Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 869
1.
Oncogene ; 40(46): 6456-6468, 2021 11.
Article En | MEDLINE | ID: mdl-34608264

Cancer metastasis accounts for nearly 90% of all cancer deaths. Metastatic cancer progression requires both cancer cell migration to the site of the metastasis and subsequent proliferation after colonization. However, it has long been recognized that cancer cell migration and proliferation can be uncoupled; but the mechanism underlying this paradox is not well understood. Here we report that TNFAIP8 (tumor necrosis factor-α-induced protein 8), a "professional" transfer protein of phosphoinositide second messengers, promotes cancer cell migration or metastasis but inhibits its proliferation or cancer growth. TNFAIP8-deficient mice developed larger tumors, but TNFAIP8-deficient tumor cells completely lost their ability to migrate toward chemoattractants and were defective in colonizing lung tissues as compared to wild-type counterparts. Mechanistically, TNFAIP8 served as a cellular "pilot" of tumor cell migration by locally amplifying PI3K-AKT and Rac signals on the cell membrane facing chemoattractant; at the same time, TNFAIP8 also acted as a global inhibitor of tumor cell growth and proliferation by regulating Hippo signaling pathway. These findings help explain the migration-proliferation paradox of cancer cells that characterizes many cancers.


Apoptosis Regulatory Proteins/metabolism , Fibrosarcoma/pathology , Lung Neoplasms/pathology , Skin Neoplasms/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Diethylnitrosamine/adverse effects , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Methylcholanthrene/adverse effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
2.
Methods Mol Biol ; 1731: 235-245, 2018.
Article En | MEDLINE | ID: mdl-29318558

For decades, proteases have been associated with cancer progression due to the ability of some members of this large group of enzymes to degrade tumor cell surroundings, thereby facilitating cancer invasion and dissemination. However, the generation of mouse models deficient in proteases has revealed the existence of a great variety of functions among proteolytic enzymes in cancer biology, including important tumor-suppressive roles. Therefore, in this chapter, we describe methods to chemically induce different types of cancer (lung adenocarcinoma, hepatocellular carcinoma, oral and esophageal carcinoma, colorectal carcinoma, skin cancer, and fibrosarcoma) in genetically modified mouse models to efficiently evaluate the specific pro- or antitumoral function of proteases in cancer.


Carcinoma/genetics , Fibrosarcoma/genetics , Neoplasms, Experimental/genetics , Neoplasms/genetics , Peptide Hydrolases/genetics , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinoma/chemically induced , Carcinoma/pathology , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/pathology , Genetic Predisposition to Disease , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/chemically induced , Neoplasms/pathology , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
3.
Vet Clin North Am Small Anim Pract ; 48(2): 279-290, 2018 Mar.
Article En | MEDLINE | ID: mdl-29195924

Vaccines are important for providing protection from infectious diseases. Vaccination initiates a process that stimulates development of a robust and long-lived immune response to the disease agents in the vaccine. Side effects are sometimes associated with vaccination. These vary from development of acute hypersensitivity responses to vaccine components to local tissue reactions that are annoying but not significantly detrimental to the patient. The pathogenesis of these responses and the consequent clinical outcomes are discussed. Overstimulation of the immune response and the potential relationship to autoimmunity is evaluated in relation to genetic predisposition.


Cat Diseases/chemically induced , Dog Diseases/chemically induced , Vaccination/veterinary , Vaccines/adverse effects , Anaphylaxis/chemically induced , Anaphylaxis/veterinary , Animals , Arthus Reaction/chemically induced , Arthus Reaction/veterinary , Autoimmunity/drug effects , Autoimmunity/genetics , Cats , Dog Diseases/genetics , Dogs , Fibrosarcoma/chemically induced , Fibrosarcoma/veterinary , Horse Diseases/chemically induced , Horses , Immunity, Herd , Immunoglobulin E/immunology , Vaccination/adverse effects
4.
Int Immunopharmacol ; 38: 332-41, 2016 Sep.
Article En | MEDLINE | ID: mdl-27344639

Diagnosis of cancer and photothermal therapy using optoelectronic properties of noble metal nanoparticles (NPs) has established a new therapeutic approach for treating cancer. Here we address the intrinsic properties of noble metal NPs (gold and silver) as well as the mechanism of their potential antitumor activity. For this, the study addresses the functional characterization of tumor associated macrophages (TAMs) isolated from murine fibrosarcoma induced by a chemical carcinogen, 3-methylcholanthrene (MCA). We have previously shown antitumor activity of both gold nanoparticles (AuNPs) and silver nanoparticle (AgNPs) in vivo in a murine fibrosarcoma model. In the present study, it has been seen that AuNPs and AgNPs modulate the reactive oxygen species (ROS) and reactive nitrogen species (RNS) production, suppressing the antioxidant system of cells (TAMs). Moreover, the antioxidant-mimetic action of these NPs maintain the ROS and RNS levels in TAMs which act as second messengers to activate the proinflammatory signaling cascades. Thus, while there is a downregulation of tumor necrosis factor-α (TNF-α) and Interleukin-10 (IL-10) in the TAMs, the proinflammatory cytokine Interleukin-12 (IL-12) is upregulated resulting in a polarization of TAMs from M2 (anti-inflammatory) to M1 (pro-inflammatory) nature.


Fibrosarcoma/immunology , Macrophages/physiology , Metal Nanoparticles/administration & dosage , Animals , Cell Differentiation , Cells, Cultured , Fibrosarcoma/chemically induced , Gold/chemistry , Interleukin-10/metabolism , Interleukin-12/metabolism , Male , Metal Nanoparticles/chemistry , Methylcholanthrene/toxicity , Mice , Oxidative Stress , Phenotype , Reactive Oxygen Species/metabolism , Silver/chemistry , Tumor Necrosis Factor-alpha/metabolism
5.
Cell Rep ; 13(11): 2470-2479, 2015 Dec 22.
Article En | MEDLINE | ID: mdl-26686633

USP15 is a deubiquitinase that negatively regulates activation of naive CD4(+) T cells and generation of IFN-γ-producing T helper 1 (Th1) cells. USP15 deficiency in mice promotes antitumor T cell responses in a transplantable cancer model; however, it has remained unclear how deregulated T cell activation impacts primary tumor development during the prolonged interplay between tumors and the immune system. Here, we find that the USP15-deficient mice are hypersensitive to methylcholantrene (MCA)-induced fibrosarcomas. Excessive IFN-γ production in USP15-deficient mice promotes expression of the immunosuppressive molecule PD-L1 and the chemokine CXCL12, causing accumulation of T-bet(+) regulatory T cells and CD11b(+)Gr-1(+) myeloid-derived suppressor cells at tumor site. Mixed bone marrow adoptive transfer studies further reveals a T cell-intrinsic role for USP15 in regulating IFN-γ production and tumor development. These findings suggest that T cell intrinsic USP15 deficiency causes excessive production of IFN-γ, which promotes an immunosuppressive tumor microenvironment during MCA-induced primary tumorigenesis.


CD4-Positive T-Lymphocytes/metabolism , Interferon-gamma/metabolism , Th1 Cells/metabolism , Ubiquitin-Specific Proteases/genetics , Animals , Antibodies, Neutralizing/immunology , B7-H1 Antigen/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemokine CXCL12/metabolism , Fibrosarcoma/chemically induced , Fibrosarcoma/mortality , Fibrosarcoma/pathology , Forkhead Transcription Factors/metabolism , Interferon-gamma/immunology , Methylcholanthrene/toxicity , Mice , Mice, Knockout , Signal Transduction , Survival Rate , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Tumor Microenvironment , Ubiquitin-Specific Proteases/deficiency
6.
Int J Nanomedicine ; 10: 6331-8, 2015.
Article En | MEDLINE | ID: mdl-26504383

The present study aimed to assess the antitumor effect of glycosphingolipid-incorporated liposomes (glycosphingosomes) in combination with liposomal doxorubicin (Lip-Dox) in a mouse model of fibrosarcoma. Glycosphingosomes were prepared by incorporating glycosphingolipids isolated from Sphingomonas paucimobilis into the liposomes of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, cholesterol, and cardiolipin. Tumors were induced by administering dimethyl-α-benzanthracene, and tumor-bearing mice were treated with various formulations of Dox, including free Dox, Lip-Dox, or glycosphingosomes + Lip-Dox. Mice were observed for 90 days to monitor their survival and tumor size. Free Dox, but not Lip-Dox or a combination of glycosphingosomes and Lip-Dox, caused the substantial depletion of leukocytes and significantly increased the levels of lactate dehydrogenase and creatinine kinase in mice. Tumor-bearing mice treated with a combination of glycosphingosomes and Lip-Dox showed restricted tumor growth and increased survival when compared to those treated with free Dox or Lip-Dox. The results of the present study suggest that a combination of glycosphingosomes and Lip-Dox may prove to be very effective in the treatment of tumors.


1,2-Dipalmitoylphosphatidylcholine/analogs & derivatives , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Doxorubicin/analogs & derivatives , Fibrosarcoma/chemically induced , Fibrosarcoma/drug therapy , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Animals , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Fibrosarcoma/pathology , Mice , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Survival Analysis
7.
Oncotarget ; 6(28): 24649-59, 2015 Sep 22.
Article En | MEDLINE | ID: mdl-26433463

Foxp3(+) regulatory T cells (Tregs) are often highly enriched within the tumor-infiltrating T cell pool. Using a well-characterised model of carcinogen-induced fibrosarcomas we show that the enriched tumor-infiltrating Treg population comprises largely of CXCR3(+) T-bet(+) 'TH1-like' Tregs which are thymus-derived Helios(+) cells. Whilst IL-2 maintains homeostatic ratios of Tregs in lymphoid organs, we found that the perturbation in Treg frequencies in tumors is IL-2 independent. Moreover, we show that the TH1 phenotype of tumor-infiltrating Tregs is dispensable for their ability to influence tumor progression. We did however find that unlike Tconvs, the majority of intra-tumoral Tregs express the activation markers CD69, CD25, ICOS, CD103 and CTLA4 and are significantly more proliferative than Tconvs. Moreover, we have found that CD69(+) Tregs are more suppressive than their CD69- counterparts. Collectively, these data indicate superior activation of Tregs in the tumor microenvironment, promoting their suppressive ability and selective proliferation at this site.


Cell Proliferation , Fibrosarcoma/metabolism , Interleukin-2/metabolism , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Sarcoma, Experimental/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Methylcholanthrene , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Sarcoma, Experimental/chemically induced , Sarcoma, Experimental/genetics , Sarcoma, Experimental/immunology , Signal Transduction , T-Box Domain Proteins/deficiency , T-Box Domain Proteins/genetics , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Tumor Microenvironment
8.
Biochem Pharmacol ; 98(1): 110-8, 2015 Nov 01.
Article En | MEDLINE | ID: mdl-26296573

Epidemiologic studies depict a negative correlation between caffeine consumption and incidence of tumors in humans. The main pharmacological effects of caffeine are mediated by antagonism of the adenosine receptor, A2AR. Here, we examine whether the targeting of A2AR by caffeine plays a role in anti-tumor immunity. In particular, the effects of caffeine are studied in wild-type and A2AR knockout (A2AR(-/-)) mice. Tumor induction was achieved using the carcinogen 3-methylcholanthrene (3-MCA). Alternatively, tumor cells, comprised of 3-MCA-induced transformed cells or B16 melanoma cells, were inoculated into animal footpads. Cytokine release was determined in a mixed lymphocyte tumor reaction (MLTR). According to our findings, caffeine-consuming mice (0.1% in water) developed tumors at a lower rate compared to water-consuming mice (14% vs. 53%, respectively, p=0.0286, n=15/group). Within the caffeine-consuming mice, tumor-free mice displayed signs of autoimmune alopecia and pronounced leukocyte recruitment intocarcinogen injection sites. Similarly, A2AR(-/-) mice exhibited reduced rates of 3-MCA-induced tumors. In tumor inoculation studies, caffeine treatment resulted in inhibition of tumor growth and elevation in proinflammatory cytokine release over water-consuming mice, as depicted by MLTR. Addition of the adenosine receptor agonist, NECA, to MLTR resulted in a sharp decrease in IFNγ levels; this was reversed by the highly selective A2AR antagonist, ZM241385. Thus, immune response modulation through either caffeine or genetic deletion of A2AR leads to a Th1 immune profile and suppression of carcinogen-induced tumorigenesis. Taken together, our data suggest that the use of pharmacologic A2AR antagonists may hold therapeutic potential in diminishing the rate of cancer development.


Adenosine A2 Receptor Antagonists/pharmacology , Caffeine/pharmacology , Fibrosarcoma/chemically induced , Neoplasms, Experimental/chemically induced , Receptor, Adenosine A2A/metabolism , Animals , Cell Line, Tumor , Cyclopentanes/toxicity , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/prevention & control , Mice , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/prevention & control , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2A/genetics , Triazines/pharmacology , Triazoles/pharmacology
9.
J Immunol ; 195(2): 726-35, 2015 Jul 15.
Article En | MEDLINE | ID: mdl-26041539

Antitumor immunity is augmented by cytotoxic lymphodepletion therapies. Adoptively transferred naive and effector T cells proliferate extensively and show enhanced antitumor effects in lymphopenic recipients. Although the impact of lymphodepletion on transferred donor T cells has been well evaluated, its influence on recipient T cells is largely unknown. The current study demonstrates that both regulatory T cells (Tregs) and effector CD8(+) T cells from lymphopenic recipients play critical roles in the development of antitumor immunity after lymphodepletion. Cyclophosphamide (CPA) treatment depleted lymphocytes more efficiently than other cytotoxic agents; however, the percentage of CD4(+)CD25(+) Foxp3(+) Tregs was significantly increased in CPA-treated lymphopenic mice. Depletion of these chemoresistant Tregs following CPA treatment and transfer of naive CD4(+) T cells augmented the antitumor immunity and significantly suppressed tumor progression. Further analyses revealed that recipient CD8(+) T cells were responsible for this augmentation. Using Rag2(-/-) mice or depletion of recipient CD8(+) T cells after CPA treatment abrogated the augmentation of antitumor effects in CPA-treated reconstituted mice. The transfer of donor CD4(+) T cells enhanced the proliferation of CD8(+) T cells and the priming of tumor-specific CD8(+) T cells originating from the lymphopenic recipients. These results highlight the importance of the recipient cells surviving cytotoxic regimens in cancer immunotherapies.


Cytotoxins/pharmacology , Fibrosarcoma/therapy , Lymphocyte Depletion , Lymphopenia/therapy , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cisplatin/pharmacology , Cyclophosphamide/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance/immunology , Etoposide/pharmacology , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphopenia/chemically induced , Lymphopenia/immunology , Lymphopenia/pathology , Methylcholanthrene , Mice , Mice, Inbred C57BL , Mice, Knockout , Paclitaxel/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Tumor Cells, Cultured , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Whole-Body Irradiation , Gemcitabine
10.
Metallomics ; 7(7): 1155-62, 2015 Jul.
Article En | MEDLINE | ID: mdl-25927891

A TAT47-57 peptide was modified on the N-terminus by elongation with a 2,3-diaminopropionic acid residue and then by coupling of two histidine residues on its N-atoms. This branched peptide could bind to Ni under physiological conditions as a 1 : 1 complex. We demonstrated that the complex was quantitatively taken up by human fibrosarcoma cells, in contrast to Ni(2+) ions. Ni localization (especially at the nuclei) was confirmed by imaging using both scanning X-ray fluorescence microscopy and Newport Green fluorescence. A competitive assay with Newport Green showed that the latter displaced the peptide ligand from the Ni-complex. Ni(2+) delivered as a complex with the designed peptide induced substantially more DNA damage than when introduced as a free ion. The availability of such a construct opens up the way to investigate the importance of the nucleus as a target for the cytotoxicity, genotoxicity or carcinogenicity of Ni(2+).


Cell Nucleus/metabolism , Fibrosarcoma/chemically induced , Fibrosarcoma/metabolism , Nickel/metabolism , Nickel/toxicity , Peptide Fragments/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Carcinogens/chemistry , Carcinogens/metabolism , Carcinogens/toxicity , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/pathology , DNA Damage/drug effects , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Humans , Nickel/administration & dosage , Optical Imaging , Peptide Fragments/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry
11.
Mol Cells ; 38(2): 151-5, 2015.
Article En | MEDLINE | ID: mdl-25518925

Matrix metalloproteinase (MMP)-9 degrades type IV collagen in the basement membrane and plays crucial roles in several pathological implications, including tumorigenesis and inflammation. In this study, we analyzed the effect of flavonols on MMP-9 expression in phorbol-12-myristate-13-acetate (PMA)-induced human fibrosarcoma HT-1080 cells. Galangin and kaempferol efficiently decreased MMP-9 secretion, whereas fisetin only weakly decreased its secretion. Galangin and kaempferol did not affect cell viability at concentrations up to 30 µM. Luciferase reporter assays showed that galangin and kaempferol decrease transcription of MMP-9 mRNA. Moreover, galangin and kaempferol strongly reduce IκBα phosphorylation and significantly decrease JNK phosphorylation. These results indicate that galangin and kaempferol suppress PMA-induced MMP-9 expression by blocking activation of NF-κB and AP-1. Therefore, these flavonols could be used as chemopreventive agents to lower the risk of diseases involving MMP-9.


Fibrosarcoma/genetics , Flavonoids/pharmacology , Kaempferols/pharmacology , Matrix Metalloproteinase 9/genetics , Tetradecanoylphorbol Acetate/analogs & derivatives , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Fibrosarcoma/chemically induced , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic , Humans , I-kappa B Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/toxicity
12.
Immunology ; 145(1): 94-104, 2015 May.
Article En | MEDLINE | ID: mdl-25495686

The frequency of CD4(+)  Foxp3(+) regulatory T (Treg) cells is often significantly increased in the blood of tumour-bearing mice and people with cancer. Moreover, Treg cell frequencies are often higher in tumours compared with blood and lymphoid organs. We wished to determine whether certain chemokines expressed within the tumour mass selectively recruit Treg cells, thereby contributing to their enrichment within the tumour-infiltrating lymphocyte pool. To achieve this goal, the chemokine profile of carcinogen-induced fibrosarcomas was determined, and the chemokine receptor expression profiles of both CD4(+)  Foxp3(-) and CD4(+)  Foxp3(+) T cells were compared. These analyses revealed that the tumours are characterized by expression of inflammatory chemokines (CCL2, CCL5, CCL7, CCL8, CCL12, CXCL9, CXCL10 and CX3CL1), reflected by an enrichment of activated Foxp3(-) and Foxp3(+) T cells expressing T helper type 1-associated chemokine receptors. Notably, we found that CXCR3(+) T cells were significantly enriched in the tumours although curiously we found no evidence that CXCR3 was required for their recruitment. Instead, CXCR3 marks a population of activated Foxp3(-) and Foxp3(+) T cells, which use multiple and overlapping ligand receptor pairs to guide their migration to tumours. Collectively, these data indicate that enrichment of Foxp3(+) cells in tumours characterized by expression of inflammatory chemokines, does not occur via a distinct chemokine axis, thus selective chemokine blockade is unlikely to represent a meaningful therapeutic strategy for preventing Treg cell accumulation in tumours.


Carcinogens/toxicity , Chemokines/immunology , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cell Movement/immunology , Chemokines/genetics , Female , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Humans , Methylcholanthrene/toxicity , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology
13.
PLoS One ; 9(11): e112415, 2014.
Article En | MEDLINE | ID: mdl-25384044

Tumor recognition by immune effector cells is mediated by antigen receptors and a variety of adhesion and costimulatory molecules. The evidence accumulated since the identification of CD155 and CD112 as ligands for DNAM-1 in humans and mice has suggested that the interactions between DNAM-1 and its ligands play an important role in T cell- and natural killer (NK) cell-mediated recognition and lysis of tumor cells. We have previously demonstrated that methylcholanthrane (MCA) accelerates tumor development in DNAM-1-deficient mice, and the Cd155 level on MCA-induced tumors is significantly higher in DNAM-1-deficient mice than in wild-type (WT) mice. By contrast, Cd112 expression on the tumors is similar in WT and DNAM-1-deficient mice, suggesting that CD155 plays a major role as a DNAM-1 ligand in activation of T cells and NK cells for tumor immune surveillance. To address this hypothesis, we examined MCA-induced tumor development in CD155-deficient mice. Unexpectedly, we observed no significant difference in tumor development between WT and CD155-deficient mice. Instead, we found that Cd112 expression was significantly higher in the MCA-induced tumors of CD155-deficient mice than in those of WT mice. We also observed higher expression of DNAM-1 and lower expression of an inhibitory receptor, TIGIT, on CD8+ T cells in CD155-deficient mice. These results suggest that modulation of the expression of receptors and CD112 compensates for CD155 deficiency in immune surveillance against MCA-induced tumors.


Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Fibrosarcoma/chemically induced , Fibrosarcoma/pathology , Methylcholanthrene/toxicity , Neoplasm Proteins/genetics , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Cytokines/genetics , Cytokines/metabolism , Fibrosarcoma/genetics , Humans , Interleukin-2 Receptor beta Subunit/genetics , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nectins , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , T-Lymphocytes/metabolism
14.
Immunol Lett ; 158(1-2): 159-66, 2014.
Article En | MEDLINE | ID: mdl-24406503

Immune suppression is well documented during tumor progression, which includes loss of effect of T cells and expansion of T regulatory (Treg) cells. IL-7 plays a key role in the proliferation, survival and homeostasis of T cells and displays a potent antitumor activity in vivo. In the present study, we investigated the antitumor effect of IL-7 in Meth A model. IL-7 inhibited tumor growth and prolonged the survival of tumor-bearing mice with corresponding increases in the frequency of CD4 and CD8 T cells, Th1 (CD4(+)IFN-γ(+)), Tc1 (CD8(+)IFN-γ(+)) and T cells cytolytic activity against Meth A cells. Neutralization of CD4 or CD8 T cells reversed the antitumor benefit of IL-7. Furthermore, IL-7 decreased regulatory T Foxp3 as well as cells suppressive activity with a reciprocal increase in SMAD7. In addition, we observed an increase of the serum concentrations of IL-6 and IFN-γ, and a significant decrease of TGF-ß and IL-10 after IL-7 treatment. Taken together, these results indicate that IL-7 augments T cell-mediated antitumor immunity and improves the effect of antitumor in Meth A model.


CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cytokines/biosynthesis , Fibrosarcoma/therapy , Interleukin-7/administration & dosage , Skin Neoplasms/therapy , T-Lymphocytes, Regulatory/drug effects , Animals , Antibodies, Blocking/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Growth Processes/drug effects , Cell Line, Tumor , Cytokines/blood , Cytokines/genetics , Cytotoxicity, Immunologic/drug effects , Disease Models, Animal , Female , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Gene Expression Regulation/drug effects , Humans , Lymphocyte Activation/drug effects , Methylcholanthrene/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Skin Neoplasms/chemically induced , Skin Neoplasms/immunology , Smad7 Protein/genetics , Smad7 Protein/metabolism , T-Lymphocytes, Regulatory/immunology
15.
Oncotarget ; 5(24): 12715-27, 2014 Dec 30.
Article En | MEDLINE | ID: mdl-25587030

Phorbol ester (PMA or TPA), a tumor promoter, can cause either proliferation or cell cycle arrest, depending on cellular context. For example, in SKBr3 breast cancer cells, PMA hyper-activates the MEK/MAPK pathway, thus inducing p21 and cell cycle arrest. Here we showed that PMA-induced arrest was followed by conversion to cellular senescence (geroconversion). Geroconversion was associated with active mTOR and S6 kinase (S6K). Rapamycin suppressed geroconversion, maintaining quiescence instead. In this model, PMA induced arrest (step one of a senescence program), whereas constitutively active mTOR drove geroconversion (step two). Without affecting Akt phosphorylation, PMA increased phosphorylation of S6K (T389) and S6 (S240/244), and that was completely prevented by rapamycin. Yet, T421/S424 and S235/236 (p-S6K and p-S6, respectively) phosphorylation became rapamycin-insensitive in the presence of PMA. Either MEK or mTOR was sufficient to phosphorylate these PMA-induced rapamycin-resistant sites because co-treatment with U0126 and rapamycin was required to abrogate them. We next tested whether activation of rapamycin-insensitive pathways would shift quiescence towards senescence. In HT-p21 cells, cell cycle arrest was caused by IPTG-inducible p21 and was spontaneously followed by mTOR-dependent geroconversion. Rapamycin suppressed geroconversion, whereas PMA partially counteracted the effect of rapamycin, revealing the involvement of rapamycin-insensitive gerogenic pathways. In normal RPE cells arrested by serum withdrawal, the mTOR/pS6 pathway was inhibited and cells remained quiescent. PMA transiently activated mTOR, enabling partial geroconversion. We conclude that PMA can initiate a senescent program by either inducing arrest or fostering geroconversion or both. Rapamycin can decrease gero-conversion by PMA, without preventing PMA-induced arrest. The tumor promoter PMA is a gero-promoter, which may be useful to study aging in mammals.


Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Carcinogens/pharmacology , Cellular Senescence/drug effects , Fibrosarcoma/chemically induced , Fibrosarcoma/pathology , Tetradecanoylphorbol Acetate/pharmacology , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Interactions , Female , Fibrosarcoma/metabolism , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation , Sirolimus/pharmacology
16.
Immunol Cell Biol ; 91(7): 451-60, 2013 Aug.
Article En | MEDLINE | ID: mdl-23817579

Toll-like receptors (TLRs) enable metazoans to mount effective innate immune responses to microbial and viral pathogens, as well as to endogenous host-derived ligands. It is understood that genetic background of the host can influence TLR responsiveness, altering susceptibility to pathogen infection, autoimmunity and cancer. Macrophage stimulatory protein (MSP), which activates the receptor tyrosine kinase recepteur d'origine nantais (RON), promotes key macrophage functions such as motility and phagocytic activity. MSP also acts via RON to modulate signaling by TLR4, which recognizes a range of pathogen or endogenous host-derived molecules. Here, we show that RON exerts divergent control over TLR4 activity in macrophages from different mouse genetic backgrounds. RON potently modulated the TLR4 response in macrophages from M2-prone FVB mice, as compared with M1-skewed C57Bl6 mice. Moreover, global expression analysis revealed that RON suppresses the TLR4-dependent type-I interferon gene signature only in FVB macrophages. This leads to attenuated production of the potent inflammatory mediator, tumor necrosis factor-α. Eliminating RON kinase activity markedly decreased carcinogen-mediated tumorigenesis in M2/Th2-biased FVB mice. We propose that host genetic background influences RON function, thereby contributing to the variability in TLR4 responsiveness in rodents and, potentially, in humans. These findings provide novel insight into the complex interplay between genetic context and immune function.


Fibrosarcoma/immunology , Macrophages, Peritoneal/immunology , Papilloma/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/immunology , Toll-Like Receptor 4/immunology , 9,10-Dimethyl-1,2-benzanthracene/administration & dosage , Animals , Carcinogenesis , Cell Movement/drug effects , Fibrosarcoma/chemically induced , Fibrosarcoma/genetics , Genotype , Hepatocyte Growth Factor/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Methylcholanthrene/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Papilloma/chemically induced , Papilloma/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Th1-Th2 Balance , Transcriptional Activation/genetics , Transcriptome
17.
J Environ Pathol Toxicol Oncol ; 31(2): 109-19, 2012.
Article En | MEDLINE | ID: mdl-23216636

This work reports the chemopreventive property of hydroalcoholic extract of the Trichosanthes dioica root (TDA) against 3-methylcholanthrene (3-MC)-induced carcinogenesis in Swiss albino mice. TDA was administered orally at 2 and 4 mg/kg for 45 days after 24 hours of a single subcutaneous administration of 3-MC (200 µg) in mice. The mice were observed for 15 weeks to record tumor incidence (fibrosarcoma) and survival. After 15 weeks the mice were killed for the evaluation of hematological profiles and hepatic biochemical parameters viz lipid peroxidation, reduced glutathione, glutathione-S-transferase, superoxide dismutase, and catalase. TDA treatment markedly reduced tumor incidence and prolonged the life span of sarcoma-bearing mice compared with 3-MC control mice. Hematological profiles of TDA-treated mice were restored significantly to normal levels. TDA treatment significantly modulated the liver biochemical parameters compared with 3-MC control. Therefore, TDA possesses remarkable cancer chemopreventive efficacy plausibly mediated by multiple mechanisms in Swiss albino mice.


Fibrosarcoma/chemically induced , Fibrosarcoma/prevention & control , Liver Neoplasms/chemically induced , Liver Neoplasms/prevention & control , Methylcholanthrene/adverse effects , Plant Extracts/therapeutic use , Plant Roots , Trichosanthes , Animals , Catalase/metabolism , Chemoprevention/methods , Disease Models, Animal , Fibrosarcoma/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Lipid Peroxidation/drug effects , Liver Neoplasms/metabolism , Male , Mice , Phytotherapy/methods , Plant Extracts/pharmacology , Superoxide Dismutase/metabolism , Treatment Outcome
18.
Pediatrics ; 130(4): e1019-25, 2012 Oct.
Article En | MEDLINE | ID: mdl-22945410

Congenital fibrosarcoma (CFS) is a rare fibrous tissue malignancy that usually presents in the first few years of life. It is unique among human sarcomas in that it has an excellent prognosis. We describe a temporal clustering of a number of cases of CFS and investigate the possible associated prenatal risk factors. The Pediatric Environmental History, a questionnaire developed in our clinic that is instrumental in determining environmental risk factors for tumor-related disease, was essential in documenting the presence or absence of risk factors considered as human carcinogens. We found a history of exposure to petroleum products in four cases of CFS that occurred at a greater than expected rate in a short time frame-an apparent cancer cluster. We call attention to the possibility that exposure to petroleum products raises the risk of developing CFS. While future studies should focus on systematic investigation of CFS and its underlying mechanisms, this report suggests the need for proactive measures to avoid exposure to solvents and petroleum products during pregnancy.


Carcinogens, Environmental/toxicity , Fibrosarcoma/chemically induced , Maternal Exposure/adverse effects , Petroleum/toxicity , Retroperitoneal Neoplasms/chemically induced , Soft Tissue Neoplasms/chemically induced , Thigh , Female , Fibrosarcoma/congenital , Humans , Infant, Newborn , Male , Paternal Exposure/adverse effects , Retroperitoneal Neoplasms/congenital , Soft Tissue Neoplasms/congenital , Spain , Surveys and Questionnaires
19.
Cancer Res ; 72(22): 5721-32, 2012 Nov 15.
Article En | MEDLINE | ID: mdl-22986739

The NLRP3 inflammasome acts as a danger signal sensor that triggers and coordinates the inflammatory response upon infectious insults or tissue injury and damage. However, the role of the NLRP3 inflammasome in natural killer (NK) cell-mediated control of tumor immunity is poorly understood. Here, we show in a model of chemical-induced carcinogenesis and a series of experimental and spontaneous metastases models that mice lacking NLRP3 display significantly reduced tumor burden than control wild-type (WT) mice. The suppression of spontaneous and experimental tumor metastases and methylcholanthrene (MCA)-induced sarcomas in mice deficient for NLRP3 was NK cell and IFN-γ-dependent. Focusing on the amenable B16F10 experimental lung metastases model, we determined that expression of NLRP3 in bone marrow-derived cells was necessary for optimal tumor metastasis. Tumor-driven expansion of CD11b(+)Gr-1(intermediate) (Gr-1(int)) myeloid cells within the lung tumor microenvironment of NLRP3(-/-) mice was coincident with increased lung infiltrating activated NK cells and an enhanced antimetastatic response. The CD11b(+)Gr-1(int) myeloid cells displayed a unique cell surface phenotype and were characterized by their elevated production of CCL5 and CXCL9 chemokines. Adoptive transfer of this population into WT mice enhanced NK cell numbers in, and suppression of, B16F10 lung metastases. Together, these data suggested that NLRP3 is an important suppressor of NK cell-mediated control of carcinogenesis and metastases and identify CD11b(+)Gr-1(int) myeloid cells that promote NK cell antimetastatic function.


Carrier Proteins/immunology , Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , CD11b Antigen/immunology , Chemokine CCL5/immunology , Chemokine CXCL9/immunology , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Inflammasomes/immunology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Male , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Methylcholanthrene , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/secondary , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology
20.
Cancer Res ; 72(16): 3987-96, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22869585

Cancer immunoediting, the process by which the immune system controls tumor growth and shapes tumor immunogenicity, consists of 3 stages: elimination, equilibrium, and escape. The molecular mechanisms that underlie the equilibrium phase, during which the immune system maintains tumor dormancy, remain incompletely defined. Here, we investigated the length of the equilibrium phase during immune control of methylcholanthrene (MCA)-induced or p53 mutant cancers and showed the critical and opposing roles of interleukin (IL)-23 and IL-12 in maintaining cancer cells in a state of immune-mediated dormancy. Inhibition of IL-23p19 was shown to reduce the malignant potential of lesions established by MCA inoculation, whereas inhibition of IL-12/23p40 enhanced tumor outgrowth. Furthermore, agonistic anti-CD40 antibody treatment mimicked the effects of anti-IL-23p19 monoclonal antibody treatment. Other cytokines such as IL-4, IL-17, TNF, and IFNαß, which are known to play important roles either in MCA tumorigenesis or in the elimination phase of cancer immunoediting, did not play critical roles in maintaining the equilibrium phase. Taken together, our findings show opposing roles for IL-23 and IL-12 in determining the outgrowth versus dormancy of occult neoplasia and suggest a potential long-term danger in using IL-12/23p40 antibodies for treating human autoimmune inflammatory disorders.


Interleukin-12/immunology , Interleukin-23/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Fibrosarcoma/chemically induced , Fibrosarcoma/immunology , Fibrosarcoma/pathology , Interferon-gamma/immunology , Lymphocyte Activation , Male , Methylcholanthrene , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology
...