Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Immunol ; 191(1): 17-23, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23794702

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid cells that play a major role in the regulation of immune responses in many pathological conditions. These cells have a common myeloid origin, relatively immature state, common genetic and biochemical profiles, and, most importantly, the ability to inhibit immune responses. Although initial studies of MDSCs were almost exclusively performed in tumor-bearing mice or cancer patients, in recent years, it became clear that MDSCs play a critical role in the regulation of different types of inflammation that are not directly associated with cancer. In this review we discuss the nature of the complex relationship between MDSCs and the different populations of CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Communication/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/pathology , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology
2.
Stem Cells ; 31(7): 1321-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23533187

ABSTRACT

Natriuretic peptide receptor A (NPRA), the signaling receptor for the cardiac hormone, atrial natriuretic peptide (ANP), is expressed abundantly in inflamed/injured tissues and tumors. NPRA deficiency substantially decreases tissue inflammation and inhibits tumor growth. However, the precise mechanism of NPRA function and whether it links inflammation and tumorigenesis remains unknown. Since both injury repair and tumor growth require stem cell recruitment and angiogenesis, we examined the role of NPRA signaling in tumor angiogenesis as a model of tissue injury repair in this study. In in vitro cultures, aortas from NPRA-KO mice show significantly lower angiogenic response compared to wild-type counterparts. The NPRA antagonist that decreases NPRA expression, inhibits lipopolysaccharide-induced angiogenesis. The reduction in angiogenesis correlates with decreased expression of vascular endothelial growth factor and chemokine (C-X-C motif) receptor 4 (CXCR4) implicating a cell recruitment defect. To test whether NPRA regulates migration of cells to tumors, mesenchymal stem cells (MSCs) were administered i.v., and the results showed that MSCs fail to migrate to the tumor microenvironment in NPRA-KO mice. However, coimplanting tumor cells with MSCs increases angiogenesis and tumorigenesis in NPRA-KO mice, in part by promoting expression of CXCR4 and its ligand, stromal cell-derived factor 1α. Taken together, these results demonstrate that NPRA signaling regulates stem cell recruitment and angiogenesis leading to tumor growth. Thus, NPRA signaling provides a key linkage between inflammation and tumorigenesis, and NPRA may be a target for drug development against cancers and tissue injury repair.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Female , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Atrial Natriuretic Factor/genetics , Signal Transduction , Tumor Microenvironment
3.
Nat Med ; 13(7): 828-35, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17603493

ABSTRACT

Antigen-specific CD8+ T-cell tolerance, induced by myeloid-derived suppressor cells (MDSCs), is one of the main mechanisms of tumor escape. Using in vivo models, we show here that MDSCs directly disrupt the binding of specific peptide-major histocompatibility complex (pMHC) dimers to CD8-expressing T cells through nitration of tyrosines in a T-cell receptor (TCR)-CD8 complex. This process makes CD8-expressing T cells unable to bind pMHC and to respond to the specific peptide, although they retain their ability to respond to nonspecific stimulation. Nitration of TCR-CD8 is induced by MDSCs through hyperproduction of reactive oxygen species and peroxynitrite during direct cell-cell contact. Molecular modeling suggests specific sites of nitration that might affect the conformational flexibility of TCR-CD8 and its interaction with pMHC. These data identify a previously unknown mechanism of T-cell tolerance in cancer that is also pertinent to many pathological conditions associated with accumulation of MDSCs.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Neoplasms/pathology , Adoptive Transfer , Animals , Cells, Cultured , Female , Gene Deletion , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms, Experimental , Nitrates/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
4.
Semin Cancer Biol ; 22(4): 282-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22313876

ABSTRACT

Myeloid derived suppressor cells play a critical role in T cell suppression in cancer. Here, we discuss the mechanisms of how MDSC suppress CD4(+) or CD8(+) T cells in an antigen dependent or non-dependent manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immune Tolerance , Tumor Escape
5.
J Exp Med ; 204(6): 1463-74, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17548519

ABSTRACT

Polymicrobial sepsis alters the adaptive immune response and induces T cell suppression and Th2 immune polarization. We identify a GR-1(+)CD11b(+) population whose numbers dramatically increase and remain elevated in the spleen, lymph nodes, and bone marrow during polymicrobial sepsis. Phenotypically, these cells are heterogeneous, immature, predominantly myeloid progenitors that express interleukin 10 and several other cytokines and chemokines. Splenic GR-1(+) cells effectively suppress antigen-specific CD8(+) T cell interferon (IFN) gamma production but only modestly suppress antigen-specific and nonspecific CD4(+) T cell proliferation. GR-1(+) cell depletion in vivo prevents both the sepsis-induced augmentation of Th2 cell-dependent and depression of Th1 cell-dependent antibody production. Signaling through MyD88, but not Toll-like receptor 4, TIR domain-containing adaptor-inducing IFN-beta, or the IFN-alpha/beta receptor, is required for complete GR-1(+)CD11b(+) expansion. GR-1(+)CD11b(+) cells contribute to sepsis-induced T cell suppression and preferential Th2 polarization.


Subject(s)
CD11b Antigen/metabolism , Immune Tolerance/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Progenitor Cells/immunology , Receptors, Chemokine/metabolism , Sepsis/immunology , Th2 Cells/immunology , Animals , Cell Proliferation , Colony-Forming Units Assay , Cytokines/metabolism , Flow Cytometry , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Signal Transduction/immunology
6.
Biol Blood Marrow Transplant ; 19(5): 692-702, 2013 May.
Article in English | MEDLINE | ID: mdl-23376089

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a group of myeloid cells composed of hematopoietic progenitor cells, immature macrophages, dendritic cells, and granulocytes, which accumulate in inflammatory diseases and various cancers. Here, we investigated the dynamic changes and effects of MDSCs in graft-versus-host disease (GVHD) development and/or tumor relapse after syngeneic and allogeneic bone marrow transplantation (BMT). We found that adding functional MDSCs in donor graft alleviated GVHD, whereas removal of MDSCs in vivo exacerbated GVHD. After T cell-deplete BMT, MDSCs transiently accumulated in the blood and spleen of recipients without GVHD. In contrast, after T cell-replete BMT, the levels of blood MDSCs were constantly elevated in recipients with GVHD. MDSC accumulation positively correlated with the severity of GVHD. Additionally, MDSC accumulation was further increased upon tumor relapse. Although MDSCs isolated from both syngeneic and allogeneic BMT recipients inhibited T cell proliferation in response to alloantigen stimulation ex vivo, MDSCs from the recipients with GVHD showed much higher suppressive potency compared with those from recipients without GVHD. These results indicate that MDSCs can regulate the immune response in acute GVHD, and possibly tumor relapse, subsequent to allogeneic BMT.


Subject(s)
Bone Marrow Transplantation/methods , Myeloid Cells/immunology , Animals , Cell Growth Processes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, Transgenic , Myeloid Cells/metabolism , Transplantation Immunology , Transplantation, Homologous/methods
7.
Curr Opin Pulm Med ; 18(1): 14-22, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22113000

ABSTRACT

PURPOSE OF REVIEW: In this review, we describe the 'state-of-the-art' in our knowledge of asthma and what gaps exist, which can be exploited in the future for effective translation of our knowledge from the bench or population studies to diagnosis and therapy. RECENT FINDINGS: The advent of microbiome research has expanded the potential role of microbes in asthma. There has been a significant increase in our understanding of the pathologic, genetic, cellular and molecular mechanisms of asthma. Nonetheless, the contribution of microbes to the genesis, exacerbation and treatment of asthma are poorly understood. SUMMARY: Asthma is a complex chronic disease of the lung whose incidence is growing at all ages despite the progress that has been made in the areas of diagnosis and treatment of asthma. The complexity is partly due to the environmental insults such as allergens and microbial infections that play differential roles in the pathogenesis of childhood vis-à-vis elderly asthma. Microbes may play important roles in the exacerbation of asthma and hence in the comorbidities due to asthma, and also in the causation of asthma.


Subject(s)
Asthma/microbiology , Corynebacterium Infections/complications , Respiratory Tract Infections/microbiology , Staphylococcal Infections/complications , Age Factors , Aged , Aged, 80 and over , Asthma/etiology , Asthma/immunology , CD4-Positive T-Lymphocytes , Child , Child, Preschool , Corynebacterium Infections/immunology , Female , Humans , Infant , Interleukin-1/immunology , Interleukin-6/immunology , Male , Metagenome , Respiratory Tract Infections/complications , Respiratory Tract Infections/immunology , Staphylococcal Infections/immunology , Tumor Necrosis Factor-alpha/immunology
8.
J Immunol ; 184(6): 3106-16, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20142361

ABSTRACT

Ag-specific T cell tolerance plays a critical role in tumor escape. Recent studies implicated myeloid-derived suppressor cells (MDSCs) in the induction of CD8(+) T cell tolerance in tumor-bearing hosts. However, the mechanism of this phenomenon remained unclear. We have found that incubation of Ag-specific CD8(+) T cells, with peptide-loaded MDSCs, did not induce signaling downstream of TCR. However, it prevented subsequent signaling from peptide-loaded dendritic cells. Using double TCR transgenic CD8(+) T cells, we have demonstrated that MDSC induced tolerance to only the peptide, which was presented by MDSCs. T cell response to the peptide specific to the other TCR was not affected. Incubation of MDSCs with Ag-specific CD8(+) T cells caused nitration of the molecules on the surface of CD8(+) T cells, localized to the site of physical interaction between MDSC and T cells, which involves preferentially only TCR specific for the peptide presented by MDSCs. Postincubation with MDSCs, only nitrotyrosine-positive CD8(+) T cells demonstrated profound nonresponsiveness to the specific peptide, whereas nitrotyrosine-negative CD8(+) T cells responded normally to that stimulation. MDSCs caused dissociation between TCR and CD3zeta molecules, disrupting TCR complexes on T cells. Thus, these data describe a novel mechanism of Ag-specific CD8(+) T cell tolerance in cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Transplantation Tolerance/immunology , Adoptive Transfer , Animals , Antigen Presentation/genetics , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Female , Genes, T-Cell Receptor beta/immunology , Leukemia, Experimental/genetics , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Myeloid Cells/transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Transplantation Tolerance/genetics , Tumor Cells, Cultured
9.
J Immunol ; 181(8): 5791-802, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832739

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are a heterogeneous group of cells that play a critical role in tumor associated immune suppression. In an attempt to identify a specific subset of MDSC primarily responsible for immunosuppressive features of these cells, 10 different tumor models were investigated. All models showed variable but significant increase in the population of MDSC. Variability of MDSC expansion in vivo matched closely the effect of tumor cell condition medium in vitro. MDSC consists of two major subsets of Ly6G(+)Ly6C(low) granulocytic and Ly6G(-)Ly6C(high) monocytic cells. Granulocytic MDSC have increased level of reactive oxygen species and undetectable level of NO whereas monocytic MDSC had increased level of NO but undetectable levels of reactive oxygen species. However, their suppressive activity per cell basis was comparable. Almost all tumor models demonstrated a preferential expansion of granulocytic subset of MDSC. We performed a phenotypical and functional analysis of several surface molecules previously suggested to be involved in MDSC-mediated suppression of T cells: CD115, CD124, CD80, PD-L1, and PD-L2. Although substantial proportion of MDSC expressed those molecules no differences in the level of their expression or the proportion, positive cells were found between MDSC and cells from tumor-free mice that lack immune suppressive activity. The level of MDSC-mediated T cell suppression did not depend on the expression of these molecules. These data indicate that suppressive features of MDSC is caused not by expansion of a specific subset but more likely represent a functional state of these cells.


Subject(s)
Granulocytes/immunology , Monocytes/immunology , Neoplasms, Experimental/immunology , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/immunology , Female , Gene Expression Regulation/immunology , Granulocytes/metabolism , Granulocytes/pathology , Mice , Mice, Inbred BALB C , Monocytes/metabolism , Monocytes/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nitric Oxide/immunology , Nitric Oxide/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism
10.
Adv Exp Med Biol ; 601: 213-23, 2007.
Article in English | MEDLINE | ID: mdl-17713008

ABSTRACT

The development of tumor-specific T cell tolerance is largely responsible for tumor escape. Accumulation of myeloid-derived suppressor cells (MDSCs) in animal tumor models as well as in cancer patients is involved in tumor-associated T cell tolerance. In recent years, it has become increasingly evident that MDSCs bring about antigen-specific T cell tolerance by various mechanisms, which is the focus of this chapter.


Subject(s)
Myeloid Cells/cytology , Animals , Antigens/metabolism , Arginine/chemistry , Cell Differentiation , Epitopes/chemistry , Humans , Immune Tolerance , Models, Biological , Neoplasms/immunology , Peroxynitrous Acid/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta/metabolism
11.
Cancer Res ; 65(20): 9525-35, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230418

ABSTRACT

Abnormal dendritic cell differentiation and accumulation of immunosuppressive myeloid cells in cancer is one of the major factors of tumor nonresponsiveness. We have previously shown that hyperactivation of the Janus-activated kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3) induced by tumor-derived factors (TDF) is responsible for abnormal dendritic cell differentiation. Here, using a novel selective inhibitor of JAK2/STAT3 JSI-124, we investigated the possibility of pharmacologic regulation of dendritic cell differentiation in cancer. Our experiments in vitro have shown that JSI-124 overcomes the differentiation block induced by TDF and promotes the differentiation of mature dendritic cells and macrophages. JSI-124 significantly reduced the presence of immature myeloid cells in vivo and promoted accumulation of mature dendritic cells. In addition to a direct antitumor effect in several animal models, JSI-124 significantly enhanced the effect of cancer immunotherapy. This indicates that pharmacologic inhibition of the JAK2/STAT3 pathway can be an important new therapeutic strategy to enhance antitumor activity of cancer immunotherapy.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Triterpenes/pharmacology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Dendritic Cells/cytology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/immunology , Female , Janus Kinase 2 , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/immunology , NIH 3T3 Cells , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , Sarcoma, Experimental/drug therapy , Sarcoma, Experimental/immunology , Sarcoma, Experimental/therapy , Signal Transduction/drug effects , Signal Transduction/immunology
12.
J Interferon Cytokine Res ; 24(8): 490-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15320962

ABSTRACT

Standard chemotherapy for pancreatic carcinoma is based on the use of gemcitabine. The clinical benefit of interferon-alpha (IFN-alpha) in advanced pancreatic cancer has been shown. However, it has been demonstrated that to be effective, there is a need for a constant amount of IFN-alpha at the site of the tumor. Therefore, we examined transfection of the human pancreatic cancer cell line DAN-G with a retrovirus encoding for IFN-alpha and the effect of IFN-alpha gene expression alone or in combination with gemcitabine on growth inhibition of DAN-G pancreatic cancer cells in vitro and in vivo in orthotopically implanted DAN-G cells in nude mice. DAN-G cells could be efficiently transfected retrovirally by the human IFN-alpha gene and significantly enhanced the levels of IFN-alpha mRNA. In vitro gemcitabine led to an alteration of G1/S phase progression in transduced as well as untransduced cells, whereas IFN-alpha led to a significant decrease in cell viability in the transduced cells via delay in the progression of the S phase but no alteration of G1/S phase progression. In vivo, tumor volume in mice was reduced significantly with gemcitabine combined with IFN-alpha (76% +/- 8.3%) compared with gemcitabine alone (62.9% +/- 7.3%) or IFN-alpha alone (24.4% +/- 5.2%) compared with untreated animals. We conclude that gemcitabine and IFN-alpha concomitantly inhibited tumor cell proliferation significantly.


Subject(s)
Cell Cycle , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Interferon-alpha/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Retroviridae/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA, Complementary/genetics , Deoxycytidine/therapeutic use , Female , Gene Expression , Genetic Therapy , Humans , Interferon-alpha/biosynthesis , Interferon-alpha/metabolism , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Transgenes/genetics , Gemcitabine
13.
Cell Stress Chaperones ; 8(4): 348-60, 2003.
Article in English | MEDLINE | ID: mdl-15115287

ABSTRACT

Previously we described an involvement of the C-type lectin receptor CD94 and the neuronal adhesion molecule CD56 in the interaction of natural killer (NK) cells with Hsp70-protein and Hsp70-peptide TKD. Therefore, differences in the cell surface density of these NK cell-specific markers were investigated comparatively in CD94-sorted, primary NK cells and in established NK cell lines NK-92, NKL, and YT after TKD stimulation. Initially, all NK cell types were positive for CD94; the CD56 expression varied. After stimulation with TKD, the mean fluorescence intensity (mfi) of CD94 and CD56 was upregulated selectively in primary NK cells but not in NK cell lines. Other cell surface markers including natural cytotoxicity receptors remained unaffected in all cell types. CD3-enriched T cells neither expressing CD94 nor CD56 served as a negative control. High receptor densities of CD94/CD56 were associated with an increased cytolytic response against Hsp70 membrane-positive tumor target cells. The major histocompatibility complex (MHC) class I-negative, Hsp70-positive target cell line K562 was efficiently lysed by primary NK cells and to a lower extent by NK lines NK-92 and NKL. YT and CD3-positive T cells were unable to kill K562 cells. MHC class-I and Hsp70-positive, Cx + tumor target cells were efficiently lysed only by CD94-sorted, TKD-stimulated NK cells with high CD94/CD56 mfi values. Hsp70-specificity was demonstrated by antibody blocking assays, comparative phenotyping of the tumor target cells, and by correlating the amount of membrane-bound Hsp70 with the sensitivity to lysis. Remarkably, a 14-mer peptide (LKD), exhibiting only 1 amino acid exchange at position 1 (T to L), neither stimulated Hsp70-reactivity nor resulted in an upregulated CD94 expression on primary NK cells. Taken together our findings indicate that an MHC class I-independent, Hsp70 reactivity could be associated with elevated cell surface densities of CD94 and CD56 after TKD stimulation.


Subject(s)
Antigens, CD/metabolism , CD56 Antigen/metabolism , HSP70 Heat-Shock Proteins/metabolism , Killer Cells, Natural/metabolism , Lectins, C-Type/metabolism , CD3 Complex/metabolism , Humans , K562 Cells , NK Cell Lectin-Like Receptor Subfamily D , T-Lymphocytes/metabolism
14.
Genet Vaccines Ther ; 2: 12, 2004.
Article in English | MEDLINE | ID: mdl-15329148

ABSTRACT

Modulation of the immune system by genetically modified immunological effector cells is of potential therapeutic value in the treatment of malignancies. Interleukin-2 (IL-2) is a crucial cytokine which induces potent antitumor response. Cytokine-induced killer cells (CIK) have been described as highly efficient cytotoxic effector cells capable of lysing tumor cell targets and are capable of recognizing these cells in a non-MHC restricted fashion. Dendritic cells (DC) are the major antigen presenting cells. This study evaluated the antitumor effect of CIK cells which were non-virally transfected with IL-2 and co-cultured with pulsed and unpulsed DC. Human CIK cells generated from peripheral blood were transfected in vitro with plasmid encoding for the human IL-2. Transfection involved a combination of electrical parameters and a specific solution to deliver plasmid directly to the cell nucleus by using the Nucleofector® electroporation system. Nucleofection resulted in the production of IL-2 with a mean of 478.5 pg/106 cells (range of 107.6-1079.3 pg /106 cells/24 h) compared to mock transfected CIK cells (31 pg/106 cells) (P = 0.05). After co-culturing with DC their functional ability was assessed in vitro by a cytotoxicity assay. On comparison with non-transfected CIK cells co-cultured with DCs (36.5 ± 5.3 %), transfected CIK cells co-cultured with DC had a significantly higher lytic activity of 58.5 ± 3.2% (P = 0.03) against Dan G cells, a human pancreatic carcinoma cell line.

16.
Cancer Res ; 72(4): 928-38, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22237629

ABSTRACT

Myeloid-derived suppressor cells (MDSC) play a major role in cancer-related immune suppression, yet the nature of this suppression remains controversial. In this study, we evaluated the ability of MDSCs to elicit CD4(+) T-cell tolerance in different mouse tumor models. In contrast to CD8(+) T-cell tolerance, which could be induced by MDSCs in all the tumor models tested, CD4(+) T-cell tolerance could be elicited in only one of the models (MC38) in which a substantial level of MHC class II was expressed on MDSCs compared with control myeloid cells. Mechanistic investigations revealed that MDSCs deficient in MHC class II could induce tolerance to CD8(+) T cells but not to CD4(+) T cells. Unexpectedly, antigen-specific CD4(+) T cells (but not CD8(+) T cells) could dramatically enhance the immune suppressive activity of MDSCs by converting them into powerful nonspecific suppressor cells. This striking effect was mediated by direct cell-cell contact through cross-linking of MHC class II on MDSCs. We also implicated an Ets-1 transcription factor-regulated increase in expression of Cox-2 and prostaglandin E2 in MDSCs in mediating this effect. Together, our findings suggest that activated CD4(+) T cells that are antigen specific may enhance the immune suppressive activity of MDSCs, a mechanism that might serve normally as a negative feedback loop to control immune responses that becomes dysregulated in cancer.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immune Tolerance , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Epitopes , Female , Histocompatibility Antigens Class II/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology
17.
Cancer J ; 16(4): 348-53, 2010.
Article in English | MEDLINE | ID: mdl-20693846

ABSTRACT

Myeloid-derived suppressor cells are one of the major factors responsible for immune suppression in cancer. They also contribute to limited efficacy of current vaccination strategies. Here, we give an overview of the myeloid-derived suppressor cells field focusing primarily on the studies in cancer patients and current and future therapeutic options targeting these cells.


Subject(s)
Antigen-Presenting Cells/microbiology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/pathology , Drug Delivery Systems/methods , Humans , Mice , Myeloid Cells/drug effects , Myeloid Cells/pathology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology
18.
J Clin Invest ; 120(4): 1111-24, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20234093

ABSTRACT

Cancer immunotherapy faces a serious challenge because of low clinical efficacy. Recently, a number of clinical studies have reported the serendipitous finding of high rates of objective clinical response when cancer vaccines are combined with chemotherapy in patients with different types of cancers. However, the mechanism of this phenomenon remains unclear. Here, we tested in mice several cancer vaccines and an adoptive T cell transfer approach to cancer immunotherapy in combination with several widely used chemotherapeutic drugs. We found that chemotherapy made tumor cells more susceptible to the cytotoxic effect of CTLs through a dramatic perforin-independent increase in permeability to GrzB released by the CTLs. This effect was mediated via upregulation of mannose-6-phosphate receptors on the surface of tumor cells and was observed in mouse and human cells. When combined with chemotherapy, CTLs raised against specific antigens were able to induce apoptosis in neighboring tumor cells that did not express those antigens. These data suggest that small numbers of CTLs could mediate a potent antitumor effect when combined with chemotherapy. In addition, these results provide a strong rationale for combining these modalities for the treatment of patients with advanced cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Cytotoxicity, Immunologic/drug effects , Immunotherapy , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cisplatin/pharmacology , Dendritic Cells/immunology , Female , Granzymes/physiology , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Paclitaxel/pharmacology , Vaccination
19.
Clin Cancer Res ; 16(6): 1812-23, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215551

ABSTRACT

PURPOSE: Myeloid-derived suppressor cells (MDSC) are one of the major factors responsible for immune suppression in cancer. Therefore, it would be important to identify effective therapeutic means to modulate these cells. EXPERIMENTAL DESIGN: We evaluated the effect of the synthetic triterpenoid C-28 methyl ester of 2-cyano-3,12-dioxooleana-1,9,-dien-28-oic acid (CDDO-Me; bardoxolone methyl) in MC38 colon carcinoma, Lewis lung carcinoma, and EL-4 thymoma mouse tumor models, as well as blood samples from patients with renal cell cancer and soft tissue sarcoma. Samples were also analyzed from patients with pancreatic cancer treated with CDDO-Me in combination with gemcitabine. RESULTS: CDDO-Me at concentrations of 25 to 100 nmol/L completely abrogated immune suppressive activity of MDSC in vitro. CDDO-Me reduced reactive oxygen species in MDSCs but did not affect their viability or the levels of nitric oxide and arginase. Treatment of tumor-bearing mice with CDDO-Me did not affect the proportion of MDSCs in the spleens but eliminated their suppressive activity. This effect was independent of antitumor activity. CDDO-Me treatment decreased tumor growth in mice. Experiments with severe combined immunodeficient-beige mice indicated that this effect was largely mediated by the immune system. CDDO-Me substantially enhanced the antitumor effect of a cancer vaccines. Treatment of pancreatic cancer patients with CDDO-Me did not affect the number of MDSCs in peripheral blood but significantly improved the immune response. CONCLUSIONS: CDDO-Me abrogated the immune suppressive effect of MDSCs and improved immune responses in tumor-bearing mice and cancer patients. It may represent an attractive therapeutic option by enhancing the effect of cancer immunotherapy.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Myeloid Cells/immunology , Neoplasms, Experimental/immunology , Oleanolic Acid/analogs & derivatives , Aged , Aged, 80 and over , Animals , Arginase/metabolism , Clinical Trials, Phase I as Topic , Dendritic Cells/pathology , Dendritic Cells/transplantation , Female , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Middle Aged , Myeloid Cells/pathology , Myeloid Cells/transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nitric Oxide/metabolism , Oleanolic Acid/pharmacology , Prognosis , Reactive Oxygen Species/metabolism
20.
Nat Med ; 16(8): 880-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622859

ABSTRACT

Dendritic cells (DCs), a type of professional antigen-presenting cells, are responsible for initiation and maintenance of immune responses. Here we report that a substantial proportion of DCs in tumor-bearing mice and people with cancer have high amounts of triglycerides as compared with DCs from tumor-free mice and healthy individuals. In our studies, lipid accumulation in DCs was caused by increased uptake of extracellular lipids due to upregulation of scavenger receptor A. DCs with high lipid content were not able to effectively stimulate allogeneic T cells or present tumor-associated antigens. DCs with high and normal lipid levels did not differ in expression of major histocompatibility complex and co-stimulatory molecules. However, lipid-laden DCs had a reduced capacity to process antigens. Pharmacological normalization of lipid abundance in DCs with an inhibitor of acetyl-CoA carboxylase restored the functional activity of DCs and substantially enhanced the effects of cancer vaccines. These findings suggest that immune responses in cancer can be improved by manipulating the lipid levels in DCs.


Subject(s)
Dendritic Cells/physiology , Lipid Metabolism/physiology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Boron Compounds/pharmacokinetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Fatty Acids/analysis , Fatty Acids/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Neoplasms/pathology , Neoplasms/physiopathology , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism , Spectrometry, Mass, Electrospray Ionization , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL