Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Transl Med ; 7: 4, 2009 Jan 12.
Article in English | MEDLINE | ID: mdl-19138409

ABSTRACT

BACKGROUND: Recently, nanometer sized vesicles (termed exosomes) have been described as a component of urine. Such vesicles may be a useful non-invasive source of markers in renal disease. Their utility as a source of markers in urological cancer remains unstudied. Our aim in this study was to investigate the feasibility and value of analysing urinary exosomes in prostate cancer patients undergoing standard therapy. METHODS: Ten patients (with locally advanced PCa) provided spot urine specimens at three time points during standard therapy. Patients received 3-6 months neoadjuvant androgen deprivation therapy prior to radical radiotherapy, comprising a single phase delivering 55 Gy in 20 fractions to the prostate and 44 Gy in 20 fractions to the pelvic nodes. Patients were continued on adjuvant ADT according to clinical need. Exosomes were purified, and the phenotype compared to exosomes isolated from the prostate cancer cell line LNcaP. A control group of 10 healthy donors was included. Serum PSA was used as a surrogate treatment response marker. Exosomes present in urine were quantified, and expression of prostate markers (PSA and PSMA) and tumour-associated marker 5T4 was examined. RESULTS: The quantity and quality of exosomes present in urine was highly variable, even though we handled all materials freshly and used methods optimized for obtaining highly pure exosomes. There was approx 2-fold decrease in urinary exosome content following 12 weeks ADT, but this was not sustained during radiotherapy. Nevertheless, PSA and PSMA were present in 20 of 24 PCa specimens, and not detected in healthy donor specimens. There was a clear treatment-related decrease in exosomal prostate markers in 1 (of 8) patient. CONCLUSION: Evaluating urinary-exosomes remains difficult, given the variability of exosomes in urine specimens. Nevertheless, this approach holds promise as a non-invasive source of multiple markers of malignancy that could provide clinically useful information.


Subject(s)
Biomarkers, Tumor , Exosomes , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/urine , Adult , Aged , Antigens, Neoplasm/analysis , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/urine , Cell Line, Tumor , Exosomes/chemistry , Exosomes/metabolism , Humans , Male , Middle Aged , Phenotype , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Treatment Outcome
2.
J Immunol ; 180(11): 7249-58, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490724

ABSTRACT

NKG2D is an activating receptor for NK, NKT, CD8(+), and gammadelta(+) T cells, whose aberrant loss in cancer is a key mechanism of immune evasion. Soluble NKG2D ligands and growth factors, such as TGFbeta1 emanating from tumors, are mechanisms for down-regulating NKG2D expression. Cancers thereby impair the capacity of lymphocytes to recognize and destroy them. In this study, we show that exosomes derived from cancer cells express ligands for NKG2D and express TGFbeta1, and we investigate the impact of such exosomes on CD8(+) T and NK cell NKG2D expression and on NKG2D-dependent functions. Exosomes produced by various cancer cell lines in vitro, or isolated from pleural effusions of mesothelioma patients triggered down-regulation of surface NKG2D expression by NK cells and CD8(+) T cells. This decrease was rapid, sustained, and resulted from direct interactions between exosomes and NK cells or CD8(+) T cells. Other markers (CD4, CD8, CD56, CD16, CD94, or CD69) remained unchanged, indicating the selectivity and nonactivatory nature of the response. Exosomal NKG2D ligands were partially responsible for this effect, as down-modulation of NKG2D was slightly attenuated in the presence of MICA-specific Ab. In contrast, TGFbeta1-neutralizing Ab strongly abrogated NKG2D down-modulation, suggesting exosomally expressed TGFbeta as the principal mechanism. Lymphocyte effector function was impaired by pretreatment with tumor exosomes, as these cells exhibited poor NKG2D-dependent production of IFN-gamma and poor NKG2D-dependent killing function. This hyporesponsiveness was evident even in the presence of IL-15, a strong inducer of NKG2D. Our data show that NKG2D is a likely physiological target for exosome-mediated immune evasion in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Killer Cells, Natural/immunology , Neoplasms/metabolism , Receptors, Immunologic/metabolism , Secretory Vesicles/metabolism , Transforming Growth Factor beta1/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation , Mesothelioma/immunology , Mesothelioma/metabolism , NK Cell Lectin-Like Receptor Subfamily K , Neoplasms/immunology , Receptors, Immunologic/immunology , Receptors, Natural Killer Cell , Secretory Vesicles/immunology , Transforming Growth Factor beta1/immunology
3.
Cancer Res ; 67(15): 7458-66, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17671216

ABSTRACT

Exosomes are nanometer-sized vesicles, secreted by normal and neoplastic cells. The outcome following interaction between the cellular immune system and cancer-derived exosomes is not well understood. Interleukin-2 (IL-2) is a key factor supporting expansion and differentiation of CTL and natural killer (NK) cells but can also support regulatory T cells and their suppressive functions. Our study examined whether tumor-derived exosomes could modify lymphocyte IL-2 responses. Proliferation of healthy donor peripheral blood lymphocytes in response to IL-2 was inhibited by tumor exosomes. In unfractionated lymphocytes, this effect was seen in all cell subsets. Separating CD4(+) T cells, CD8(+) T cells, and NK cells revealed that CD8(+) T-cell proliferation was not inhibited in the absence of CD4(+) T cells and that NK cell proliferation was only slightly impaired. Other exosome effects included selective impairment of IL-2-mediated CD25 up-regulation, affecting all but the CD3(+)CD8(-) T-cell subset. IL-2-induced Foxp3 expression by CD4(+)CD25(+) cells was not inhibited by tumor exosomes, and the suppressive function of CD4(+)CD25(+) T cells was enhanced by exosomes. In contrast, exosomes directly inhibited NK cell killing function in a T-cell-independent manner. Analysis of tumor exosomes revealed membrane-associated transforming growth factor beta(1) (TGFbeta(1)), which contributed to the antiproliferative effects, shown by using neutralizing TGFbeta(1)-specific antibody. The data show an exosome-mediated mechanism of skewing IL-2 responsiveness in favor of regulatory T cells and away from cytotoxic cells. This coordinated "double hit" to cellular immunity strongly implicates the role of exosomes in tumor immune evasion.


Subject(s)
Exocytosis/immunology , Interleukin-2/pharmacology , Mesothelioma/immunology , Pleural Neoplasms/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes/drug effects , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-10/metabolism , Male , Mesothelioma/drug therapy , Mesothelioma/metabolism , Pleural Neoplasms/drug therapy , Pleural Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
4.
Eur J Immunol ; 33(2): 522-31, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12645951

ABSTRACT

Exosomes are secreted nanometer-sized vesicles derived from antigen-presenting cells, which have attracted recent interest as they likely play important roles in immune regulation, and their use as cell-free tools for immunotherapy has been proposed. Liposomes used clinically as transport vehicles can activate the complement system, leading to their rapid degradation and significant inflammatory toxicity. The use of isolated exosomes in therapy, therefore, may also elicit complement activation, reducing their potential efficacy. We have examined the expression and functional roles of the membrane regulators of complement (CD46, CD55 and CD59) on antigen-presenting cell-derived exosomes. Exosomes express the glycosylphosphatidylinositol (GPI)-anchored regulators CD55 and CD59, but not the transmembrane protein CD46. Antibody blocking of CD55 in the presence of sensitizing antibody (w6/32) and human serum resulted in increased C3b deposition and significantly increased exosome lysis. Blockade of CD59 also resulted in significant lysis, while blocking both CD55 and CD59 increased lysis still further. We conclude that exosomes express GPI-anchored complement regulators in order to permit their survival in the extracellular environment.


Subject(s)
Antigen Presentation/immunology , CD55 Antigens/immunology , CD59 Antigens/immunology , Complement System Proteins/immunology , Dendritic Cells/immunology , Organelles/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD/analysis , Cell Differentiation , Cells, Cultured/immunology , Complement Activation , Complement C3b/metabolism , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Humans , Membrane Cofactor Protein , Membrane Glycoproteins/analysis , Monocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...