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 Invest Dermatol ; 136(10): 1981-1989, 2016 10.
Article in English | MEDLINE | ID: mdl-27375111

ABSTRACT

Sterile alpha motif (SAM) and histidine-aspartic (HD) domains protein 1 (SAMHD1) was previously identified as a critical post-entry restriction factor to HIV-1 infection in myeloid dendritic cells. Here we show that SAMHD1 is also expressed in epidermis-isolated Langerhans cells (LC), but degradation of SAMHD1 does not rescue HIV-1 or vesicular stomatitis virus G-pseudotyped lentivectors infection in LC. Strikingly, using Langerhans cells model systems (mutz-3-derived LC, monocyte-derived LC [MDLC], and freshly isolated epidermal LC), we characterize previously unreported post-entry restriction activity to HIV-1 in these cells, which acts at HIV-1 reverse transcription, but remains independent of restriction factors SAMHD1 and myxovirus resistance 2 (MX2). We demonstrate that transforming growth factor-ß signaling confers this potent HIV-1 restriction in MDLC during their differentiation and blocking of mothers against decapentaplegic homolog 2 (SMAD2) signaling in MDLC restores cells' infectivity. Interestingly, maturation of MDLC with a toll-like receptor 2 agonist or transforming growth factor-α significantly increases cells' susceptibility to HIV-1 infection, which may explain why HIV-1 acquisition is increased during coinfection with sexually transmitted infections. In conclusion, we report a SAMHD1-independent post-entry restriction in MDLC and LC isolated from epidermis, which inhibits HIV-1 replication. A better understanding of HIV-1 restriction and propagation from LC to CD4(+) T cells may help in the development of new microbicides or vaccines to curb HIV-1 infection at its earliest stages during mucosal transmission.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Langerhans Cells/virology , Monomeric GTP-Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Cell Line , Humans , Monocytes/metabolism , Myxovirus Resistance Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Transforming Growth Factor alpha/metabolism , Virus Replication/physiology
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.
J Immunol Methods ; 335(1-2): 98-105, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18423480

ABSTRACT

Exosomes are nanometer-sized vesicles, secreted from most cell types, with documented immune-modulatory functions. Exosomes can be purified from cultured cells but to do so effectively, requires maintenance of cells at high density in order to obtain sufficient accumulation of exosomes in the culture medium, prior to purification. Whilst high density cultures can be achieved with cells in suspension, this remains difficult with adherent cells, resulting in low quantity of exosomes for subsequent study. We have used the Integra CELLine culture system, originally designed for hybridoma cultures, to achieve a significant increase in obtainable exosomes from adherent and non-adherent tumour cells. Traditional cultures of mesothelioma cells (cultured in 75 cm(2) flasks) gave an average yield of 0.78 microg+/-0.14 microg exosome/ml of conditioned medium. The CELLine Adhere 1000 (CLAD1000) flask, housing the same cell line, increased exosome yield approximately 12 fold to 10.06 microg+/-0.97 microg/ml. The morphology, phenotype and immune function of these exosomes were compared, and found to be identical in all respects. Similarly an 8 fold increase in exosome production was obtained from NKL cells (a suspension cell line) using a CELLine 1000 (CL1000) flask. The CELLine system also incurred ~5.5 fold less cost and reduced labour for cell maintenance. This simple culture system is a cost effective, useful method for significantly increasing the quantity of exosomes available from cultured cells, without detrimental effects. This tool should prove advantageous in future studies of exosome-immune modulation in cancer and other settings.


Subject(s)
Cell Culture Techniques , Killer Cells, Natural/metabolism , Leukemia, Large Granular Lymphocytic/metabolism , Mesothelioma/metabolism , Pleural Neoplasms/metabolism , Secretory Vesicles/metabolism , Cell Adhesion , Cell Line , Cell Proliferation , Culture Media, Conditioned/metabolism , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Leukemia, Large Granular Lymphocytic/immunology , Leukemia, Large Granular Lymphocytic/pathology , Mesothelioma/immunology , Mesothelioma/pathology , Microscopy, Electron, Transmission , Phenotype , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Secretory Vesicles/immunology , Secretory Vesicles/ultrastructure , Time Factors , Tumor Cells, Cultured
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...