Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Leukoc Biol ; 103(3): 395-407, 2018 03.
Article in English | MEDLINE | ID: mdl-29345342

ABSTRACT

Obesity is a risk factor for cancer incidence and cancer mortality. The association of obesity and cancer is attributed to multiple factors, but the tightest linkage is with the chronic, low-grade inflammation that accompanies obesity. Myeloid-derived suppressor cells (MDSC) are known facilitators of cancer progression that act by suppressing the activation and function of tumor-reactive T cells. Because MDSC quantity and function are driven by chronic inflammation, we hypothesized that MDSC may accumulate in obese individuals and facilitate tumor growth by suppressing antitumor immunity. To test this hypothesis, tumor-bearing mice on a high fat or low fat diet (HFD or LFD) were assessed for tumor progression and the metabolic dysfunction associated with obesity. HFD enhanced the accumulation of MDSC, and the resulting MDSC had both beneficial and detrimental effects. HFD-induced MDSC protected mice against diet-induced metabolic dysfunction and reduced HFD-associated inflammation, but also increased the accumulation of fat, enhanced tumor progression, and spontaneous metastasis and reduced survival time. HFD-induced MDSC facilitated tumor growth by limiting the activation of tumor-reactive CD8+ T cells. Leptin, an adipokine that regulates appetite satiety and is overexpressed in obesity, undergoes crosstalk with MDSC in which leptin drives the accumulation of MDSC while MDSC down-regulate the production of leptin. Collectively, these studies demonstrate that although MDSC protect against some metabolic dysfunction associated with HFD they enhance tumor growth in HFD mice and that leptin is a key regulator linking HFD, chronic inflammation, immune suppression, and tumor progression.


Subject(s)
Breast Neoplasms/pathology , Diet, High-Fat/adverse effects , Leptin/adverse effects , Myeloid-Derived Suppressor Cells/immunology , Animals , Breast Neoplasms/etiology , Female , Inflammation/complications , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/metabolism , Obesity/complications , Obesity/immunology , Obesity/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Cancer Immunol Res ; 6(1): 59-68, 2018 01.
Article in English | MEDLINE | ID: mdl-29122838

ABSTRACT

Tumor cells use various immune-suppressive strategies to overcome antitumor immunity. One such method is tumor expression of programmed death ligand-1 (PD-L1), which triggers apoptotic death or anergy upon binding programmed death-1 (PD-1) on T cells. Our previous in vitro cellular studies with human and mouse PD-L1+ tumor cells demonstrated that a soluble form of the costimulatory molecule CD80 prevented PD-L1-mediated immune suppression and restored T-cell activation by binding PD-L1 and blocking interaction with PD-1. We now report that in vivo treatment of established syngeneic PD-L1+ CT26 colon carcinoma and B16F10 melanoma tumors with CD80-Fc delays tumor growth and promotes tumor-infiltrating T cells. Studies with PD-1-/- and CD28-/- mice demonstrate that soluble CD80 acts in vivo by simultaneously neutralizing PD-1 suppression and activating through CD28. We also report that soluble CD80 mediates its effects by activating transcription factors EGR1-4, NF-κB, and MAPK, downstream signaling components of the CD28 and T-cell receptor pathways. Soluble CD80 binds to CTLA-4 on activated human peripheral blood mononuclear cells. However, increasing quantities of CTLA-4 antagonist antibodies do not increase T-cell activation. These results indicate that soluble CD80 does not suppress T-cell function through CTLA-4 and suggest that CTLA-4 acts as a decoy receptor for CD80, rather than functioning as a suppressive signaling receptor. Collectively, these studies demonstrate that soluble CD80 has therapeutic efficacy in vivo in mouse tumor systems and that its effects are due to its ability to inhibit PD-1-mediated suppression while concurrently activating T cells through CD28. Cancer Immunol Res; 6(1); 59-68. ©2017 AACR.


Subject(s)
B7-1 Antigen/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Animals , CD28 Antigens/metabolism , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunomodulation , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental , Mice , Neoplasms/drug therapy , Neoplasms/mortality , Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
J Proteome Res ; 17(1): 315-324, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29061044

ABSTRACT

Ubiquitinated proteins carried by the extracellular vesicles (EV) released by myeloid-derived suppressor cells (MDSC) have been investigated using proteomic strategies to examine the effect of tumor-associated inflammation. EV were collected from MDSC directly following isolation from tumor-bearing mice with low and high inflammation. Among the 1092 proteins (high inflammation) and 925 proteins (low inflammation) identified, more than 50% were observed as ubiquitinated proteoforms. More than three ubiquitin-attachment sites were characterized per ubiquitinated protein, on average. Multiple ubiquitination sites were identified in the pro-inflammatory proteins S100 A8 and S100 A9, characteristic of MDSC and in histones and transcription regulators among other proteins. Spectral counting and pathway analysis suggest that ubiquitination occurs independently of inflammation. Some ubiquitinated proteins were shown to cause the migration of MDSC, which has been previously connected with immune suppression and tumor progression. Finally, MDSC EV are found collectively to carry all the enzymes required to catalyze ubiquitination, and the hypothesis is presented that a portion of the ubiquitinated proteins are produced in situ.


Subject(s)
Extracellular Vesicles/pathology , Inflammation , Myeloid-Derived Suppressor Cells/ultrastructure , Ubiquitin/metabolism , Animals , Binding Sites , Cell Movement , Mice , Ubiquitinated Proteins/analysis , Ubiquitination
4.
J Proteome Res ; 17(1): 486-498, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29139296

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are immature myeloid cells that accumulate in the circulation and the tumor microenvironment of most cancer patients. There, MDSC suppress both adaptive and innate immunity, hindering immunotherapies. The inflammatory milieu often present in cancers facilitates MDSC suppressive activity, causing aggressive tumor progression and metastasis. MDSC from tumor-bearing mice release exosomes, which carry biologically active proteins and mediate some of the immunosuppressive functions characteristic of MDSC. Studies on other cell types have shown that exosomes may also carry RNAs which can be transferred to local and distant cells, yet the mRNA and microRNA cargo of MDSC-derived exosomes has not been studied to date. Here, the cargo of MDSC and their exosomes was interrogated with the goal of identifying and characterizing molecules that may facilitate MDSC suppressive potency. Because inflammation is an established driving force for MDSC suppressive activity, we used the well-established 4T1 mouse mammary carcinoma system, which includes "conventional" as well as "inflammatory" MDSC. We provide evidence that MDSC-derived exosomes carry proteins, mRNAs, and microRNAs with different quantitative profiles than those of their parental cells. Several of these molecules have known or predicted functions consistent with MDSC suppressive activity, suggesting a potential mechanistic redundancy.


Subject(s)
Exosomes/chemistry , Myeloid-Derived Suppressor Cells/chemistry , Animals , Exosomes/immunology , Exosomes/physiology , Immunity , Inflammation , Mice , MicroRNAs/analysis , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/physiology , Proteins/analysis , RNA, Messenger/analysis
5.
Oncotarget ; 8(35): 57964-57980, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28938530

ABSTRACT

Bi-specific T cell engagers (BiTEs) activate T cells through CD3 and target activated T cells to tumor-expressed antigens. BiTEs have shown therapeutic efficacy in patients with liquid tumors; however, they do not benefit all patients. Anti-tumor immunity is limited by Programmed Death 1 (PD1) pathway-mediated immune suppression, and patients who do not benefit from existing BiTES may be non-responders because their T cells are anergized via the PD1 pathway. We have designed a BiTE that activates and targets both T cells and NKT cells to PDL1+ cells. In vitro studies demonstrate that the CD3xPDL1 BiTE simultaneously binds to both CD3 and PDL1, and activates healthy donor CD4+ and CD8+ T cells and NKT cells that are specifically cytotoxic for PDL1+ tumor cells. Cancer patients' PBMC are also activated and cytotoxic, despite the presence of myeloid-derived suppressor cells. The CD3xPDL1 BiTE significantly extends the survival time and maintains activated immune cell levels in humanized NSG mice reconstituted with human PBMC and carrying established human melanoma tumors. These studies suggest that the CD3xPDL1 BiTE may be efficacious for patients with PDL1+ solid tumors, in combination with other immunotherapies that do not specifically neutralize PD1 pathway-mediated immune suppression.

6.
J Leukoc Biol ; 101(5): 1091-1101, 2017 05.
Article in English | MEDLINE | ID: mdl-28007981

ABSTRACT

During successful pregnancy, a woman is immunologically tolerant of her genetically and antigenically disparate fetus, a state known as maternal-fetal tolerance. How this state is maintained has puzzled investigators for more than half a century. Diverse, immune and nonimmune mechanisms have been proposed; however, these mechanisms appear to be unrelated and to act independently. A population of immune suppressive cells called myeloid-derived suppressor cells (MDSCs) accumulates in pregnant mice and women. Given the profound immune suppressive function of MDSCs, it has been suggested that this cell population may facilitate successful pregnancy by contributing to maternal-fetal tolerance. We now report that myeloid cells with the characteristics of MDSCs not only accumulate in the circulation and uterus of female mice following mating but also suppress T cell activation and function in pregnant mice. Depletion of cells with the phenotype and function of MDSCs from gestation d 0.5 through d 7.5 resulted in implantation failure, increased T cell activation, and increased T cell infiltration into the uterus, whereas induction of MDSCs restored successful pregnancy and reduced T cell activation. MDSC-mediated suppression during pregnancy was accompanied by the down-regulation of L-selectin on naïve T cells and a reduced ability of naïve T cells to enter lymph nodes and become activated. Because MDSCs regulate many of the immune and nonimmune mechanisms previously attributed to maternal-fetal tolerance, MDSCs may be a unifying mechanism promoting maternal-fetal tolerance, and their induction may facilitate successful pregnancy in women who spontaneously abort or miscarry because of dysfunctional maternal-fetal tolerance.


Subject(s)
Cell Communication/immunology , Immune Tolerance , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Animals , Cell Count , Embryo Implantation , Embryo, Mammalian , Female , Histocompatibility, Maternal-Fetal , Humans , Immunophenotyping , Lymphocyte Activation , Lymphocyte Depletion , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/cytology , Pregnancy , T-Lymphocytes/cytology
7.
J Proteome Res ; 16(1): 238-246, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27728760

ABSTRACT

In this report, we use a proteomic strategy to identify glycoproteins on the surface of exosomes derived from myeloid-derived suppressor cells (MDSCs), and then test if selected glycoproteins contribute to exosome-mediated chemotaxis and migration of MDSCs. We report successful modification of a surface chemistry method for use with exosomes and identify 21 surface N-glycoproteins on exosomes released by mouse mammary carcinoma-induced MDSCs. These glycoprotein identities and functionalities are compared with 93 N-linked glycoproteins identified on the surface of the parental cells. As with the lysate proteomes examined previously, the exosome surface N-glycoproteins are primarily a subset of the glycoproteins on the surface of the suppressor cells that released them, with related functions and related potential as therapeutic targets. The "don't eat me" molecule CD47 and its binding partners thrombospondin-1 (TSP1) and signal regulatory protein α (SIRPα) were among the surface N-glycoproteins detected. Functional bioassays using antibodies to these three molecules demonstrated that CD47, TSP1, and to a lesser extent SIRPα facilitate exosome-mediated MDSC chemotaxis and migration.


Subject(s)
CD47 Antigen/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Myeloid-Derived Suppressor Cells/metabolism , Proteome/genetics , Thrombospondin 1/genetics , Amino Acid Sequence , Animals , CD47 Antigen/metabolism , Chemotaxis/genetics , Exosomes/chemistry , Exosomes/metabolism , Female , Glycosylation , Mammary Glands, Animal , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Myeloid-Derived Suppressor Cells/pathology , Proteome/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Thrombospondin 1/metabolism
8.
Elife ; 52016 12 08.
Article in English | MEDLINE | ID: mdl-27929373

ABSTRACT

Myeloid-derived suppressor cells (MDSC) contribute to an immunosuppressive network that drives cancer escape by disabling T cell adaptive immunity. The prevailing view is that MDSC-mediated immunosuppression is restricted to tissues where MDSC co-mingle with T cells. Here we show that splenic or, unexpectedly, blood-borne MDSC execute far-reaching immune suppression by reducing expression of the L-selectin lymph node (LN) homing receptor on naïve T and B cells. MDSC-induced L-selectin loss occurs through a contact-dependent, post-transcriptional mechanism that is independent of the major L-selectin sheddase, ADAM17, but results in significant elevation of circulating L-selectin in tumor-bearing mice. Even moderate deficits in L-selectin expression disrupt T cell trafficking to distant LN. Furthermore, T cells preconditioned by MDSC have diminished responses to subsequent antigen exposure, which in conjunction with reduced trafficking, severely restricts antigen-driven expansion in widely-dispersed LN. These results establish novel mechanisms for MDSC-mediated immunosuppression that have unanticipated implications for systemic cancer immunity.


Subject(s)
Adaptive Immunity , Immune Tolerance , L-Selectin/biosynthesis , Lymph Nodes/immunology , Lymphocytes/immunology , Myeloid-Derived Suppressor Cells/physiology , Neoplasms/physiopathology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Lymphocytes/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , RNA Interference , Transplantation, Heterologous
9.
Cancer Res ; 74(20): 5723-33, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25164013

ABSTRACT

Chronic inflammation often precedes malignant transformation and later drives tumor progression. Likewise, subversion of the immune system plays a role in tumor progression, with tumoral immune escape now well recognized as a crucial hallmark of cancer. Myeloid-derived suppressor cells (MDSC) are elevated in most individuals with cancer, where their accumulation and suppressive activity are driven by inflammation. Thus, MDSCs may define an element of the pathogenic inflammatory processes that drives immune escape. The secreted alarmin HMGB1 is a proinflammatory partner, inducer, and chaperone for many proinflammatory molecules that MDSCs develop. Therefore, in this study, we examined HMGB1 as a potential regulator of MDSCs. In murine tumor systems, HMGB1 was ubiquitous in the tumor microenvironment, activating the NF-κB signal transduction pathway in MDSCs and regulating their quantity and quality. We found that HMGB1 promotes the development of MDSCs from bone marrow progenitor cells, contributing to their ability to suppress antigen-driven activation of CD4(+) and CD8(+) T cells. Furthermore, HMGB1 increased MDSC-mediated production of IL-10, enhanced crosstalk between MDSCs and macrophages, and facilitated the ability of MDSCs to downregulate expression of the T-cell homing receptor L-selectin. Overall, our results revealed a pivotal role for HMGB1 in the development and cancerous contributions of MDSCs.


Subject(s)
Cell Differentiation , HMGB1 Protein/physiology , Myeloid Cells/physiology , Tumor Escape , Animals , Antigens, Neoplasm/immunology , Bone Marrow Cells/physiology , Cell Line, Tumor , Coculture Techniques , Female , Interleukin-10/metabolism , L-Selectin/metabolism , Lymphocyte Activation , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Neoplasm Transplantation , Stem Cells/physiology , T-Lymphocytes/immunology , Tumor Microenvironment
10.
J Immunol ; 191(5): 2829-36, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918985

ABSTRACT

Many tumor cells escape anti-tumor immunity through their expression of programmed death ligand-1 (PDL1 or B7-H1), which interacts with T cell-expressed PD1 and results in T cell apoptosis. We previously reported that transfection of human tumor cells with a membrane-bound form of the human costimulatory molecule CD80 prevented PD1 binding and restored T cell activation. We now report that a membrane-bound form of murine CD80 similarly reduces PDL1-PD1-mediated suppression by mouse tumor cells and that a soluble protein consisting of the extracellular domains of human or mouse CD80 fused to the Fc domain of IgG1 (CD80-Fc) overcomes PDL1-mediated suppression by human and mouse tumor cells, respectively. T cell activation experiments with human and mouse tumor cells indicate that CD80-Fc facilitates T cell activation by binding to PDL1 to inhibit PDL1-PD1 interactions and by costimulating through CD28. CD80-Fc is more effective in preventing PD1-PDL1-mediated suppression and restoring T cell activation compared with treatment with mAb to either PD1 or PDL1. These studies identify CD80-Fc as an alternative and potentially more efficacious therapeutic agent for overcoming PDL1-induced immune suppression and facilitating tumor-specific immunity.


Subject(s)
B7-1 Antigen/immunology , B7-H1 Antigen/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Animals , B7-1 Antigen/metabolism , B7-H1 Antigen/metabolism , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes/metabolism , Transfection
11.
Semin Cancer Biol ; 22(4): 275-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22313874

ABSTRACT

The tumor microenvironment is a complex milieu of tumor and host cells. Host cells can include tumor-reactive T cells capable of killing tumor cells. However, more frequently the tumor and host components interact to generate a highly immune suppressive environment that frustrates T cell cytotoxicity and promotes tumor progression through a variety of immune and non-immune mechanisms. Myeloid-derived suppressor cells (MDSC) are a major host component contributing to the immune suppressive environment. In addition to their inherent immune suppressive function, MDSC amplify the immune suppressive activity of macrophages and dendritic cells via cross-talk. This article will review the cell-cell interactions used by MDSC to inhibit anti-tumor immunity and promote progression, and the role of inflammation in promoting cross-talk between MDSC and other cells in the tumor microenvironment.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Tumor Escape , Animals , Dendritic Cells/metabolism , Humans , Immunotherapy , Inflammation , Inflammation Mediators/metabolism , Macrophages/metabolism , Myeloid Cells/metabolism , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology
12.
Blood ; 117(20): 5381-90, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21450901

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) inhibit adaptive and innate immunity and accumulate in the blood of persons with cancer, chronic inflammation, trauma, infection, and stress. Some of the factors inducing their accumulation are known; however, mechanisms regulating their turnover have not been identified. Mass spectrometry showed prominent expression of apoptosis pathway proteins, suggesting that MDSC turnover may be regulated by Fas-FasL-mediated apoptosis. This hypothesis was confirmed by showing that blood MDSCs induced by 3 mouse tumors were Fas(+) and apoptosed in response to Fas agonist in vitro and to activated FasL(+) T cells in vivo. FasL-deficient mice contained significantly more blood MDSCs than FasL(+/+) mice, and after removal of primary tumors MDSCs regressed in STAT6(-/-) and CD1(-/-) mice but not in STAT6(-/-)FasL(-/-) or CD1(-/-)FasL(-/-) mice. Fas(+) macrophages and dendritic cells did not apoptose in response to activated T cells, indicating that Fas-FasL regulation of myeloid cells was restricted to MDSCs. These results identify a new mechanism regulating MDSC levels in vivo and show a retaliatory relationship between T cells and MDSCs in that MDSCs suppress T-cell activation; however, once activated, T cells mediate MDSC apoptosis.


Subject(s)
Fas Ligand Protein/metabolism , Myeloid Cells/cytology , Myeloid Cells/immunology , T-Lymphocytes/immunology , fas Receptor/metabolism , Adoptive Transfer , Animals , Apoptosis , Cell Line, Tumor , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Female , Lymphocyte Activation , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cells/metabolism , T-Lymphocytes/metabolism
13.
Cancer Res ; 70(1): 68-77, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20028852

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are present in most cancer patients and are potent inhibitors of T-cell-mediated antitumor immunity. Their inhibitory activity is attributed to production of arginase, reactive oxygen species, inducible nitric oxide synthase, and interleukin-10. Here we show that MDSCs also block T-cell activation by sequestering cystine and limiting the availability of cysteine. Cysteine is an essential amino acid for T-cell activation because T cells lack cystathionase, which converts methionine to cysteine, and because they do not have an intact xc- transporter and therefore cannot import cystine and reduce it intracellularly to cysteine. T cells depend on antigen-presenting cells (APC), such as macrophages and dendritic cells, to export cysteine, which is imported by T cells via their ASC neutral amino acid transporter. MDSCs express the xc- transporter and import cystine; however, they do not express the ASC transporter and do not export cysteine. MDSCs compete with APC for extracellular cystine, and in the presence of MDSCs, APC release of cysteine is reduced, thereby limiting the extracellular pool of cysteine. In summary, MDSCs consume cystine and do not return cysteine to their microenvironment, thereby depriving T cells of the cysteine they require for activation and function.


Subject(s)
Cysteine/metabolism , Cystine/metabolism , Lymphocyte Activation/immunology , Myeloid Cells/metabolism , T-Lymphocytes/immunology , Amino Acid Transport System y+/metabolism , Animals , Cysteine/immunology , Cystine/immunology , Flow Cytometry , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism
14.
J Immunol ; 183(2): 937-44, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19553533

ABSTRACT

Effective cell-mediated antitumor immunity requires the activation of tumor-reactive T cells and the trafficking of activated T cells to tumor sites. These processes involve the extravasation of lymphocytes from the blood and lymphatics, and their homing to lymph nodes and tumors. L-selectin (CD62L) is an important molecule in these processes. It directs naive lymphocytes to peripheral lymph nodes where they become activated and it traffics naive lymphocytes to inflammatory environments, such as tumors. Individuals with advanced cancer are immune suppressed due to myeloid-derived suppressor cells (MDSC), a population of immature myeloid cells that accumulate to high levels in response to tumor-secreted and proinflammatory factors. We now demonstrate that the reduction in T cell levels of L-selectin that is commonly seen in individuals with cancer inversely correlates with MDSC levels. Three lines of evidence demonstrate that MDSC directly down-regulate L-selectin on naive T cells: 1) naive T cells cocultured with tumor-induced MDSC have reduced L-selectin; 2) T cells in tumor-free aged mice with elevated levels of MDSC have reduced L-selectin, and 3) peritoneal exudate T cells of tumor-free mice treated with plasminogen activator urokinase to elevate MDSC have reduced levels of L-selectin. MDSC are likely to down-regulate L-selectin through their plasma membrane expression of ADAM17 (a disintegrin and metalloproteinase domain 17), an enzyme that cleaves the ectodomain of L-selectin. Therefore, MDSC down-regulate L-selectin levels on naive T cells, decreasing their ability to home to sites where they would be activated. This is another mechanism by which MDSC inhibit antitumor immunity.


Subject(s)
CD4-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/chemistry , Down-Regulation/immunology , L-Selectin/biosynthesis , Myeloid Cells/physiology , Paracrine Communication/immunology , ADAM Proteins/metabolism , ADAM17 Protein , Aging/immunology , Animals , Cell Line, Tumor , Cell Movement/immunology , Coculture Techniques , Immunity, Cellular , L-Selectin/analysis , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Neoplasms, Experimental/immunology , Urokinase-Type Plasminogen Activator/pharmacology
15.
J Leukoc Biol ; 85(6): 996-1004, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19261929

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are potent inhibitors of anti-tumor immunity that facilitate tumor progression by blocking the activation of CD4(+) and CD8(+) T cells and by promoting a type 2 immune response through their production of IL-10 and down-regulation of macrophage production of IL-12. MDSC accumulate in many cancer patients and are a significant impediment to active cancer immunotherapies. Chronic inflammation has been shown recently to enhance the accumulation of MDSC and to increase their suppression of T cells. These findings led us to hypothesize that inflammation contributes to tumor progression through the induction of MDSC, which create a favorable environment for tumor growth. As chronic inflammation also drives type 2 immune responses, which favor tumor growth, we asked if inflammation mediates this effect through MDSC. We find that IL-1beta-induced inflammation increased IL-10 production by MDSC and induces MDSC, which are more effective at down-regulating macrophage production of IL-12 as compared with MDSC isolated from less-inflammatory tumor microenvironments, thereby skewing tumor immunity toward a type 2 response. Inflammation heightens MDSC phenotype by signaling through the TLR4 pathway and involves up-regulation of CD14. Although this pathway is well-recognized in other myeloid cells, it has not been implicated previously in MDSC function. These studies demonstrate that MDSC are an intermediary through which inflammation promotes type 2 immune responses, and they identify the TLR4 pathway in MDSC as a potential target for down-regulating immune suppression and promoting anti-tumor immunity.


Subject(s)
Inflammation/immunology , Myeloid Cells/immunology , Receptor Cross-Talk , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Interleukin-1beta , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Myeloid Cells/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects
16.
Cancer Immunol Immunother ; 57(3): 389-98, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17724589

ABSTRACT

Activation of tumor-reactive T lymphocytes is a promising approach for the prevention and treatment of patients with metastatic cancers. Strategies that activate CD8(+) T cells are particularly promising because of the cytotoxicity and specificity of CD8(+) T cells for tumor cells. Optimal CD8(+) T cell activity requires the co-activation of CD4(+) T cells, which are critical for immune memory and protection against latent metastatic disease. Therefore, we are developing "MHC II" vaccines that activate tumor-reactive CD4(+) T cells. MHC II vaccines are MHC class I(+) tumor cells that are transduced with costimulatory molecules and MHC II alleles syngeneic to the prospective recipient. Because the vaccine cells do not express the MHC II-associated invariant chain (Ii), we hypothesized that they will present endogenously synthesized tumor peptides that are not presented by professional Ii(+) antigen presenting cells (APC) and will therefore overcome tolerance to activate CD4(+) T cells. We now report that MHC II vaccines prepared from human MCF10 mammary carcinoma cells are more efficient than Ii(+) APC for priming and boosting Type 1 CD4(+) T cells. MHC II vaccines consistently induce greater expansion of CD4(+) T cells which secrete more IFNgamma and they activate an overlapping, but distinct repertoire of CD4(+) T cells as measured by T cell receptor Vbeta usage, compared to Ii(+) APC. Therefore, the absence of Ii facilitates a robust CD4(+) T cell response that includes the presentation of peptides that are presented by traditional APC, as well as peptides that are uniquely presented by the Ii(-) vaccine cells.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Antigen-Presenting Cells/immunology , Antigens, Differentiation, B-Lymphocyte/genetics , Breast Neoplasms/genetics , Cancer Vaccines/genetics , Cell Line, Tumor , Flow Cytometry/methods , Genetic Vectors/genetics , Histocompatibility Antigens Class II/genetics , Humans , Th1 Cells/immunology
17.
Cancer Res ; 67(20): 10019-26, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17942936

ABSTRACT

Chronic inflammation is frequently associated with malignant growth and is thought to promote and enhance tumor progression, although the mechanisms which regulate this relationship remain elusive. We reported previously that interleukin (IL)-1beta promoted tumor progression by enhancing the accumulation of myeloid-derived suppressor cells (MDSC), and hypothesized that inflammation leads to cancer through the production of MDSC which inhibit tumor immunity. If inflammation-induced MDSC promote tumor progression by blocking antitumor immunity, then a reduction in inflammation should reduce MDSC levels and delay tumor progression, whereas an increase in inflammation should increase MDSC levels and hasten tumor progression. We have tested this hypothesis using the 4T1 mammary carcinoma and IL-1 receptor (IL-1R)-deficient mice which have a reduced potential for inflammation, and IL-1R antagonist-deficient mice, which have an increased potential for inflammation. Consistent with our hypothesis, IL-1R-deficient mice have a delayed accumulation of MDSC and reduced primary and metastatic tumor progression. Accumulation of MDSC and tumor progression are partially restored by IL-6, indicating that IL-6 is a downstream mediator of the IL-1beta-induced expansion of MDSC. In contrast, excessive inflammation in IL-1R antagonist-deficient mice promotes the accumulation of MDSC and produces MDSC with enhanced suppressive activity. These results show that immune suppression by MDSC and tumor growth are regulated by the inflammatory milieu and support the hypothesis that the induction of suppressor cells which down-regulate tumor immunity is one of the mechanisms linking inflammation and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Animals , Disease Progression , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1/immunology , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , T-Lymphocytes/immunology , Transfection
18.
J Immunol ; 179(2): 977-83, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17617589

ABSTRACT

Although the immune system has the potential to protect against malignancies, many individuals with cancer are immunosuppressed. Myeloid-derived suppressor cells (MDSC) are elevated in many patients and animals with tumors, and contribute to immune suppression by blocking CD4(+) and CD8(+) T cell activation. Using the spontaneously metastatic 4T1 mouse mammary carcinoma, we now demonstrate that cross-talk between MDSC and macrophages further subverts tumor immunity by increasing MDSC production of IL-10, and by decreasing macrophage production of IL-12. Cross-talk between MDSC and macrophages requires cell-cell contact, and the IL-12 decrease is dependent on MDSC production of IL-10. Treatment with the chemotherapeutic drug gemcitabine, which reduces MDSC, promotes rejection of established metastatic disease in IL-4Ralpha(-/-) mice that produce M1 macrophages by allowing T cell activation, by maintaining macrophage production of IL-12, and by preventing increased production of IL-10. Therefore, MDSC impair tumor immunity by suppressing T cell activation and by interacting with macrophages to increase IL-10 and decrease IL-12 production, thereby promoting a tumor-promoting type 2 response, a process that can be partially reversed by gemcitabine.


Subject(s)
Macrophages/immunology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Receptor Cross-Talk/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape/immunology , Animals , Coculture Techniques , Flow Cytometry , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic
19.
Cancer Res ; 67(9): 4507-13, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483367

ABSTRACT

A causative relationship between chronic inflammation and cancer has been postulated for many years, and clinical observations and laboratory experiments support the hypothesis that inflammation contributes to tumor onset and progression. However, the precise mechanisms underlying the relationship are not known. We recently reported that the proinflammatory cytokine, interleukin-1beta, induces the accumulation and retention of myeloid-derived suppressor cells (MDSC), which are commonly found in many patients and experimental animals with cancer and are potent suppressors of adaptive and innate immunity. This finding led us to hypothesize that inflammation leads to cancer through the induction of MDSC, which inhibit immunosurveillance and thereby allow the unchecked persistence and proliferation of premalignant and malignant cells. We now report that host MDSC have receptors for prostaglandin E2 (PGE2) and that E-prostanoid receptor agonists, including PGE2, induce the differentiation of Gr1(+)CD11b(+) MDSC from bone marrow stem cells, whereas receptor antagonists block differentiation. BALB/c EP2 knockout mice inoculated with the spontaneously metastatic BALB/c-derived 4T1 mammary carcinoma have delayed tumor growth and reduced numbers of MDSC relative to wild-type mice, suggesting that PGE2 partially mediates MDSC induction through the EP2 receptor. Treatment of 4T1-tumor-bearing wild-type mice with the cyclooxygenase 2 inhibitor, SC58236, delays primary tumor growth and reduces MDSC accumulation, further showing that PGE2 induces MDSC and providing a therapeutic approach for reducing this tumor-promoting cell population.


Subject(s)
Dinoprostone/immunology , Mammary Neoplasms, Experimental/immunology , Myeloid Cells/immunology , Animals , Cell Differentiation/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Progression , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/pathology , Receptors, Prostaglandin E/immunology , Receptors, Prostaglandin E/metabolism
20.
J Immunol ; 176(1): 284-90, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365420

ABSTRACT

Epidemiological and experimental observations support the hypothesis that chronic inflammation contributes to cancer development and progression; however, the mechanisms underlying the relationship between inflammation and cancer are poorly understood. To study these mechanisms, we have transfected the mouse 4T1 mammary carcinoma with the proinflammatory cytokine IL-1beta to produce a chronic inflammatory microenvironment at the tumor site. Mice with 4T1/IL-1beta tumors have a decreased survival time and elevated levels of immature splenic Gr1+CD11b+ myeloid-derived cells. These myeloid suppressor cells (MSC) are present in many patients with cancer and inhibit the activation of CD4+ and CD8+ T lymphocytes. 4T1/IL-1beta-induced MSC do not express the IL-1R, suggesting that the cytokine does not directly activate MSC. Neither T or B cells nor NKT cells are involved in the IL-1beta-induced increase of MSC because RAG2-/- mice and nude mice with 4T1/IL-1beta tumors also have elevated MSC levels. MSC levels remain elevated in mice inoculated with 4T1/IL-1beta even after the primary tumor is surgically removed, indicating that the IL-1beta effect is long lived. Collectively, these findings suggest that inflammation promotes malignancy via proinflammatory cytokines, such as IL-1beta, which enhance immune suppression through the induction of MSC, thereby counteracting immune surveillance and allowing the outgrowth and proliferation of malignant cells.


Subject(s)
Inflammation/immunology , Interleukin-1/metabolism , Mammary Neoplasms, Experimental/immunology , Myeloid Cells/immunology , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Inflammation/physiopathology , Interleukin-1/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Mice , Mice, Transgenic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...