Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Zhonghua Xue Ye Xue Za Zhi ; 39(10): 839-844, 2018 Oct 14.
Article in Chinese | MEDLINE | ID: mdl-30373357

ABSTRACT

Objective: To elucidate the expression levels of key immune biomarkers, phosphate and tension homology deleted on chromosome ten (PTEN) and programmed cell death protein1(PD-1),of different immune tolerance pathway in classic Hodgkin's lymphoma (CHL) to further determine their clinical role and prognostic significance. Methods: The clinical features and prognostic factors of 56 CHL patients, who were admitted to the TianJin Medical University Cancer Institute from February 2003 to August 2013, were retrospectively analyzed. PTEN and PD-1 protein expression levels were analyzed by immunohistochemistry, Epstein-Barr virus encoded RNA (EBER) was performed by in situ hybridization assay. Correlations between the expression of biomarkers and clinicopathologic parameters were examined and survival analyses were performed. Results: This cohort of 56 CHL patients included 34 males and 22 females with a median age of 25 years (ranged from 7 to 71 years). In a univariate analysis, age≥45, IPS score >2, EBER positive, high expression of PTEN protein conferred inferior 5-year OS and 5-year PFS; In a multivariate model, age≥45, IPS score >2, EBER positive, high expression of PTEN protein were identified as the independent adverse prognostic factors for CHL. Conclusions: This study suggested for the first time that PTEN was independent prognostic immune biomarkers in CHL, which provided the novel therapeutic strategy of immune therapy for CHL.


Subject(s)
Hodgkin Disease , PTEN Phosphohydrolase/analysis , Programmed Cell Death 1 Receptor/analysis , Adolescent , Adult , Aged , Child , Female , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Young Adult
2.
Mol Oncol ; 9(7): 1447-1457, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25957812

ABSTRACT

The median survival for metastatic melanoma is in the realm of 8-16 months and there are few therapies that offer significant improvement in overall survival. One of the recent advances in cancer treatment focuses on epigenetic modifiers to alter the survivability and immunogenicity of cancer cells. Our group and others have previously demonstrated that pan-HDAC inhibitors induce apoptosis, cell cycle arrest and changes in the immunogenicity of melanoma cells. Here we interrogated specific HDACs which may be responsible for this effect. We found that both genetic abrogation and pharmacologic inhibition of HDAC6 decreases in vitro proliferation and induces G1 arrest of melanoma cell lines without inducing apoptosis. Moreover, targeting this molecule led to an important upregulation in the expression of tumor associated antigens and MHC class I, suggesting a potential improvement in the immunogenicity of these cells. Of note, this anti-melanoma activity was operative regardless of mutational status of the cells. These effects translated into a pronounced delay of in vivo melanoma tumor growth which was, at least in part, dependent on intact immunity as evidenced by the restoration of tumor growth after CD4+ and CD8+ depletion. Given our findings, we provide the initial rationale for the further development of selective HDAC6 inhibitors as potential therapeutic anti-melanoma agents.


Subject(s)
Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/drug effects , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Animals , Cell Line, Tumor , G1 Phase/drug effects , Histone Deacetylase 6 , Humans , Melanoma, Experimental/enzymology , Mice , Mice, Inbred C57BL
3.
Leukemia ; 25(1): 145-52, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20966935

ABSTRACT

B-cell lymphoma 6 (BCL6) and PR domain containing 1 (PRDM1) are considered as master regulators for germinal center (GC) formation and terminal B-cell differentiation. Dysregulation of BCL6 and PRDM1 has been associated with lymphomagenesis. Here, we show for the first time that direct cell-cell contact between follicular dendritic cells (FDC) and B-lymphocytes, by influencing the expression of a set of microRNAs (miRNAs), regulates the expression of BCL6 and PRDM1. We identify that, on cell adhesion to FDC, FDC induces upregulation of PRDM1 expression through downregulation of miR-9 and let-7 families and induces downregulation of BCL-6 through upregulation of miR-30 family in B-lymphocytes and lymphoma cells. We further demonstrate that the miR-30 family directly controls BCL-6 expression and miR-9-1 and let-7a directly control PRDM-1 expression through targeting their 3'UTR, mediating the FDC effect. Our studies define a novel regulatory mechanism in which the FDC, through induction of miRNAs in B-lymphocytes, orchestrates the regulation of transcription factors, promotes germinal center B-cell survival and differentiation. Dysregulation of miRNAs may interfere with B-cell survival and maturation, thus representing a novel molecular mechanism, as well as a potential therapeutic target in B-cell lymphomas.


Subject(s)
DNA-Binding Proteins/genetics , Dendritic Cells, Follicular/physiology , Lymphoma, B-Cell/metabolism , MicroRNAs/physiology , Repressor Proteins/genetics , 3' Untranslated Regions/physiology , Cell Communication , Cell Line, Tumor , Cell Survival , Down-Regulation , Humans , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6 , Up-Regulation
4.
Oncogene ; 29(2): 157-73, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19855430

ABSTRACT

The initiation, magnitude and duration of an immune response against antigens are a tightly regulated process involving a dynamic, orchestrated balance of pro- and anti-inflammatory pathways in immune cells. Such a delicate balance is critical for allowing efficient immune response against foreign antigens while preventing autoimmune attack against self-antigens. In recent years, much effort has been devoted to understanding immune evasion by cancer cells. Also, significant advances have been made in mechanistically understanding the role of pro- and anti-inflammatory cytokines in the regulation of immune responses against antigens, including those expressed by tumors. However, we still know very little about the regulation of inflammatory/anti-inflammatory genes in their natural setting, the chromatin substrate. Several mechanisms have been identified to influence chromatin flexibility and allow dynamic changes in gene expression. Among those, chromatin modifications induced by acetylation and deacetylation of histone tails have gained wide attention. In this study, we discuss the role of histone deacetylases in the transcriptional regulation of genes involved in the inflammatory response and how these enzymes coordinate the dynamic expression of these genes during an immune response. This emerging knowledge is opening new avenues to better understand epigenetic regulation of inflammatory responses and providing new molecular targets for either amplifying or ameliorating immune responses.


Subject(s)
Cytokines/metabolism , Histone Deacetylases/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Animals , Cytokines/genetics , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation Mediators/metabolism , Models, Biological , Neoplasms/genetics , Signal Transduction
5.
Leuk Res ; 26(5): 461-71, 2002 May.
Article in English | MEDLINE | ID: mdl-11916520

ABSTRACT

We report single institution outcome of brief, intensive ara-C-based chemotherapy using bone marrow transplantation as primary intensification for untreated adult patients with acute lymphoblastic leukemia (ALL). Overall disease-free and overall survival were inferior to those reported with prolonged chemotherapy modeled on pediatric protocols. Survival and disease-free survival were superior for patients receiving allogeneic BMT compared with chemopurged autologous transplant or maintenance chemotherapy (patients ineligible for or declining BMT). In multivariate analysis, non-L2-FAB, higher ara-C dose, absence of CNS disease, non-Ph1+ karyotype, allogeneic BMT, T cell phenotype, and younger age were associated with improved disease-free survival. Autologous BMT was not superior to chemotherapy, and appears unlikely to provide adequate curative treatment for most adult ALL patients if not followed by maintenance.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Bone Marrow Transplantation , Cytarabine/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Aged , Follow-Up Studies , Humans , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Transplantation, Autologous
6.
Blood ; 98(4): 1070-7, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11493453

ABSTRACT

Tumor antigen-specific T-cell tolerance may limit the efficacy of therapeutic cancer vaccines. Direct presentation of antigens by tumor cells incapable of providing adequate costimulation to tumor-specific T cells has been suggested as the basis for this unresponsiveness. Using parent-into-F1 bone marrow (BM) chimeras, this study unambiguously demonstrates that the induction of this tolerant state requires T-cell recognition of tumor antigen presented by BM-derived antigen-presenting cells (APCs), not tumor cells themselves. In the absence of host APC presentation, tumor-specific T cells remained functional, even in the setting of antigen expressed by B-cell lymphomas residing in secondary lymphoid tissues. The intrinsic APC capacity of tumor cells has therefore little influence over T-cell priming versus tolerance, a decision that is regulated at the level of host APCs. (Blood. 2001;98:1070-1077)


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Lymphoma, B-Cell/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/pharmacology , Bone Marrow Cells/immunology , Bone Marrow Transplantation , CD4-Positive T-Lymphocytes/transplantation , Clone Cells/immunology , Disease Progression , Hemagglutinins, Viral/immunology , Immune Tolerance/drug effects , Lymphocyte Activation/immunology , Lymphoma, B-Cell/pathology , Male , Mice , Mice, SCID , Mice, Transgenic , Models, Biological , Neoplasm Transplantation , Transplantation Chimera , Tumor Cells, Cultured/immunology , Tumor Cells, Cultured/transplantation
7.
J Immunol ; 166(9): 5398-406, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11313376

ABSTRACT

The mechanism of tumor-associated T cell dysfunction remains an unresolved problem of tumor immunology. Development of T cell defects in tumor-bearing hosts are often associated with increased production of immature myeloid cells. In tumor-bearing mice, these immature myeloid cells are represented by a population of Gr-1(+) cells. In this study we investigated an effect of these cells on T cell function. Gr-1(+) cells were isolated from MethA sarcoma or C3 tumor-bearing mice using cell sorting. These Gr-1(+) cells expressed myeloid cell marker CD11b and MHC class I molecules, but they lacked expression of MHC class II molecules. Tumor-induced Gr-1(+) cells did not affect T cell responses to Con A and to a peptide presented by MHC class II. In sharp contrast, Gr-1(+) cells completely blocked T cell response to a peptide presented by MHC class I in vitro and in vivo. Block of the specific MHC class I molecules on the surface of Gr-1(+) cells completely abrogated the observed effects of these cells. Thus, immature myeloid cells specifically inhibited CD8-mediated Ag-specific T cell response, but not CD4-mediated T cell response. Differentiation of Gr-1(+) cells in the presence of growth factors and all-trans retinoic acid completely eliminated inhibitory potential of these cells. This may suggest a new approach to cancer treatment.


Subject(s)
Immune Tolerance/immunology , Myeloid Cells/immunology , Sarcoma, Experimental/immunology , Sarcoma, Experimental/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Separation , Cells, Cultured , Female , Histocompatibility Antigens Class I/biosynthesis , Injections, Subcutaneous , Lymphocyte Activation/immunology , Methylcholanthrene , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/metabolism , Neoplasm Transplantation , Sarcoma, Experimental/chemically induced , Spleen/cytology , Spleen/immunology
8.
Blood ; 95(10): 3011-9, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10807763

ABSTRACT

For many cancers, autologous bone marrow transplantation (BMT) achieves a minimal residual disease state, yet relapse rates remain high. Using a syngeneic murine bone marrow transplant model, we demonstrate that vaccination with irradiated granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing autologous tumor cells is effective in the post-BMT period and actually results in a greater tumor-free survival than vaccination in the nontransplant setting. Employing T cells specific for a model tumor-antigen, we find that transplantation of the tumor-bearing host results in a massive expansion and activation of tumor-specific T cells in the early posttransplant period, but this response rapidly declines in association with tumor progression. Immunization with irradiated GM-CSF tumor cells during the period of immune reconstitution results in the sustained amplification and activation of this response that closely correlates with freedom from relapse. These results demonstrate the feasibility of integrating GM-CSF vaccines in the postautologous BMT setting and suggest mechanisms that may contribute to the observed efficacy of immunization during the critical period of immune reconstitution.


Subject(s)
Bone Marrow Transplantation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Graft vs Tumor Effect , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunization , Mice , Mice, Inbred BALB C , Secondary Prevention , T-Lymphocytes/immunology
9.
Proc Natl Acad Sci U S A ; 96(20): 11476-81, 1999 Sep 28.
Article in English | MEDLINE | ID: mdl-10500201

ABSTRACT

The efficacy of therapeutic vaccination for the treatment of cancer is limited by peripheral tolerance to tumor antigens. In vivo blockade of CTLA-4, a negative regulator of T cell function, can induce the regression of established tumors and can augment the tumor rejection achieved through therapeutic vaccination. These outcomes may reflect enhanced tumor-specific T cell priming and/or interference with the development of tolerance to tumor antigens. We examined the effect of CTLA-4 blockade on the fate and function of T cells specific for a model tumor antigen in the tumor-bearing host. We found that while CTLA-4 blockade enhanced the priming of responsive T cells, it did not prevent the induction of tolerance to tumor antigens. These results demonstrate that there is a critical window in which the combination of CTLA-4 blockade and vaccination achieves an optimal response, and they point to mechanisms other than CTLA-4 engagement in mediating peripheral T cell tolerance to tumor antigens.


Subject(s)
Antigens, Differentiation/physiology , Antigens, Neoplasm/immunology , Immune Tolerance , Immunoconjugates , T-Lymphocytes/immunology , Abatacept , Animals , Antigens, CD/physiology , B7-1 Antigen/physiology , B7-2 Antigen , CTLA-4 Antigen , Male , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Vaccination
10.
Hum Gene Ther ; 10(12): 1983-91, 1999 Aug 10.
Article in English | MEDLINE | ID: mdl-10466632

ABSTRACT

Irradiated tumor cells transduced with the gene encoding the cytokine GM-CSF have been extensively studied as a vaccine formulation capable of priming systemic antitumor immune responses in the tumor-bearing host. In spite of the therapeutic promise of this vaccine strategy demonstrated in both animal models and early-phase clinical trials, clinical development has been limited by difficulties pertaining to the need to establish in culture the tumor of each patient and to perform individualized gene transfer. To circumvent these issues, we generated an HLA-negative human cell line producing large quantities of human GM-CSF for use as a universal bystander cell to be mixed with unmodified autologous tumor cells in the formulation of a vaccine. This line is easily propagated as a suspension culture in defined, serum-free medium. In a mouse model, we find that vaccination with a mixture of autologous tumor cells and an MHC-negative allogeneic GM-CSF-producing bystander cell primes antitumor immune responses that are equivalent or better than those achieved using autologous tumor cells directly transduced to secrete GM-CSF. This strategy greatly simplifies further clinical development of autologous tumor cell-based vaccines.


Subject(s)
Cancer Vaccines , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor Cells, Cultured , Animals , Cell Line , Cell Transplantation , Disease Models, Animal , Gene Transfer Techniques , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Lymphoma, B-Cell/therapy , Mice , Mice, Inbred BALB C , Transduction, Genetic
11.
Nat Med ; 5(7): 780-7, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395323

ABSTRACT

Tumor antigen-specific T-cell tolerance limits the efficacy of therapeutic cancer vaccines. Antigen-presenting cells mediate the induction of T-cell tolerance to self-antigens. We therefore assessed the fate of tumor-specific CD4+ T cells in tumor-bearing recipients after in vivo activation of antigen-presenting cells with antibodies against CD40. Such treatment not only preserved the responsiveness of this population, but resulted in their endogenous activation. Established tumors regressed in vaccinated mice treated with antibody against CD40 at a time when no response was achieved with vaccination alone. These results indicate that modulation of antigen-presenting cells may be a useful strategy for enhancing responsiveness to immunization.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Cancer Vaccines , Carcinoma, Renal Cell/prevention & control , Kidney Neoplasms/prevention & control , Lung Neoplasms/secondary , Membrane Glycoproteins/immunology , Adoptive Transfer , Animals , CD40 Ligand , Carcinoma, Renal Cell/immunology , Immune Tolerance , Kidney Neoplasms/immunology , Lung Neoplasms/prevention & control , Lymphocyte Transfusion , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics , Tumor Cells, Cultured
12.
Crit Rev Oncog ; 7(5-6): 433-56, 1996.
Article in English | MEDLINE | ID: mdl-9467666

ABSTRACT

Self-antigens are the most relevant and abundant antigens to which the host's immune system must be tolerant. Induction and maintenance of tolerance to self-antigens is mediated by several mechanisms that prevent inappropriate damage to normal tissues. However, these same mechanisms may impose potential barriers for the full development of effective immune responses against antigens expressed by tumors. A critical issue in tumor immunology is whether antigen presented by a progressively expanding tumor cell population results in T-cell tolerance. Utilizing a T cell receptor transgenic mice specific for a model tumor antigen expressed on a B-cell lymphoma, recently we have obtained direct evidence supporting the existence of tumor-induced antigen-specific tolerance. A better identification and understanding of the factor(s) involved in tumor-induced tolerance has clear implications for the development of novel cancer immunotherapies aimed at safely breaking tolerance, for example, releasing the brakes on antitumor immune responses while still limiting the induction of undesirable autoimmune responses.


Subject(s)
Immune Tolerance , Neoplasms/immunology , Tumor Escape , Animals , Antigens, Neoplasm , Humans , Immunosuppression Therapy , Lymphocytes/physiology , Mice , Signal Transduction
13.
Int J Cancer ; 60(5): 660-7, 1995 Mar 03.
Article in English | MEDLINE | ID: mdl-7860141

ABSTRACT

Peritoneal-exudate macrophages (PEM) from mammary-tumor-bearing mice have impaired cytotoxic activity against syngeneic and allogeneic tumor targets. The ability of PEM from normal and tumor-bearing mice to bind tumor targets was found to be similar in the presence or the absence of surrogate receptors, which enhanced the binding but not the killing of tumor targets by PEM from tumor-bearing mice, suggesting that other mechanisms are involved in their impaired cytolytic activity. Soluble and membrane-bound TNF-alpha, as well as H2O2, were found in higher amounts in PEM from tumor bearers upon stimulation with LPS, as compared with PEM from normal mice. However, tumor-bearers' macrophages displayed decreased capacity to produce and/or release nitric oxide, which could be reversed by the addition of increasing levels of IFN-gamma. These results indicate that the lack of macrophage cytotoxicity in mammary-tumor-bearing mice is related to impaired production and/or release of NO by these effector cells, possibly aggravated by the insufficient IFN-gamma production previously reported in these animals. Moreover, mammary-tumor progression results in dis-regulation of synthesis of macrophage-mediators, with over-production of molecules to which mammary-tumor cells are insensitive and deficient production of NO, the crucial molecule to which these cells appear to be highly sensitive.


Subject(s)
Macrophages, Peritoneal/immunology , Mammary Neoplasms, Experimental/immunology , Nitric Oxide/physiology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Cytotoxicity, Immunologic/drug effects , Endotoxins/pharmacology , Hydrogen Peroxide/metabolism , Immunoglobulin Fab Fragments/metabolism , Interferon-gamma/deficiency , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Nitric Oxide/biosynthesis , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , omega-N-Methylarginine
14.
J Exp Med ; 180(3): 945-58, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-8064242

ABSTRACT

Macrophages from mice bearing large D1-DMBA-3 mammary tumors have a decreased capacity to kill tumor targets. This effect is due to an impaired ability to produce nitric oxide (NO) in response to lipopolysaccharide (LPS) stimulation. Here we report that the DA-3 tumor cell line, derived from the in vivo adenocarcinoma D1-DMBA-3, produces a factor that inhibits both NO production/release and cytotoxicity of LPS-activated peritoneal exudate macrophages (PEM). However, other complex macrophage functions such as phagocytosis, superoxide production, mitochondrial dehydrogenase activity, and synthesis of proteins were not reduced by this factor. The NO inhibitor has been found to be lipid in nature. Lipid extracts from DA-3 cell culture supernatants were purified by repeated silica gel column chromatography. The active molecule was unambiguously characterized as phosphatidyl serine (PS) by fast atom bombardment tandem mass spectrometry. Preliminary results indicate a lack of induced NO synthase (iNOS) activity in the lysates of LPS-activated PEM pretreated with PS. The ubiquity of PS in the inner leaflet of biological membranes and its NO inhibitory property, suggest that this phospholipid may be one of the long elusive molecules responsible for regulating physiological levels of NO in the host and hence preventing cellular dysfunction and/or tissue damage. Furthermore, the possible overexpression and shedding of PS by DA-3 tumor cells may represent a novel mechanism to impair macrophage cytotoxicity, a host function that contributes to the protection against developing neoplasms.


Subject(s)
Mammary Neoplasms, Experimental/chemistry , Nitric Oxide/antagonists & inhibitors , Phosphatidylserines/isolation & purification , Animals , Female , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mass Spectrometry , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Phosphatidylserines/pharmacology , Type C Phospholipases/pharmacology
15.
Drugs Exp Clin Res ; 18 Suppl: 5-11, 1992.
Article in English | MEDLINE | ID: mdl-1305044

ABSTRACT

Ukrain is a semisynthetic drug with immunomodulatory properties, derived from Chelidonium majus L. alkaloids and thiophosphoric acid. The effect of this compound on the growth of Balb/c syngenic mammary adenocarcinoma was assessed. Intravenous, but not subcutaneous or intraperitoneal, administration of this drug was found to be effective in delaying tumour growth in an actual therapeutic protocol initiated five days after tumour implantation. No untoward side-effects were observed using these in vivo treatment modalities. The role of macrophages in the observed retardation of tumour development was investigated using peritoneal exudate macrophages (PEM) in cytotoxicity assays. In previous studies, the authors have found that PEM of mammary tumour bearing mice lose their capacity to kill a variety of tumour target cells including the in vitro cultured homologous tumour cells (DA-3). Pretreatment of PEM from normal mice with 2.5 microM Ukrain for 24 h followed by stimulation with either IFN-gamma or with LPS+IFN-gamma enhanced their cytotoxic activity. Treatment of PEM from tumour bearing mice with 2.5 microM Ukrain and LPS results in a reversal of their defective cytotoxic response against the DA-3 target cells. Furthermore, Ukrain alone, in the absence of a secondary signal, induced the activation of tumouricidal function of PEM from tumour bearing but not from normal mice. These data indicate that Ukrain's in vivo effects against the development of mammary tumours may be due, at least in part, to its ability to restore macrophage cytolytic function.


Subject(s)
Adenocarcinoma/prevention & control , Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Macrophages/drug effects , Mammary Neoplasms, Experimental/prevention & control , Adenocarcinoma/chemically induced , Animals , Berberine Alkaloids , Exudates and Transudates/cytology , Humans , In Vitro Techniques , Indicators and Reagents , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mammary Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phenanthridines , Transplantation, Homologous
16.
J Immunol ; 147(8): 2816-23, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-1918995

ABSTRACT

Peritoneal elicited macrophages (PEM) from mammary tumor-bearing mice have a decreased capacity to become cytotoxic against syngeneic, allogeneic, and xenogeneic target cells upon in vitro stimulation with LPS, as compared with PEM of normal mice. A regulatory mechanism other than PG release is suggested because the addition of both indomethacin and LPS to macrophage cultures from tumor-bearing mice caused no changes in their cytotoxic capability. Because tumor products have been implicated in the down-regulation of immune responses, we investigated whether pretreatment with supernatants from the tumor cell line DA-3, derived from the in vivo mammary adenocarcinoma D1-DMBA-3, affects the cytolytic capacity of macrophages. This treatment inhibits, in a dose-dependent fashion, the ability of stimulated normal PEM to kill target cells. Partial purification of DA-3 cell line supernatant showed that most of the inhibitory activity was exerted by factors with a molecular mass greater than 10 kDa and less than 30 kDa. However, slight inhibition could also be observed with fractions containing molecules less than 10 kDa. The data suggest that more than one factor released by the mammary tumor cells may be involved in the down-regulation of macrophage-mediated cytotoxicity. Because the DA-3 cells constitutively produce granulocyte-macrophage CSF (GM-CSF), which has a molecular mass of 27 kDa, we pretreated PEM from normal mice in vitro with rGM-CSF for 24 h. This resulted in a dose-dependent decrease in their capacity to kill tumor target cells upon LPS stimulation. Furthermore, PEM from normal mice injected with rGM-CSF for 25 days displayed a profound decrease in their cytolytic ability against DA-3 targets upon in vitro stimulation with increasing amounts of LPS. The pretreatment of PEM from normal mice with a combination of DA-3 cell supernatants and specific anti-GM-CSF partially neutralized the inhibitory effect of the DA-3 supernatant on macrophage tumoricidal capability. These results indicate that tumor-derived GM-CSF is an important factor involved in the decreased macrophage cytotoxicity during mammary adenocarcinoma progression.


Subject(s)
Adenocarcinoma/immunology , Cytotoxicity, Immunologic/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Mammary Neoplasms, Experimental/immunology , Adenocarcinoma/metabolism , Animals , Dinoprostone/biosynthesis , Down-Regulation , Interferon-gamma/pharmacology , Lipopolysaccharides , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...