bay-11-7082 and Prostatic-Neoplasms

bay-11-7082 has been researched along with Prostatic-Neoplasms* in 12 studies

Other Studies

12 other study(ies) available for bay-11-7082 and Prostatic-Neoplasms

ArticleYear
Hsp-27 and NF-κB pathway is associated with AR/AR-V7 expression in prostate cancer cells.
    Gene, 2019, May-20, Volume: 697

    In the present study, NF-κB inhibitor BAY 11-7082 and/or Hsp-27 inhibitor KRIBB-3 agents were used to investigate the molecular mechanisms mediating androgen receptor expression on prostate cancer cell lines. The decrease observed in androgen receptor and p65 expressions, particularly at 48 h, in parallel with the decrease in the phosphorylation of the p-IKK α/β and p-Hsp-27 proteins in the LNCaP cells, indicated that androgen receptor inactivation occurred after the inhibition of the NF-κB and Hsp-27. In 22Rv1 cells, androgen receptor variant-7 was also observed to be decreased in the combined dose of 48 h. The association of this decrease with the decrease in androgen receptor and p65 expressions is a supportive result for the role of NF-κB signaling in the formation of androgen receptor variant. In androgen receptor variant-7 siRNA treatment in 22Rv1 cell lines, decrease of expression of androgen receptor variant-7 as well as decrease of expression of androgen receptor and p65 were observed. The decrease statistically significant in androgen receptor and p65 expressions was even greater when siRNA treatment was followed with low dose and time (6 h) combined treatment after transfection. We also showed that increased Noxa and decreased Bcl-2 protein level, indicated that apoptotic induction after this combination. In conclusion, inhibition of NF-κB and Hsp-27 is also important, along with therapies for androgen receptor variant-7 inhibition.

    Topics: Anisoles; Apoptosis; Cell Line, Tumor; Heat-Shock Proteins; HSP27 Heat-Shock Proteins; Humans; I-kappa B Kinase; Isoxazoles; Male; Molecular Chaperones; NF-kappa B; Nitriles; Prostatic Neoplasms; Receptors, Androgen; Signal Transduction; Sulfones

2019
NF-κB inhibition promotes apoptosis in androgen-independent prostate cancer cells by the photothermal effect via the IκBα/AR signaling pathway.
    Biomaterials science, 2019, May-28, Volume: 7, Issue:6

    The photothermal response of nanomaterials provides a basis for many biomedical applications, including diagnosis (e.g., biosensor and photoacoustic imaging) and treatment (e.g., drug delivery and photothermal therapy). The use of nano-materials for cancer phototherapy (solid tumor ablation) can cause cell necrosis and apoptosis. However, photothermal effects using the same material can differ among tumor cell types, and the molecular mechanisms underlying these differences are not clear. We used polydopamine (PDA)-coated branched Au-Ag nanoparticles (Au-Ag@PDA NPs) for the photothermal treatment of two prostate cancer cell lines. The therapeutic effect was evaluated by CCK8, flow cytometry, and expression analyses of related genes by western blotting. Photothermal therapy resulted in oxidative stress in prostate cancer cells and activated the mitochondrial-related apoptosis pathway, increasing the Bax expression. In addition, we observed a greater photothermal treatment effect on the androgen-dependent cells LNCaP than the androgen-independent cells DU145. Pretreatment with an inhibitor of the NF-κB signaling pathway (BAY 11-7082) enhanced the expression of BAX in the DU145 cells and increased the sensitivity of the cells to the heat treatment of Au-Ag@PDA NPs both in vitro and in vivo. Our findings explain the differences in the observed effects of photothermal therapy and provide the direction for further improvements to this strategy.

    Topics: Apoptosis; bcl-2-Associated X Protein; Cell Line, Tumor; Gene Expression Regulation, Neoplastic; Gold; Hot Temperature; Humans; Indoles; Male; Metal Nanoparticles; Mitochondria; NF-kappa B; NF-KappaB Inhibitor alpha; Nitriles; Oxidative Stress; Phototherapy; Polymers; Prostatic Neoplasms; Receptors, Androgen; Signal Transduction; Silver; Sulfones

2019
NFκB-mediated CXCL1 production in spinal cord astrocytes contributes to the maintenance of bone cancer pain in mice.
    Journal of neuroinflammation, 2014, Mar-01, Volume: 11

    Bone cancer pain (BCP) is one of the most disabling factors in patients suffering from primary bone cancer or bone metastases. Recent studies show several chemokines (for example, CCL2, CXCL10) in the spinal cord are involved in the pathogenesis of BCP. Here we investigated whether and how spinal CXCL1 contributes to BCP.. Mouse prostate tumor cell line, RM-1 cells were intramedullary injected into the femur to induce BCP. The mRNA expression of CXCL1 and CXCR2 was detected by quantitative real-time PCR. The protein expression and distribution of CXCL1, NFκB, and CXCR2 was examined by immunofluorescence staining and western blot. The effect of CXCL1 neutralizing antibody, NFκB antagonist, and CXCR2 antagonist on pain hypersensitivity was checked by behavioral testing.. Intramedullary injection of RM-1 cells into the femur induced cortical bone damage and persistent (>21 days) mechanical allodynia and heat hyperalgesia. Tumor cell inoculation also produced CXCL1 upregulation in activated astrocytes in the spinal cord for more than 21 days. Inhibition of CXCL1 by intrathecal administration of CXCL1 neutralizing antibody at 7 days after inoculation attenuated mechanical allodynia and heat hyperalgesia. In cultured astrocytes, TNF-α induced robust CXCL1 expression, which was dose-dependently decreased by NFκB inhibitor. Furthermore, inoculation induced persistent NFκB phosphorylation in spinal astrocytes. Intrathecal injection of NFκB inhibitor attenuated BCP and reduced CXCL1 increase in the spinal cord. Finally, CXCR2, the primary receptor of CXCL1, was upregulated in dorsal horn neurons after inoculation. Inhibition of CXCR2 by its selective antagonist SB225002 attenuated BCP.. NFκB mediates CXCL1 upregulation in spinal astrocytes in the BCP model. In addition, CXCL1 may be released from astrocytes and act on CXCR2 on neurons in the spinal cord and be involved in the maintenance of BCP. Inhibition of the CXCL1 signaling may provide a new therapy for BCP management.

    Topics: Animals; Animals, Newborn; Antibodies; Astrocytes; Bone Neoplasms; Cell Line, Tumor; Cells, Cultured; Cerebral Cortex; Chemokine CXCL1; Disease Models, Animal; Enzyme Inhibitors; Gene Expression Regulation, Neoplastic; Male; Mice; Mice, Inbred C57BL; Neoplasm Transplantation; NF-kappa B; Nitriles; Pain; Prostatic Neoplasms; Spinal Cord; Sulfones

2014
Induction of steroid sulfatase expression in PC-3 human prostate cancer cells by insulin-like growth factor II.
    Toxicology letters, 2013, Nov-25, Volume: 223, Issue:2

    Human steroid sulfatase (STS) plays an important role in regulating the formation of biologically active estrogens and may be a promising target for treating estrogen-mediated carcinogenesis. The molecular mechanism of STS gene expression, however, is still not clear. Growth factors are known to increase STS activity but the changes in STS expression have not been completely understood. To determine whether insulin-like growth factor (IGF)-II can induce STS gene expression, the effects of IGF-II on STS expression were studied in PC-3 human prostate cancer cells. RT-PCR and Western blot analysis showed that IGF-II treatment significantly increased the expression of STS mRNA and protein in concentration- and time-dependent manners. To understand the signaling pathway by which IGF-II induces STS gene expression, the effects of specific PI3-kinase/Akt and NF-κB inhibitors were determined. When the cells were treated with IGF-II and PI3-kinase/Akt inhibitors, such as LY294002, wortmannin, or Akt inhibitor IV, STS expression induced by IGF-II was significantly blocked. Moreover, we found that NF-κB inhibitors, such as MG-132, bortezomib, Bay 11-7082 or Nemo binding domain (NBD) binding peptide, also strongly prevented IGF-II from inducing STS gene expression. We assessed whether IGF-II activates STS promoter activity using transient transfection with a luciferase reporter. IGF-II significantly stimulated STS reporter activity. Furthermore, IGF-II induced expression of 17β-hydroxysteroid dehydrogenase (HSD) 1 and 3, whereas it reduced estrone sulfotransferase (EST) gene expression, causing enhanced estrone and β-estradiol production. Taken together, these results strongly suggest that IGF-II induces STS expression via a PI3-kinase/Akt-NF-κB signaling pathway in PC-3 cells and may induce estrogen production and estrogen-mediated carcinogenesis.

    Topics: 17-Hydroxysteroid Dehydrogenases; Androstadienes; Boronic Acids; Bortezomib; Cell Line, Tumor; Chromones; Enzyme Inhibitors; Gene Expression Regulation, Enzymologic; Humans; Insulin-Like Growth Factor II; Leupeptins; Male; Morpholines; NF-kappa B; Nitriles; Phosphatidylinositol 3-Kinases; Phosphoinositide-3 Kinase Inhibitors; Polymerase Chain Reaction; Prostatic Neoplasms; Proto-Oncogene Proteins c-akt; Pyrazines; Sequence Analysis, DNA; Signal Transduction; Steryl-Sulfatase; Sulfones; Sulfotransferases; Wortmannin

2013
ER stress drives Lipocalin 2 upregulation in prostate cancer cells in an NF-κB-dependent manner.
    BMC cancer, 2011, Jun-07, Volume: 11

    Tumor cells adapt to endoplasmic reticulum (ER) stress through a set of conserved intracellular pathways, as part of a process termed the unfolded protein response (UPR). The expression of UPR genes/proteins correlates with increasing progression and poor clinical outcome of several tumor types, including prostate cancer. UPR signaling can activate NF-κB, a master regulator of transcription of pro-inflammatory, tumorigenic cytokines. Previous studies have shown that Lipocalin 2 (Lcn2) is upregulated in several epithelial cancers, including prostate cancer, and recently Lcn2 was implicated as a key mediator of breast cancer progression. Here, we hypothesize that the tumor cell UPR regulates Lcn2 production.. We interrogated Lcn2 regulation in murine and human prostate cancer cells undergoing pharmacological and physiological ER stress, and tested UPR and NF-κB dependence by using pharmacological inhibitors of these signaling pathways.. Induction of ER stress using thapsigargin (Tg), a canonical pharmacologic ER stress inducer, or via glucose deprivation, a physiologic ER stressor present in the tumor microenvironment, upregulates LCN2 production in murine and human prostate cancer cells. Inhibition of the UPR using 4-phenylbutyric acid (PBA) dramatically decreases Lcn2 transcription and translation. Inhibition of NF-κB in prostate cancer cells undergoing Tg-mediated ER stress by BAY 11-7082 abrogates Lcn2 upregulation.. We conclude that the UPR activates Lcn2 production in prostate cancer cells in an NF-κB-dependent manner. Our results imply that the observed upregulation of Lipocalin 2 in various types of cancer cells may be the direct consequence of concomitant UPR activation, and that the ER stress/Lipocalin 2 axis is a potential new target for intervention in cancer progression.

    Topics: Acute-Phase Proteins; Adenocarcinoma; Animals; Cell Line, Tumor; Endoplasmic Reticulum; Gene Expression Regulation, Neoplastic; Glucose; Humans; Lipocalin-2; Lipocalins; Male; Mice; Neoplasm Proteins; NF-kappa B; Nitriles; Oncogene Proteins; Phenylbutyrates; Prostatic Neoplasms; Protein Biosynthesis; Proto-Oncogene Proteins; Sulfones; Thapsigargin; Transcription, Genetic; Tunicamycin; Unfolded Protein Response; Up-Regulation

2011
Characterisation and manipulation of docetaxel resistant prostate cancer cell lines.
    Molecular cancer, 2011, Oct-07, Volume: 10

    There is no effective treatment strategy for advanced castration-resistant prostate cancer. Although Docetaxel (Taxotere®) represents the most active chemotherapeutic agent it only gives a modest survival advantage with most patients eventually progressing because of inherent or acquired drug resistance. The aims of this study were to further investigate the mechanisms of resistance to Docetaxel. Three Docetaxel resistant sub-lines were generated and confirmed to be resistant to the apoptotic and anti-proliferative effects of increasing concentrations of Docetaxel.. The resistant DU-145 R and 22RV1 R had expression of P-glycoprotein and its inhibition with Elacridar partially and totally reversed the resistant phenotype in the two cell lines respectively, which was not seen in the PC-3 resistant sublines. Resistance was also not mediated in the PC-3 cells by cellular senescence or autophagy but multiple changes in pro- and anti-apoptotic genes and proteins were demonstrated. Even though there were lower basal levels of NF-κB activity in the PC-3 D12 cells compared to the Parental PC-3, docetaxel induced higher NF-κB activity and IκB phosphorylation at 3 and 6 hours with only minor changes in the DU-145 cells. Inhibition of NF-κB with the BAY 11-7082 inhibitor reversed the resistance to Docetaxel.. This study confirms that multiple mechanisms contribute to Docetaxel resistance and the central transcription factor NF-κB plays an immensely important role in determining docetaxel-resistance which may represent an appropriate therapeutic target.

    Topics: Acridines; Antineoplastic Agents; Apoptosis; Apoptosis Regulatory Proteins; ATP Binding Cassette Transporter, Subfamily B, Member 1; beta-Galactosidase; Cell Line, Tumor; Cell Survival; Cellular Senescence; Docetaxel; Drug Resistance, Neoplasm; Gene Expression; Gene Expression Profiling; Genes, Reporter; Humans; Luciferases, Renilla; Male; Molecular Targeted Therapy; NF-kappa B; Nitriles; Prostatic Neoplasms; Sulfones; Taxoids; Tetrahydroisoquinolines

2011
Inhibition of constitutive and cxc-chemokine-induced NF-kappaB activity potentiates ansamycin-based HSP90-inhibitor cytotoxicity in castrate-resistant prostate cancer cells.
    British journal of cancer, 2009, Nov-03, Volume: 101, Issue:9

    We determined how CXC-chemokine signalling and necrosis factor-kappaB (NF-kappaB) activity affected heat-shock protein 90 (Hsp90) inhibitor (geldanamycin (GA) and 17-allylamino-demethoxygeldanamycin (17-AAG)) cytotoxicity in castrate-resistant prostate cancer (CRPC).. Geldanamycin and 17-AAG toxicity, together with the CXCR2 antagonist AZ10397767 or NF-kappaB inhibitor BAY11-7082, was assessed by 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay in two CRPC lines, DU145 and PC3. Flow cytometry quantified apoptotic or necrosis profiles. Necrosis factor-kappaB activity was determined by luciferase readouts or indirectly by quantitative PCR and ELISA-based determination of CXCL8 expression.. Geldanamycin and 17-AAG reduced PC3 and DU145 cell viability, although PC3 cells were less sensitive. Addition of AZ10397767 increased GA (e.g., PC3 IC(20): from 1.67+/-0.4 to 0.18+/-0.2 nM) and 17-AAG (PC3 IC(20): 43.7+/-7.8 to 0.64+/-1.8 nM) potency in PC3 but not DU145 cells. Similarly, BAY11-7082 increased the potency of 17-AAG in PC3 but not in DU145 cells, correlating with the elevated constitutive NF-kappaB activity in PC3 cells. AZ10397767 increased 17-AAG-induced apoptosis and necrosis and decreased NF-kappaB activity/CXCL8 expression in 17-AAG-treated PC3 cells.. Ansamycin cytotoxicity is enhanced by inhibiting NF-kappaB activity and/or CXC-chemokine signalling in CRPC cells. Detecting and/or inhibiting NF-kappaB activity may aid the selection and treatment response of CRPC patients to Hsp90 inhibitors.

    Topics: Apoptosis; Benzoquinones; Cell Line, Tumor; Cell Survival; HSP90 Heat-Shock Proteins; Humans; Interleukin-8; Lactams, Macrocyclic; Male; Necrosis; NF-kappa B; Nitriles; Orchiectomy; Prostatic Neoplasms; Receptors, Interleukin-8B; Rifabutin; Signal Transduction; Sulfones

2009
Inhibition of NF-kappaB by (E)3-[(4-methylphenyl)-sulfonyl]-2-propenenitrile (BAY11-7082; BAY) is associated with enhanced 12-O-tetradecanoylphorbol-13-acetate-induced growth suppression and apoptosis in human prostate cancer PC-3 cells.
    International journal of oncology, 2008, Volume: 32, Issue:1

    The effects of 12-O-tetradecanoylphorbol-13-acetate (TPA) alone or in combination with an NF-kappaB inhibitor, (E)3-[(4-methylphenyl)-sulfonyl]-2-propenenitrile (BAY 11-7082; BAY), on the growth and apoptosis of human prostate cancer PC-3 cells cultured in vitro or grown in immunodeficient mice were studied. Treatment of cultured PC-3 cells with TPA (0.2-10 ng/ml) for 96 h resulted in growth inhibition and apoptosis in a concentration-dependent manner. BAY inhibited NF-kappaB activity in PC-3 cells as determined by a luciferase reporter assay and enhanced TPA-induced growth inhibition and apoptosis in cultured PC-3 cells. In animal studies, NCr immunodeficient mice were injected subcutaneously with PC-3 cells in Matrigel. Mice with well-established tumors received daily i.p. injections with TPA (100 ng/g body weight/day), BAY (4 microg/g/day), or a combination of TPA (100 ng/g/day) and BAY (4 microg/g/day) for 36 days. Tumor growth occurred in all of the vehicle-treated control mice. The percent of animals with some tumor regression after 36 days of treatment was 0% for the control group, 40% for the TPA group, 50% for the BAY group and 100% for the TPA + BAY group. Mechanistic studies indicated that treatment of the mice with TPA or TPA + BAY decreased proliferation and increased apoptosis in the tumors. Results from our studies indicate that inhibition of NF-kappaB activity is associated with enhanced TPA-induced growth inhibition and apoptosis in PC-3 cells. Inhibition of NF-kappaB activity by suitable pharmacological inhibitors may be an effective strategy for improving the therapeutic efficacy of TPA in prostate cancer.

    Topics: Animals; Apoptosis; Cell Proliferation; Humans; Male; Mice; Mice, Nude; NF-kappa B; Nitriles; Prostatic Neoplasms; Sulfones; Tetradecanoylphorbol Acetate

2008
Id-1 expression induces androgen-independent prostate cancer cell growth through activation of epidermal growth factor receptor (EGF-R).
    Carcinogenesis, 2004, Volume: 25, Issue:4

    The failure of prostate cancer treatment is largely due to the development of androgen independence, since the androgen depletion therapy remains the front-line option for this cancer. Previously, we reported that over-expression of the helix-loop-helix protein Id-1 was associated with progression of prostate cancer and ectopic expression of Id-1 induced serum-independent proliferation in prostate cancer cells. In the present study, we investigated if exogenous Id-1 expression in the androgen sensitive LNCaP cells had any effect on androgen-dependent cell growth and studied the molecular mechanisms involved. Using stable Id-1 transfectants, we found that expression of Id-1 was able to reduce androgen-stimulated growth and S phase fraction of the cell cycle in LNCaP cells, indicating that Id-1 may be involved in the development of androgen independence in these cells. The Id-1-induced androgen-independent prostate cancer cell growth was correlated with up-regulation of EGF-R (epidermal growth factor-receptor) and PSA (prostate specific antigen) expression, as confirmed by western blotting analysis and luciferase assays. In contrast, down-regulation of Id-1 in androgen-independent DU145 cells by its antisense oligonucleotides resulted in suppression of EGF-R expression at both transcriptional and protein levels. In addition, the results from immunohistochemistry study showed that Id-1 expression was significantly elevated in hormone refractory prostate cancer tissues when compared with the hormone-dependent tumours. Our results suggest that up-regulation of Id-1 in prostate cancer cells may be one of the mechanisms responsible for developing androgen independence and this process may be regulated through induction of EGF-R expression. Inactivation of Id-1 may provide a potential therapeutic strategy leading to inhibition of androgen-independent prostate cancer cell growth.

    Topics: Adenocarcinoma; Androgens; Animals; Cell Division; Cell Line, Tumor; Cell Nucleus; ErbB Receptors; Humans; Immunohistochemistry; Inhibitor of Differentiation Protein 1; Male; Mice; Mice, Nude; NF-kappa B; Nitriles; Promoter Regions, Genetic; Prostatic Neoplasms; Repressor Proteins; Sulfones; Transcription Factors; Transfection; Transplantation, Heterologous

2004
The molecular mechanism of sensitization to Fas-mediated apoptosis by 2-methoxyestradiol in PC3 prostate cancer cells.
    Molecular carcinogenesis, 2004, Volume: 39, Issue:1

    It is widely known that death receptor Fas-dependent apoptotic signals are associated with development of prostate cancer, but the key pathways involved in sensitivity to the apoptosis remain unclear. Here we investigated the molecular mechanism by which 2-methoxyestradiol (2-ME) effectively sensitizes a human prostate cancer cell line, PC3, to Fas-mediated apoptosis. 2-ME significantly inhibited nuclear factor-kappaB (NF-kappaB) activation and downregulated Fas-associated death domain (FADD) protein interluekin-1beta-converting enzyme inhibitory protein (FLIP). Overexpression of the dominant negative mutant form of IkappaBalpha (d/n IkappaBalpha) or treatment with Ikappa kinase-specific inhibitor Bay117082 gave the same results, although the sensitizing effect was not as pronounced. A selective inhibitor of Akt phosphorylation, LY294002, accelerated formation of the death-inducing signaling complex (DISC) not only by FLIP reduction but also by enhancement of recruitment of the FADD to Fas, thereby sensitizing PC3 cells to apoptosis similar to the case with 2-ME stimulation. Moreover, we found that inhibition of 2-ME-induced extracellular signal-regulated kinase (ERK) activation by the upstream kinase inhibitor PD98059 significantly enhanced 2-ME-mediated suppression of Akt activation, resulting in much greater sensitization to apoptosis. Taken together, the present findings indicate that 2-ME suppresses NF-kappaB/FLIP signaling and enhances DISC formation through inhibition of Akt, and that PC3 cells thereby are being sensitized to Fas-mediated apoptosis and by a process closely associated with ERK.

    Topics: 2-Methoxyestradiol; Apoptosis; Arabidopsis Proteins; Carrier Proteins; CASP8 and FADD-Like Apoptosis Regulating Protein; Chromones; Death Domain Receptor Signaling Adaptor Proteins; Drug Resistance, Neoplasm; Electrophoretic Mobility Shift Assay; Enzyme Activation; Enzyme Inhibitors; Estradiol; fas Receptor; Fatty Acid Desaturases; Flavonoids; Genes, Dominant; Humans; I-kappa B Proteins; Intracellular Signaling Peptides and Proteins; Luciferases; Male; Mitogen-Activated Protein Kinases; Morpholines; NF-kappa B; NF-KappaB Inhibitor alpha; Nitriles; Organic Chemicals; Phosphorylation; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Proto-Oncogene Proteins; Proto-Oncogene Proteins c-akt; Receptors, Tumor Necrosis Factor; Signal Transduction; Sulfones; Tumor Cells, Cultured

2004
Contributions of mitogen-activated protein kinase and nuclear factor kappa B to N-(4-hydroxyphenyl)retinamide-induced apoptosis in prostate cancer cells.
    Molecular carcinogenesis, 2002, Volume: 35, Issue:3

    The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4-HPR) has been shown to induce apoptosis in various types of tumors, including prostate cancer. We sought to examine the key mechanisms affecting the resistance to 4-HPR-induced apoptosis in three human prostate cancer cell lines, PC-3, DU145, and LNCaP. Concentrations of more than 40 microM 4-HPR produced apoptosis to almost the same extent in all cell lines; however, only the LNCaP line remained highly sensitive to concentrations less than 10 microM. These differing sensitivities at low concentrations correlated well with the level of constitutive activation of nuclear factor kappa B (NFkappaB) in the individual cell lines. We found that NFkappaB activation inhibited c-jun NH(2)-terminal kinase and caspase 3 activation induced by 4-HPR and that NFkappaB inhibition by the I kappa B alpha phosphorylation inhibitor compound Bay 117082 resulted in increasing sensitization of both PC-3 and DU145 lines to apoptosis induced by 4-HPR at low concentrations. Furthermore, we found that inhibition of extracellular signal-regulated kinase (ERK) enhanced the suppression of NFkappaB by 4-HPR and also resulted in sensitization to apoptosis in the DU145 cell line, in which ERK is activated constitutively. It thus appears that mitogen-activated protein kinase associated with the activity of NFkappaB plays an important role in the degree of resistance to 4-HPR-induced apoptosis in human prostate cancer cells.

    Topics: Antineoplastic Agents; Apoptosis; beta-Galactosidase; Caspase 3; Caspases; Cell Survival; Drug Resistance, Neoplasm; Electrophoretic Mobility Shift Assay; Fenretinide; Humans; I-kappa B Kinase; JNK Mitogen-Activated Protein Kinases; Male; MAP Kinase Kinase 4; MAP Kinase Signaling System; Mitogen-Activated Protein Kinase 1; Mitogen-Activated Protein Kinase 3; Mitogen-Activated Protein Kinase 6; Mitogen-Activated Protein Kinase Kinases; Mitogen-Activated Protein Kinases; NF-kappa B; Nitriles; Organic Chemicals; Prostatic Neoplasms; Protein Serine-Threonine Kinases; Sulfones; Tumor Cells, Cultured

2002
Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer.
    Oncogene, 2001, Nov-01, Volume: 20, Issue:50

    This work presents direct evidence that the bcl-2 gene is transcriptionally regulated by nuclear factor-kappa B (NF-kappa B) and directly links the TNF-alpha/NF-kappa B signaling pathway with Bcl-2 expression and its pro-survival response in human prostate carcinoma cells. DNase I footprinting, gel retardation and supershift analysis identified a NF-kappa B site in the bcl-2 p2 promoter. In the context of a minimal promoter, this bcl-2 p2 site 1 increased transcription 10-fold in the presence of the p50/p65 expression vectors, comparable to the increment observed with the consensus NF-kappa B site, while for the full p2 promoter region transcriptional activity was increased sixfold by over-expression of NF-kappa B, an effect eliminated by mutating the bcl-2 p2 site 1. The expression of Bcl-2 has been linked to the hormone-resistant phenotype of advanced prostate cancer. Here we show that an increase in the level of expression of Bcl-2 in the human prostate carcinoma cell line LNCaP observed in response to hormone withdrawal is further augmented by TNF-alpha treatment, and this effect is abated by inhibitors of NF-kappa B. Concomitantly, bcl-2 p2 promoter studies in LNCaP cells show a 40-fold increase in promoter activity after stimulation with TNF-alpha in the absence of hormone.

    Topics: Adenocarcinoma; Androgens; Antineoplastic Agents; Binding Sites; Consensus Sequence; Dimerization; DNA Footprinting; DNA-Binding Proteins; Gene Expression Regulation, Neoplastic; Genes, bcl-2; Genetic Vectors; Humans; I-kappa B Proteins; Male; Mutagenesis, Site-Directed; Neoplasms, Hormone-Dependent; NF-kappa B; NF-KappaB Inhibitor alpha; Nitriles; Organic Chemicals; Peptide Fragments; Promoter Regions, Genetic; Prostatic Neoplasms; Protein Subunits; Proto-Oncogene Proteins c-bcl-2; Signal Transduction; Sulfones; Transcriptional Activation; Transfection; Tumor Cells, Cultured; Tumor Necrosis Factor-alpha

2001