Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 54(4): 1446-1456, 2019 04.
Article in English | MEDLINE | ID: mdl-30720069

ABSTRACT

Metastatic castration resistant prostate cancer (mCRPC) relapse due to acquired resistance to chemotherapy, such as docetaxel, remains a major threat to patient survival. Resistance of mCRPC to docetaxel can be associated with elevated levels of soluble clusterin (sCLU) and growth differentiation factor­15 (GDF­15). Any strategies aiming to modulate sCLU and/or GDF­15 in docetaxel­resistant prostate cancer cells present a therapeutic interest. The present study reports the cytotoxic effect of a nitric oxide donor, glyceryl trinitrate (GTN), on docetaxel­resistant mCRPC human cell lines and demonstrates that GTN displays greater inhibition of cell viability toward docetaxel­resistant mCRPC cells than on mCRPC cells. It is also demonstrated that GTN modulates the level of expression of clusterin (CLU) which is dependent of GDF­15, two markers associated with docetaxel resistance in prostate cancer. The results indicate that GTN represses the level of expression of the cytoprotective isoform of CLU (sCLU) and can increase the level of expression of the cytotoxic isoform (nuclear CLU) in docetaxel resistant cells. Furthermore, it was observed that GTN differentially regulates the level of the precursor form of GDF­15 between resistant and parental cells, and that recombinant GDF­15 can modulate the expression of CLU isoforms and counteract GTN­induced cytotoxicity in resistant cells. A link was established between GDF­15 and the expression of CLU isoforms. The present study thus revealed GTN as a potential therapeutic strategy to overcome docetaxel­resistant mCRPC.


Subject(s)
Clusterin/metabolism , Docetaxel/pharmacology , Drug Resistance, Neoplasm/drug effects , Nitroglycerin/administration & dosage , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Clusterin/genetics , Gene Expression Regulation, Neoplastic/drug effects , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Humans , Male , Nitroglycerin/pharmacology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Xenograft Model Antitumor Assays , Zebrafish
2.
Oncotarget ; 9(47): 28364-28378, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29983866

ABSTRACT

Neutrophils are known to possess both pro- and anti-tumor properties, a feature that could be related to the diversity and plasticity of these cells. Here we explored the hypothesis that under an appropriate environment and stimuli, neutrophils could induce an effective response against tumor cells. In a rat and mouse models, we show that a substantial amount of colon tumor associated-neutrophils (TAN) expressed the cytolytic enzyme granzyme B, which is absent in spleen or blood circulating neutrophils. This TAN population was also found into tumors of patients with colon cancer. Tumor neutrophil infiltration was correlated with an increase of chemokines known to attract neutrophils in both rat models and patients. These cells were involved in a Lipid A analog-mediated colon tumor regression. Mechanistically, treating the rats with the Lipid A analog triggered granzyme B release from neutrophils in tumor cell vicinity, which was correlated to tumor regression. Alteration of granzyme B function in tumor cells decreased the cytotoxic effect of Lipid A in rat and mouse models. Granzyme B expression in neutrophils could be induced by the lipid A analog but also by some of the cytokines that were detected in the tumor microenvironment. These results identify a subpopulation of neutrophils expressing granzyme B that can act as a key player of lipid A-mediated colon cancer regression in rat and mouse models and the molecular mechanisms involved may provide novel approaches for human therapeutic intervention.

3.
Cell Mol Life Sci ; 69(15): 2609-19, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22349263

ABSTRACT

Expression of the human inducible nitric oxide synthase (hiNOS) is generally undetectable in resting cells, but stimulation by a variety of signals including cytokines induces transcription in most cell types. The tight transcriptional regulation of the enzyme is a complex mechanism many aspects of which remain unknown. Here, we describe an octamer (Oct) element in hiNOS proximal promoter, located close to the TATA box. This site constitutively binds Oct-1 and its deletion abrogates cytokine-induced transcription, showing that it is indispensable though not sufficient for transcription. Increasing the distance between Oct and the TATA box by inserting inert DNA sequence inhibits transcription, and footprinting of this region shows no other protein binding in resting cells, suggesting an interaction between the two complexes. Chromatin immunoprecipitation assays detect the presence of Oct-1, RNA polymerase II and trimethyl K4 histone H3 on the proximal promoter in resting cells, confirming that the gene is primed for transcription before stimulation. RT-PCR of various fragments along the hiNOS gene shows that transcription is initiated in resting cells and this is inhibited by interference with Oct-1 binding to the proximal site of the promoter. We propose that, through interaction with the initiation complex, Oct-1 regulates hiNOS transcription by priming the gene for the rapid response required in an immune response.


Subject(s)
Nitric Oxide Synthase Type II/genetics , Octamer Transcription Factor-1/metabolism , Base Sequence , Cell Line , DNA Primers/genetics , HCT116 Cells , Humans , Models, Biological , Mutagenesis, Site-Directed , Octamer Transcription Factor-1/genetics , Promoter Regions, Genetic , TATA Box , Transcription, Genetic
4.
Ann N Y Acad Sci ; 1090: 188-202, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17384262

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) induces scattering, morphogenesis, and survival of epithelial cells through activation of the MET tyrosine kinase receptor. HGF/SF and MET are involved in normal development and tumor progression of many tissues and organs, including the mammary gland. In order to find target genes of HGF/SF involved in its survival function, we used an oligonucleotide microarray representing 1,920 genes known to be involved in apoptosis, transcriptional regulation, and signal transduction. MCF-10A human mammary epithelial cells were grown in the absence of serum and treated or not with HGF/SF for 2 h. Total RNA was reverse-transcribed to cDNA in the presence of fluorescent Cy3-dUTP or Cy5-dUTP to generate fluorescently labeled cDNA probes. Microarrays were performed and the ratios of Cy5/Cy3 fluorescence were determined. The expression of three apoptotic genes was modified by HGF/SF, with A20 being upregulated, and DAXX and SMAC being downregulated. These changes of expression were confirmed by real-time quantitative PCR. According to current-knowledge, A20 is antiapoptotic and SMAC is proapoptotic, while a pro- or antiapoptotic function of DAXX is controversial. The fact that HGF/SF upregulates an antiapoptotic gene (A20) and downregulates a proapoptotic gene (SMAC) is in agreement with its survival effect in MCF-10A cells. This study identified novel apoptotic genes regulated by HGF/SF, which can contribute to its survival effect.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/physiology , Hepatocyte Growth Factor/physiology , Mammary Glands, Human/metabolism , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Epithelial Cells/metabolism , Humans , Mammary Glands, Human/cytology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
5.
Bull Cancer ; 91(9): 705-12, 2004 Sep.
Article in French | MEDLINE | ID: mdl-15544996

ABSTRACT

NO is a molecule produced in different amounts by two types of enzymes, constitutive NO-synthases and inducible NO-synthase, the last one produce large amount of NO. In tumor outcome, its role may be either beneficial or detrimental due to its actions in the different steps of tumor growth and metastasis. This review deals with mammary tumors and the mechanisms lying behind NO effects. In human patients, increased amounts of NO were evidenced in blood, linked with the presence and activity of NO-synthase in breast tumors. Non-unequivocal correlations were established with tumor grade, invasiveness and metastatic potential. Studies in animal models have given hints to explain these discrepancies by the type of the involved NO-synthase, the amount of NO it produces, and its belonging to tumoral or stromal cells. Indeed, it was recently shown that NO produced by tumor cells inhibits, while NO produced by stromal cells facilitates tumor growth, at least in the model which was studied. On the one hand, NO toxicity against tumor cells is a well known phenomenon, but on the other hand, NO may promote tumor invasiveness due to its effect on extracellular matrix, and to its angiogenetic properties. So the role of NO in mammary tumor outcome is not clear-cut, and, at the present time, it cannot be ascribed a pronostic value in breast tumor. However, researches aimed at managing tumor cells to produce NO sufficient to induce their death may be fruitful since, be tumor targeting successful, no general toxicity would be encountered.


Subject(s)
Breast Neoplasms/metabolism , Nitric Oxide/metabolism , Animals , Apoptosis , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Capillary Permeability , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/therapy , Neoplasm Invasiveness , Neovascularization, Pathologic/etiology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III
6.
Carcinogenesis ; 25(9): 1559-65, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15059928

ABSTRACT

To study the role of nitric oxide (NO) in lung metastasis of breast carcinoma, we isolated two cell clones (H and J) from the parental EMT-6 murine breast carcinoma cell line, based on their differential NO production. In vitro, EMT-6 J cells, but not EMT-6H cells, constitutively expressed inducible NO synthase (NOS II) and secreted high levels of NO. IL-1beta increased NO production in both clones, and TNF-alpha had a synergistic effect on IL-1beta-induced NO production, but NO production by EMT-6 J cells was always higher than by EMT-6H cells. Proliferation, survival and adhesion to lung-derived endothelial cells of both clones were similar and were not affected by NO. In vivo, both clones similarly located in the lungs of syngeneic mice 48 h after injection. However, EMT-6H cells were significantly more tumorigenic than EMT-6 J cells as assessed at later time points. Injection of EMT-6 J cells and simultaneous treatment of mice with aminoguanidine (AG), a NOS II inhibitor, significantly increased tumour formation. Injection of EMT-6H and EMT-6 J cells into NOS II-deficient mice resulted in a significant survival increase as compared with wild-type animals. Simultaneous administration of AG increased the death rate of NOS II-deficient mice injected with EMT-6 J cells. These results demonstrate that: (i) NO does not influence the early stages of tumour metastasis to the lungs and (ii) NOS II expression in tumour cells reduces, while NOS II expression in host cells enhances, tumour nodule development. In conclusion, the cellular origin and the local NO production are critical in the metastatic process.


Subject(s)
Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide/physiology , Animals , Cell Adhesion , Cell Division , Cell Survival , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Inhibitors/pharmacology , Female , Guanidines/pharmacology , Indium Radioisotopes , Interleukin-1/pharmacology , Lung Neoplasms/enzymology , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Survival Rate , Tumor Cells, Cultured
7.
J Biol Chem ; 279(23): 23953-60, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15033982

ABSTRACT

Nitric oxide (NO) produced by inducible nitric-oxide synthase (NOSII) is mainly regulated at the transcriptional level by the nuclear factor-kappaB (NF-kappaB). In the present study, we further analyzed the role of NF-kappaB in the in vivo transcriptional regulation of NOSII gene by comparing two clones isolated from the EMT-6 mouse mammary cancer cell line. In response to interleukin (IL)-1beta or lipopolysaccharide (LPS), EMT-6 clone J (EMT-6J) cells produce 3-fold more NO than EMT-6 clone H (EMT-6H) cells, an effect correlated with enhanced activation of NF-kappaB in EMT-6J cells. In response to IL-1beta, the kinetics of degradation of NF-kappaB inhibitors IkappaB-alpha and IkappaB-beta, the nucleo-cytoplasmic shuttling of the transcription factor and its binding to a specific DNA sequence were similar in both clones. In contrast, an IL-1beta-induced phosphorylation of serine residues in NF-kappaB p65 subunit was observed in EMT-6J, but not in EMT-6H, cells. This IL-1beta-induced phosphorylation of p65 was specifically prevented by pretreatment of EMT-6J cells with the casein kinase II inhibitor DRB. Small interfering RNA-mediated depletion of casein kinase II-alpha subunit also decreased NF-kappaB transcriptional activity and NOSII gene transcription in IL-1beta and LPS-stimulated EMT-6J cells to the levels observed in EMT-6H cells treated in the same conditions. Altogether, these data indicate that casein kinase II-mediated phosphorylation of p65 subunit can enhance the transcriptional activity of NF-kappaB in vivo. This post-translational modification of the transcription factor can be responsible for increased NOSII gene transcription and NO production in tumor cells exposed to either IL-1beta or LPS.


Subject(s)
NF-kappa B/metabolism , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Animals , Blotting, Northern , Casein Kinase II , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA/metabolism , Enzyme Activation , Genes, Reporter , Immunoblotting , Interleukin-1/metabolism , Kinetics , Lipopolysaccharides/metabolism , Mice , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Serine/chemistry , Time Factors , Transcription Factor RelA , Transcriptional Activation , Transfection
8.
Ann N Y Acad Sci ; 1010: 100-3, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15033702

ABSTRACT

We investigated whether repression of JNK by hepatocyte growth factor/scatter factor (HGF/SF) in MDCK epithelial cells is linked to its ability to protect cells from apoptosis. To this purpose, cells were treated by TNF-alpha, a well-known inducer of JNK and of cell death, and the effects of HGF/SF were investigated under these conditions. We identified repression of JNK as a signaling target of HGF/SF for protection against TNF-alpha-induced cell death. This effect of HGF/SF occurs via the activation of the PI3K and MEK1 pathways.


Subject(s)
Apoptosis/physiology , Hepatocyte Growth Factor/pharmacology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Chromones/pharmacology , Dogs , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 4 , Morpholines/pharmacology , Urothelium
10.
Oncogene ; 21(15): 2309-19, 2002 Apr 04.
Article in English | MEDLINE | ID: mdl-11948414

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) induces scattering and morphogenesis of epithelial cells through the activation of the MET tyrosine kinase receptor. Although the activated MET receptor recruits a number of signaling proteins, little is known of the downstream signaling pathways activated by HGF/SF. In this study, we wished to examine the signaling pathway leading to activation of the ETS1 transcription factor. Using in vitro and in vivo kinase assays, we found that HGF/SF activates the ERK1 MAP kinase, leading to the phosphorylation of the threonine 38 residue of ETS1 within a putative MAP kinase phosphorylation site (PLLT38P). This threonine residue was neither phosphorylated by JNK1, nor by p38 MAP kinases and was required for the induction of transcriptional activity of ETS1 by HGF/SF. Using kinase and transcription assays, we further demonstrated that phosphorylation and activation of ETS1 occurs downstream of a RAS-RAF-MEK-ERK pathway. The functional involvement of this pathway in HGF/SF action was demonstrated using U0126, a pharmacological inhibitor of MEK, which blocked phosphorylation and activation of ETS1, RAS-dependent transcriptional responses, cell scattering and morphogenesis. These data demonstrated that ETS1 is a downstream target of HGF/SF acting through a RAS-RAF-MEK-ERK pathway and provides a signaling pathway leading to the regulation of gene expression by HGF/SF.


Subject(s)
Hepatocyte Growth Factor/pharmacology , MAP Kinase Signaling System , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Morphogenesis , Phosphorylation , Phosphothreonine/metabolism , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-ets , Proto-Oncogene Proteins c-raf/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/chemistry , Transcriptional Activation
11.
FASEB J ; 16(6): 631-3, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11919177

ABSTRACT

The human inducible nitric oxide synthase (iNOS or NOSII) gene is regulated through an extended and complex promoter. In this study, the transcriptional regulation of human NOSII is investigated in the human colon cell line HCT-8R. Stimulation with a cytokine mix (interferon-gamma, interleukin 1-beta, and tumor necrosis factor alpha) induces NOSII mRNA accumulation, as well as promoter activity in these cells. Several random deletions were performed within the proximal 7 kb of the promoter, which led to the identification of a region, whose deletion provokes a marked increase in transcriptional activity upon cytokine stimulation. Furthermore, this region is shown to repress a viral-driven luciferase construct, mainly at basal levels. An AP-1-like sequence present in this region that is specifically recognized by nuclear proteins is shown to be involved in the repressive effect. This element is capable of repressing a viral promoter, and its deletion augments cytokine-stimulated transcription. These findings are confirmed in various cell lines and suggest a general mechanism for the control of basal levels of NOSII expression, to avoid unnecessary toxicity under normal conditions.


Subject(s)
Gene Silencing , Nitric Oxide Synthase/genetics , Promoter Regions, Genetic , Response Elements , Base Sequence , Binding Sites , Consensus Sequence , Genes, Reporter , Humans , Models, Genetic , Nitric Oxide Synthase Type II , Repressor Proteins/metabolism , Sequence Deletion , Transcription Factor AP-1/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...