) and plasmacytoid (CD123(hi)) DC and a concurrent accumulation of CD11c(-)CD123- immature cells expressing HLA-DR (DR(+)IC). Notably, DR(+)IC from cancer patients have a reduced capacity to stimulate allogeneic T-cells. DR(+)IC are also present in healthy donors, albeit in smaller numbers. In this study, we assessed whether DR(+)IC could have an impact on the immune response by comparing their function with DC counterparts. For this purpose, DR(+)IC and DC were purified and tested in the presentation of antigens through major histocompatibility complex (MHC) II and MHC-I molecules. DR(+)IC were less efficient than DC at presenting antigens to T-cells. DR(+)IC induced a limited activation of T-cells, eliciting poor T-helper (Th) 1 and preferentially inducing Th2-biased responses. Importantly, despite DR(+)IC's poor responsiveness to inflammatory factors, in samples from healthy volunteers and breast cancer patients, CD40 ligation induced phenotypic maturation and interleukin 12 secretion, in turn generating more efficient T-cell responses. These data underscore the importance of inefficient antigen presentation as a mechanism for tumor evasion and suggest an approach to improve the efficacy of DC-based immunotherapy for cancer.


Subject(s)
Antigen Presentation/physiology , Antigen-Presenting Cells/immunology , Breast Neoplasms/immunology , CD40 Antigens/pharmacology , Dendritic Cells/immunology , HLA-DR Antigens/metabolism , Adenocarcinoma/blood , Adenocarcinoma/immunology , Adenocarcinoma/secondary , Adult , Aged , Aged, 80 and over , Antigens, CD/immunology , Antigens, CD/metabolism , Breast Neoplasms/blood , Breast Neoplasms/pathology , Case-Control Studies , Cell Proliferation , Female , Humans , Interferon-gamma/metabolism , Middle Aged , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neoplasm Recurrence, Local/blood , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism
4.
Immunol Cell Biol ; 83(5): 451-61, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16174093

ABSTRACT

Several reports have demonstrated that tumours are not intrinsically resistant to the immune response. However, neoplasias commonly fail to initiate and maintain adequate immunity. A number of factors have been implicated in causing the failure, including aberrant antigen processing by tumour cells, anergy or deletion of T cells, and recruitment of inhibitory/regulatory cell types. It has been suggested that dysfunction of dendritic cells (DC) induced by the tumour is one of the critical mechanisms to escape immune surveillance. As a minor subset of leucocytes, DC are the key APC for initiating immune responses. DC are poised at the boundaries of the periphery and the inner tissues, sampling antigens of diverse origin. Following their encounter with antigen or danger signals, DC migrate to lymph nodes, where they activate effector cells essential for tumour clearance. Although the DC system is highly heterogeneous, the differentiation and function of DC populations is largely regulated by exogenous factors. Malignancies appear to exploit this by producing a plethora of immunosuppressive factors capable of affecting DC, thus exerting systemic effects on immune function. This review examines recent findings on the effects of tumour-derived factors inducing DC dysfunction and in particular examines the findings on alteration of DC differentiation, maturation and longevity as a potent mechanism for immune suppression in cancer.


Subject(s)
Dendritic Cells/immunology , Immunosuppression Therapy , Neoplasms/immunology , Neoplasms/pathology , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans
5.
Eur J Immunol ; 35(3): 681-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15688345

ABSTRACT

The circumsporozoite (CS) is the most abundant surface protein of the Plasmodium sporozoite, and is also present early in the liver stage of the infection. Following successful protective experiments in mice and monkeys, the synthetic 102-mer malaria vaccine polypeptide representing the C-terminal region of the CS of Plasmodium falciparum was tested in a clinical trial with healthy human volunteers. This vaccine induced strong CD8(+), CD4(+) T lymphocyte and antibody responses specific for the immunizing peptide. CD8(+) T lymphocyte responses elicited in HLA-A*0201 volunteers recognized two well-defined cytotoxic T lymphocyte epitopes within the CS. Here, we show that both monocyte-derived dendritic cells (Mo-DC) and Epstein-Barr virus-transformed B-lymphoblastoid cells (LCL) can present a cytotoxic T lymphocyte epitope contained within the 102-mer synthetic peptide. Paraformaldehyde and low temperature inhibited presentation, indicating that cellular processing was required. Using specific inhibitors, we show that, in both cell types, processing requires the proteasome and the MHC class I pathway, while the endosomal compartment appears to be critical only for the presentation by Mo-DC. Antigen uptake is associated with actin polymerization in both cell types. These in vitro results demonstrate the likely pathway of antigen presentation achieved via vaccination with this synthetic peptide.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class I/immunology , Malaria Vaccines/immunology , Peptides/immunology , Plasmodium falciparum/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Herpesvirus 4, Human , Humans , Protozoan Proteins/immunology , Vaccines, Synthetic
SELECTION OF CITATIONS
SEARCH DETAIL