bay-11-7082 and benzyloxycarbonylleucyl-leucyl-leucine-aldehyde

bay-11-7082 has been researched along with benzyloxycarbonylleucyl-leucyl-leucine-aldehyde* in 19 studies

Other Studies

19 other study(ies) available for bay-11-7082 and benzyloxycarbonylleucyl-leucyl-leucine-aldehyde

ArticleYear
Constitutive activation of the canonical NF-κB signaling pathway in EBV-associated gastric carcinoma.
    Virology, 2019, Volume: 532

    EBV-associated gastric carcinoma (EBVaGC) is a specific subgroup of gastric carcinoma, and the multifunctional transcriptional factor NF-κB may contribute to its tumorigenesis. In this study, we comprehensively characterized NF-κB signaling in EBVaGC using qRT-PCR, western blot, immunofluorescence assays, ELISA, and immunohistochemistry staining. NF-κB-signaling inhibitors may inhibit the growth of EBVaGC cells and induce significant apoptosis. IκBα is a key regulatory molecule, and repression of IκBα can contribute to aberrant NF-κB activation. Overexpression of LMP1 and LMP2A in the EBV-negative GC cell line SGC7901 could inhibit the expression of IκBα and induce NF-κB activation. These findings indicate that the canonical NF-κB signal is constitutively activated and plays an important role in EBVaGC tumorigenesis.

    Topics: Carcinogenesis; Carcinoma; Cell Line, Tumor; Cell Proliferation; Epstein-Barr Virus Infections; Epstein-Barr Virus Nuclear Antigens; Gene Expression Regulation, Neoplastic; Herpesvirus 4, Human; Humans; Leupeptins; NF-kappa B; NF-KappaB Inhibitor alpha; Nitriles; RNA, Small Interfering; Signal Transduction; Stomach Neoplasms; Sulfones; TNF Receptor-Associated Factor 1; Viral Matrix Proteins

2019
Silymarin induces cyclin D1 proteasomal degradation via its phosphorylation of threonine-286 in human colorectal cancer cells.
    International immunopharmacology, 2015, Volume: 24, Issue:1

    Silymarin from milk thistle (Silybum marianum) plant has been reported to show anti-cancer, anti-inflammatory, antioxidant and hepatoprotective effects. For anti-cancer activity, silymarin is known to regulate cell cycle progression through cyclin D1 downregulation. However, the mechanism of silymarin-mediated cyclin D1 downregulation still remains unanswered. The current study was performed to elucidate the molecular mechanism of cyclin D1 downregulation by silymarin in human colorectal cancer cells. The treatment of silymarin suppressed the cell proliferation in HCT116 and SW480 cells and decreased cellular accumulation of exogenously-induced cyclin D1 protein. However, silymarin did not change the level of cyclin D1 mRNA. Inhibition of proteasomal degradation by MG132 attenuated silymarin-mediated cyclin D1 downregulation and the half-life of cyclin D1 was decreased in the cells treated with silymarin. In addition, silymarin increased phosphorylation of cyclin D1 at threonine-286 and a point mutation of threonine-286 to alanine attenuated silymarin-mediated cyclin D1 downregulation. Inhibition of NF-κB by a selective inhibitor, BAY 11-7082 suppressed cyclin D1 phosphorylation and downregulation by silymarin. From these results, we suggest that silymarin-mediated cyclin D1 downregulation may result from proteasomal degradation through its threonine-286 phosphorylation via NF-κB activation. The current study provides new mechanistic link between silymarin, cyclin D1 downregulation and cell growth in human colorectal cancer cells.

    Topics: Antineoplastic Agents; Cell Cycle; Cell Proliferation; Colorectal Neoplasms; Cyclin D1; HCT116 Cells; Humans; Leupeptins; NF-kappa B; Nitriles; Phosphorylation; Point Mutation; Proteasome Endopeptidase Complex; Proteolysis; Silybum marianum; Silymarin; Sulfones; Threonine

2015
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
Opioid-induced chemokine expression requires NF-κB activity: the role of PKCζ.
    Journal of leukocyte biology, 2011, Volume: 89, Issue:2

    Opioid receptor agonists induce broad immunomodulatory activity, which substantially alters host defense and the inflammatory response. Previous studies have shown that the MOR selective agonist DAMGO has the capacity to increase the expression of the proinflammatory chemokines CCL2, CCL5, and CXCL10 in human PBMCs. NF-κB is a transcription factor that plays a pivotal role in innate and adaptive immune responses. We report that NF-κB is a vital player in the DAMGO-induced, MOR-mediated regulation of chemokine expression. Results show that NF-κB inhibitors prevent the induction of CCL2 expression in response to DAMGO administration and that the NF-κB subunit, p65, is phosphorylated at serine residues 311 and 536 in response to MOR activation. Furthermore, we demonstrate that PKCζ is phosphorylated following DAMGO-induced MOR activation, and this kinase is essential for NF-κB activation as well as CCL2 expression and transcriptional activity. Finally, ChIP analysis shows that DAMGO administration induces binding of p65 to the enhancer region of the CCL2 promoter. These data are consistent with the notion that MOR activation promotes a proinflammatory response, which involves NF-κB activation. Our results also suggest a significant and novel role for PKCζ as an essential participant in the MOR-mediated regulation of proinflammatory chemokine expression.

    Topics: Aldehydes; Analgesics, Opioid; Cells, Cultured; Chemokine CCL2; Enkephalin, Ala(2)-MePhe(4)-Gly(5)-; HEK293 Cells; Humans; Leupeptins; NF-kappa B; Nitriles; Protein Kinase C; Protein Kinase Inhibitors; Receptors, Opioid, mu; Sulfones

2011
NFκB inhibitors induce cell death in glioblastomas.
    Biochemical pharmacology, 2011, Feb-01, Volume: 81, Issue:3

    Identification of novel target pathways in glioblastoma (GBM) remains critical due to poor prognosis, inefficient therapies and recurrence associated with these tumors. In this work, we evaluated the role of nuclear-factor-kappa-B (NFκB) in the growth of GBM cells, and the potential of NFκB inhibitors as antiglioma agents. NFκB pathway was found overstimulated in GBM cell lines and in tumor specimens compared to normal astrocytes and healthy brain tissues, respectively. Treatment of a panel of established GBM cell lines (U138MG, U87, U373 and C6) with pharmacological NFκB inhibitors (BAY117082, parthenolide, MG132, curcumin and arsenic trioxide) and NFκB-p65 siRNA markedly decreased the viability of GBMs as compared to inhibitors of other signaling pathways such as MAPKs (ERK, JNK and p38), PKC, EGFR and PI3K/Akt. In addition, NFκB inhibitors presented a low toxicity to normal astrocytes, indicating selectivity to cancerous cells. In GBMs, mitochondrial dysfunction (membrane depolarization, bcl-xL downregulation and cytochrome c release) and arrest in the G2/M phase were observed at the early steps of NFκB inhibitors treatment. These events preceded sub-G1 detection, apoptotic body formation and caspase-3 activation. Also, NFκB was found overstimulated in cisplatin-resistant C6 cells, and treatment of GBMs with NFκB inhibitors overcame cisplatin resistance besides potentiating the effects of the chemotherapeutics, cisplatin and doxorubicin. These findings support NFκB as a potential target to cell death induction in GBMs, and that the NFκB inhibitors may be considered for in vivo testing on animal models and possibly on GBM therapy.

    Topics: Animals; Antineoplastic Agents; Apoptosis; Arsenic Trioxide; Arsenicals; Astrocytes; Brain Neoplasms; Cell Cycle; Cell Death; Cell Line, Tumor; Cisplatin; Curcumin; Doxorubicin; Drug Synergism; Glioblastoma; Humans; Leupeptins; Molecular Targeted Therapy; NF-kappa B; Nitriles; Oxides; Rats; Sesquiterpenes; Signal Transduction; Sulfones

2011
An IκBα phosphorylation inhibitor induces heme oxygenase-1(HO-1) expression through the activation of reactive oxygen species (ROS)-Nrf2-ARE signaling and ROS-PI3K/Akt signaling in an NF-κB-independent mechanism.
    Cellular signalling, 2011, Volume: 23, Issue:9

    Reactive oxygen species (ROS) are important signaling molecules in cells. Excessive ROS induce expression of inflammatory mediators, such as iNOS and COX2. Antioxidant enzymes, such as, heme oxygenase-1 (HO-1), tightly regulate ROS levels within cells. Here, we show that Bay 11-7082 (Bay) increased HO-1 mRNA and protein expression in human colon cancer HT29 cells. Bay induced translocation of NF-E2-related factor 2 (Nrf2) into nuclei and increased the binding activity of the antioxidant response element (ARE). In addition, PI3K/Akt inhibitor (LY294002) blocked Bay-induced HO-1 expression. Pretreatment with anti-oxidants (N-acetylcysteine (NAC) or glutathione) significantly reduced Bay-induced HO-1 mRNA/protein expression, nuclear translocation of Nrf2 and phosphorylation of Akt. However, PI3K/Akt signaling was independent of Bay-induced Nrf2 translocation and ARE binding activity. Furthermore, other NF-κB inhibitors, such as pyrrolidine dithiocarbamate (PDTC) and MG132, also increased HO-1 mRNA and protein expression. However, although overexpression of dominant negative inhibitory κB (IκB) reduced NF-κB-driven transcriptional activity, IκB overexpression did not increase HO-1 expression. Taken together, our results suggest that in human colon cancer HT29 cells, Bay induces HO-1 expression by increasing ROS production in an Nrf2-ARE and PI3K dependent manner, but Bay acts independently of NF-κB.

    Topics: Analysis of Variance; Electrophoretic Mobility Shift Assay; Gene Expression Regulation; Glutathione; Heme Oxygenase-1; HT29 Cells; Humans; Immunohistochemistry; Leupeptins; NF-E2-Related Factor 2; NF-kappa B; Nitriles; Phosphatidylinositol 3-Kinases; Phosphorylation; Proline; Proto-Oncogene Proteins c-akt; Reactive Oxygen Species; Response Elements; RNA, Messenger; Signal Transduction; Sulfones; Thiocarbamates; Transfection

2011
Proteasome inhibitors induce apoptosis and reduce viral replication in primary effusion lymphoma cells.
    Biochemical and biophysical research communications, 2011, Dec-02, Volume: 415, Issue:4

    Primary effusion lymphoma (PEL) is an aggressive neoplasm caused by Kaposi's sarcoma-associated herpesvirus (KSHV). This study provides evidence that proteasomal activity is required for both survival of PEL cells stably harboring the KSHV genome and viral replication of KSHV. We evaluated the cytotoxic effects of proteasome inhibitors on PEL cells. The proteasome inhibitors MG132, lactacystin, and proteasome inhibitor I dramatically inhibited cell proliferation and induced apoptosis of PEL cells through the accumulation of p21 and p27. Furthermore, proteasome inhibitors induced the stabilization of NF-κB inhibitory molecule (IκBα) and suppressed the transcriptional activity of NF-κB in PEL cells. The NF-κB specific inhibitor BAY11-7082 also induced apoptosis in PEL cells. The constitutive activation of NF-κB signaling is essential for the survival and growth of B cell lymphoma cells, including PEL cells. NF-κB signaling is upregulated by proteasome-dependent degradation of IκBα. The suppression of NF-κB signaling by proteasome inhibitors may contribute to the induction of apoptosis in PEL cells. In addition, proteasome activity is required for KSHV replication in KSHV latently infected PEL cells. MG132 reduced the production of progeny virus from PEL cells at low concentrations, which do not affect PEL cell growth. These findings suggest that proteasome inhibitors may represent a novel strategy for the treatment of KSHV infection and KSHV-associated lymphomas.

    Topics: Acetylcysteine; Antineoplastic Agents; Apoptosis; Cell Line, Tumor; Cysteine Proteinase Inhibitors; Herpesvirus 8, Human; Humans; Leupeptins; Lymphoma, Primary Effusion; NF-kappa B; Nitriles; Oligopeptides; Proteasome Inhibitors; Sulfones; Virus Replication

2011
The pharmacological NFkappaB inhibitors BAY117082 and MG132 induce cell arrest and apoptosis in leukemia cells through ROS-mitochondria pathway activation.
    Cancer letters, 2010, Feb-28, Volume: 288, Issue:2

    A growing body of evidence suggests the inhibition of NFkappaB as a strategy to induce cell death in tumor cells. In this work, we evaluated the effects of the pharmacological NFkappaB inhibitors BAY117082 and MG132 on leukemia cells apoptosis. BAY117082 and MG132 presented potent apoptotic effects compared to inhibitors of MAPKs, EGFR, PI3K/Akt, PKC and PKA signaling pathways. Non-tumor peripheral blood cells were insensitive to BAY117082 and MG132 apoptotic effects. BAY117082 and MG132-induced apoptosis was dependent on their ability to increase ROS as a prelude to mitochondria membrane potential (MMP) depolarization, permeability transition pore opening and cytochrome c release. Antioxidants blocked MG132 and BAY117082 effects on ROS, MMP and cell death. Although apoptotic markers as phosphatidylserine externalization, chromatin condensation and sub-G1 were detected in BAY117082-treated cells, caspases activation did not occur and apoptosis was insensitive to caspase inhibitors, suggesting a caspase-independent mechanism. In contrast, MG132 induced classical apoptosis through ROS-mitochondria and subsequent caspase-9/caspase-3 activation. At sub-apoptotic concentrations, BAY117082 and MG132 arrested cells in G2/M phase of the cell cycle and blocked doxorubicin-induced NFkappaB, which sensitized doxorubicin-resistant cells. Data suggest that the NFkappaB inhibitors MG132 and BAY117082 are potential anti-leukemia agents.

    Topics: Antibiotics, Antineoplastic; Antineoplastic Agents; Apoptosis; Caspases; Cell Proliferation; Cytochromes c; Dose-Response Relationship, Drug; Doxorubicin; Drug Resistance, Neoplasm; Enzyme Activation; Humans; Jurkat Cells; K562 Cells; Leukemia; Leupeptins; Membrane Potential, Mitochondrial; Mitochondria; Mitochondrial Membrane Transport Proteins; Mitochondrial Permeability Transition Pore; NF-kappa B; Nitriles; Reactive Oxygen Species; Sulfones; Time Factors; U937 Cells

2010
Akt-dependent NF-kappaB activation is required for bile acids to rescue colon cancer cells from stress-induced apoptosis.
    Experimental cell research, 2009, Feb-01, Volume: 315, Issue:3

    Conjugated secondary bile acids promote human colon cancer cell proliferation by activating EGF receptors (EGFR). We hypothesized that bile acid-induced EGFR activation also mediates cell survival by downstream Akt-regulated activation of NF-kappaB. Deoxycholyltaurine (DCT) treatment attenuated TNF-alpha-induced colon cancer cell apoptosis, and stimulated rapid and sustained NF-kappaB nuclear translocation and transcriptional activity (detected by NF-kappaB binding to an oligonucleotide consensus sequence and by activation of luciferase reporter gene constructs). Both DCT-induced NF-kappaB nuclear translocation and attenuation of TNF-alpha-stimulated apoptosis were dependent on EGFR activation. Inhibitors of nuclear translocation, proteosome activity, and IkappaBalpha kinase attenuated NF-kappaB transcriptional activity. Cell transfection with adenoviral vectors encoding a non-degradable IkappaBalpha 'super-repressor' blocked the actions of DCT on both NF-kappaB activation and TNF-alpha-induced apoptosis. Likewise, transfection with mutant akt and treatment with a chemical inhibitor of Akt attenuated effects of DCT on NF-kappaB transcriptional activity and TNF-alpha-induced apoptosis. Chemical inhibitors of Akt and NF-kappaB activation also attenuated DCT-induced rescue of H508 cells from ultraviolet radiation-induced apoptosis. Collectively, these observations indicate that, downstream of EGFR, bile acid-induced colon cancer cell survival is mediated by Akt-dependent NF-kappaB activation. These findings provide a mechanism whereby bile acids increase resistance of colon cancer to chemotherapy and radiation.

    Topics: Active Transport, Cell Nucleus; Apoptosis; Bile Acids and Salts; Cell Line, Tumor; Cell Nucleus; Cell Survival; Chromones; Colonic Neoplasms; ErbB Receptors; Humans; I-kappa B Kinase; Leupeptins; Morpholines; Mutation; NF-kappa B; Nitriles; Peptides; Proto-Oncogene Proteins c-akt; Sulfones; Taurodeoxycholic Acid; Tumor Necrosis Factor-alpha; Ultraviolet Rays

2009
Discovery of novel small molecule cell type-specific enhancers of NF-kappaB nuclear translocation.
    Bioorganic & medicinal chemistry letters, 2009, Feb-15, Volume: 19, Issue:4

    An IKKbeta inhibitor reported to block NF-kappaB transcriptional activities in Jurkat T cells, was found to enhance NF-kappaB translocation in HUVEC cells. These studies suggested a noncanonical NF-kappaB signaling pathway independent of IKKbeta in HUVEC cells.

    Topics: Combinatorial Chemistry Techniques; Humans; Jurkat Cells; Leupeptins; Molecular Structure; NF-kappa B; Nitriles; Quinazolines; Sulfones; Sulfoxides; Tetrazoles

2009
Novel cytostatic activity of the trypanocidal drug Benznidazole.
    International immunopharmacology, 2009, Volume: 9, Issue:6

    We have shown that Benznidazole (BZL), a compound with well documented trypanocidal activity, possesses anti-inflammatory properties and inhibits the nuclear factor kappaB (NF-kappaB). Given the relationship between this transcription factor and cell growth, in this study we address the role of NF-kappaB blockade by BZL in the proliferation of different cell lines. Our studies demonstrate that this compound significantly reduced proliferation of RAW 264.7 macrophage cell line, as assessed by trypan blue exclusion, MTT reduction and [(3)H]-thymidine incorporation, at a concentration shown to inhibit NF-kappaB. Treatment with BZL also led to growth arrest in CHO, MDCK and HeLa cells. Interestingly, growth inhibition was found to be a reversible process, not accompanied by significant cell death, indicating that the drug behaves mainly as a cytostatic compound. As this effect might be related to NF-kappaB inhibition, we next evaluated whether other NF-kappaB inhibitors could induce growth arrest in RAW 264.7 and HeLa cells. We found that IKK inhibition led to growth arrest in both cell lines, indicating that NF-kappaB inhibition may be the potential mechanism by which BZL inhibits cell proliferation. To the best of our knowledge, this is the first report of an anti-proliferative activity of the trypanocidal drug against different cell lines and provides a mechanistic insight that may help understand some of the adverse effects associated with prolonged treatment.

    Topics: Animals; Cell Line; Cell Line, Tumor; Cell Proliferation; CHO Cells; Cricetinae; Cricetulus; Cytostatic Agents; Dogs; HeLa Cells; Humans; Leupeptins; Mice; NF-kappa B; Nitric Oxide; Nitriles; Nitroimidazoles; Sulfones; Trypanocidal Agents

2009
The HMG-CoA reductase inhibitor rosuvastatin inhibits plasminogen activator inhibitor-1 expression and secretion in human adipocytes.
    Atherosclerosis, 2008, Volume: 196, Issue:2

    Human preadipocytes and adipocytes are known to produce the proatherogenic factor PAI-1 and proinflammatory cytokines, and obesity was found to be state of increased adipose production of these factors. In the present study, we investigated the effect of rosuvastatin on the regulation of PAI-1 gene expression in human adipocytes. Human preadipocytes, adipocytes in primary culture and the SGBS cell line were used as cell models. Cells were transfected using various constructs and promoter activity was measured as luciferase activity. PAI-1 expression was measured by quantitative RT-PCR and ELISA. Rosuvastatin inhibited PAI-1 mRNA expression and secretion of the protein in a concentration-dependent manner. This effect was reversed by isoprenoids. Addition of MEK-inhibitors and NFkappaB inhibitors also reduced PAI-1 expression and PAI-1 promoter luciferase activity. Further experiments revealed that rosuvastatin down-regulated the MEKK-1 mediated activation of the PAI-1 promoter. In conclusion our data suggest that rosuvastatin inhibits PAI-1 expression and release from human adipocytes via a MEKK-1-dependent but not a NFkappaB-dependent mechanism.

    Topics: 3T3-L1 Cells; Adipocytes; Adult; Animals; Cells, Cultured; Down-Regulation; Female; Fluorobenzenes; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Leupeptins; Macrolides; Male; MAP Kinase Kinase Kinase 1; Mice; Middle Aged; NF-kappa B; Nitriles; Plasminogen Activator Inhibitor 1; Promoter Regions, Genetic; Pyrimidines; Rosuvastatin Calcium; Signal Transduction; Sulfonamides; Sulfones; Terpenes; Transfection

2008
Involvement of mitogen-activated protein kinases and NFkappaB in LPS-induced CD40 expression on human monocytic cells.
    Toxicology and applied pharmacology, 2008, Apr-15, Volume: 228, Issue:2

    CD40 is a costimulatory molecule linking innate and adaptive immune responses to bacterial stimuli, as well as a critical regulator of functions of other costimulatory molecules. The mechanisms regulating lipopolysaccharide (LPS)-induced CD40 expression have not been adequately characterized in human monocytic cells. In this study we used a human monocytic cell line, THP-1, to investigate the possible mechanisms of CD40 expression following LPS exposure. Exposure to LPS resulted in a dose- and time-dependent increase in CD40 expression. Further studies using immunoblotting and pharmacological inhibitors revealed that mitogen-activated protein kinases (MAPKs) and NFkappaB were activated by LPS exposure and involved in LPS-induced CD40 expression. Activation of MAPKs was not responsible for LPS-induced NFkappaB activation. TLR4 was expressed on THP-1 cells and pretreatment of cells with a Toll-like receptor 4 (TLR4) neutralizing antibody (HTA125) significantly blunted LPS-induced MAPK and NFkappaB activation and ensuing CD40 expression. Additional studies with murine macrophages expressing wild type and mutated TLR4 showed that TLR4 was implicated in LPS-induced ERK and NFkappaB activation, and CD40 expression. Moreover, blockage of MAPK and NFkappaB activation inhibited LPS-induced TLR4 expression. In summary, LPS-induced CD40 expression in monocytic cells involves MAPKs and NFkappaB.

    Topics: Animals; Anthracenes; Butadienes; CD40 Antigens; Cell Line; Cell Line, Tumor; Dose-Response Relationship, Drug; Electrophoretic Mobility Shift Assay; Flow Cytometry; Humans; Imidazoles; Immunoblotting; Leupeptins; Mitogen-Activated Protein Kinases; Monocytes; NF-kappa B; Nitriles; Phosphorylation; Polysaccharides, Bacterial; Protein Kinase Inhibitors; Pyridines; Signal Transduction; Sulfones; Time Factors; Toll-Like Receptor 4

2008
Decreased epithelial barrier function evoked by exposure to metabolic stress and nonpathogenic E. coli is enhanced by TNF-alpha.
    American journal of physiology. Gastrointestinal and liver physiology, 2008, Volume: 294, Issue:3

    A defect in mitochondrial activity contributes to many diseases. We have shown that monolayers of the human colonic T84 epithelial cell line exposed to dinitrophenol (DNP, uncouples oxidative phosphorylation) and nonpathogenic Escherichia coli (E. coli) (strain HB101) display decreased barrier function. Here the impact of DNP on macrophage activity and the effect of TNF-alpha, DNP, and E. coli on epithelial permeability were assessed. DNP treatment of the human THP-1 macrophage cell line resulted in reduced ATP synthesis, and, although hyporesponsive to LPS, the metabolically stressed macrophages produced IL-1beta, IL-6, and TNF-alpha. Given the role of TNF-alpha in inflammatory bowel disease (IBD) and the association between increased permeability and IBD, recombinant TNF-alpha (10 ng/ml) was added to the DNP (0.1 mM) + E. coli (10(6) colony-forming units), and this resulted in a significantly greater loss of T84 epithelial barrier function than that elicited by DNP + E. coli. This increased epithelial permeability was not due to epithelial death, and the enhanced E. coli translocation was reduced by pharmacological inhibitors of NF-kappabeta signaling (pyrrolidine dithiocarbamate, NF-kappabeta essential modifier-binding peptide, BAY 11-7082, and the proteosome inhibitor, MG132). In contrast, the drop in transepithelial electrical resistance was unaffected by the inhibitors of NF-kappabeta. Thus, as an integrative model system, our findings support the induction of a positive feedback loop that can severely impair epithelial barrier function and, as such, could contribute to existing inflammation or trigger relapses in IBD. Thus metabolically stressed epithelia display increased permeability in the presence of viable nonpathogenic E. coli that is exaggerated by TNF-alpha released by activated immune cells, such as macrophages, that retain this ability even if they themselves are experiencing a degree of metabolic stress.

    Topics: Adenosine Triphosphate; Blotting, Western; Caspases; Cell Death; Cell Line; Cells, Cultured; Dinitrophenols; Electric Impedance; Epithelium; Escherichia coli Infections; Feedback, Physiological; Humans; Interleukin-1beta; Interleukin-6; Leupeptins; Lipopolysaccharides; Macrophages; NF-kappa B; Nitriles; Permeability; Stress, Physiological; Sulfones; Tetrazolium Salts; Thiazoles; Tumor Necrosis Factor-alpha; Uncoupling Agents

2008
Differential involvement of NF-kappaB and MAP kinase pathways in the generation of inflammatory cytokines by human neutrophils.
    Journal of leukocyte biology, 2007, Volume: 81, Issue:2

    The ability of human neutrophils to express a variety of genes encoding inflammatory mediators is well documented, and mounting evidence suggests that neutrophil-derived cytokines and chemokines contribute to the recruitment of discrete leukocyte populations at inflammatory sites. Despite this, our understanding of the signaling intermediates governing the generation of inflammatory cytokines by neutrophils remains fragmentary. Here, we report that inhibitors of the p38 MAPK and MEK pathways substantially diminish the release of (and in the case of p38 inhibitors, the gene expression of) several inflammatory cytokines in neutrophils stimulated with LPS or TNF. In addition, various NF-kappaB inhibitors were found to profoundly impede the inducible gene expression and release of inflammatory cytokines in these cells. The MAPK inhibitors did not affect NF-kappaB activation; instead, the transcriptional effects of the p38 MAPK inhibitor appear to involve transcriptional factor IID. Conversely, the NF-kappaB inhibitors failed to affect the activation of MAPKs. Finally, the MAPK inhibitors were found to prevent the activation a key component of the translational machinery, S6 ribosomal protein, in keeping with their post-transcriptional impact on cytokine generation. To our knowledge, this constitutes the first demonstration that in neutrophils, the inducible expression of proinflammatory cytokines by physiological stimuli largely reflects the ability of the latter to activate NF-kappaB and selected MAPK pathways. Our data also raise the possibility that NF-kappaB or MAPK inhibitors could be useful in the treatment of inflammatory disorders in which neutrophils predominate.

    Topics: Boronic Acids; Cell Differentiation; Cytokines; Humans; Inflammation; Leupeptins; MAP Kinase Signaling System; Mitogen-Activated Protein Kinases; Neutrophils; NF-kappa B; Nitriles; Proline; Protein Kinase Inhibitors; Structure-Activity Relationship; Sulfones; Thiocarbamates

2007
Differential effects of NF-kappaB on apoptosis induced by DNA-damaging agents: the type of DNA damage determines the final outcome.
    Oncogene, 2006, Oct-12, Volume: 25, Issue:47

    The transcription factor nuclear factor kappa-B (NF-kappaB) is generally regarded as an antiapoptotic factor. Accordingly, NF-kappaB activation inhibits death ligand-induced apoptosis. In contrast, ultraviolet light B (UVB)-induced apoptosis is not inhibited but even enhanced upon NF-kappaB activation by interleukin-1 (IL-1). This study was performed to identify the molecular mechanisms underlying this switch of NF-kappaB. Enhancement of UVB-induced apoptosis was always associated with increased release of tumour necrosis factor-alpha (TNF-alpha), which was dependent on NF-kappaB activation. The same was observed when UVA and cisplatin were used, which like UVB induce base modifications. In contrast, apoptosis caused by DNA strand breaks was not enhanced by IL-1, indicating that the type of DNA damage is critical for switching the effect of NF-kappaB on apoptosis. Surprisingly, activated NF-kappaB induced TNF-alpha mRNA expression in the presence of all DNA damage-inducing agents. However, in the presence of DNA strand breaks, there was no release of the TNF-alpha protein, which is so crucial for enhancing apoptosis. Together, this indicates that induction of DNA damage may have a significant impact on biological effects but it is the type of DNA damage that determines the final outcome. This may have implications for the role of NF-kappaB in carcinogenesis and for the application of NF-kappaB inhibitors in anticancer therapy.

    Topics: Acetylcysteine; Antineoplastic Agents; Apoptosis; Chromosome Breakage; Cisplatin; Comet Assay; DNA Damage; Doxorubicin; Etoposide; Gamma Rays; Gene Expression Regulation; Humans; I-kappa B Kinase; I-kappa B Proteins; Interleukin-1; Interleukin-6; KB Cells; Leupeptins; NF-kappa B; NF-KappaB Inhibitor alpha; Nitriles; Proteasome Inhibitors; Receptors, Tumor Necrosis Factor, Type I; RNA, Messenger; Sulfones; Transcription, Genetic; Tumor Necrosis Factor-alpha; Ultraviolet Rays

2006
Effects of mechanistically distinct NF-kappaB inhibitors on glial inducible nitric-oxide synthase expression.
    Nitric oxide : biology and chemistry, 2005, Volume: 12, Issue:4

    Nuclear factor (NF)-kappaB is an important regulator of inflammatory gene expression. Transcriptional regulation of Nos2, the inducible nitric-oxide synthase (iNOS) gene, is complex and not fully understood, but appears to be regulated in part by NF-kappaB. To further understand the role of NF-kappaB in Nos2 expression, we compared three functionally distinct NF-kappaB inhibitors on NF-kappaB transactivation and iNOS induction by rat C6 glial cells. Cytokine-induced activation of a consensus NF-kappaB-reporter gene was concentration-dependently inhibited by BAY 11-7082, MG-132, and helenalin. The rank order of potency was MG-132>helenalin>BAY 11-7082, with low concentrations of helenalin stimulating reporter gene activity. Cytokine-stimulated iNOS expression, measured by nitrite accumulation and in vitro l-citrulline production, was similarly reduced by exposing C6 cells to the NF-kappaB inhibitors. Surprisingly, activation of Nos2-reporter gene constructs containing the proximal 188 bp (containing one kappaB site) or proximal 94 bp (no kappaB site) of the rat promoter also was inhibited with the same rank order of potency. Interestingly, low concentrations of helenalin increased activity of both promoter constructs, while BAY 11-7082 poorly inhibited the 94-bp activity. This is the first report describing BAY 11-7082 and helenalin effects on iNOS expression in astroglia. Given the reported mechanism of actions for these inhibitors, cytokine-induced glial iNOS expression appears more sensitive to disruption of proteasome degradation and p65 function than modulation of IkappaB phosphorylation. These findings may foster the design of therapeutic agents aimed at NF-kappaB-associated pathways involved in neuroinflammation, especially iNOS expression.

    Topics: Animals; Cell Line; Cytokines; Leupeptins; Neuroglia; NF-kappa B; Nitric Oxide Synthase; Nitric Oxide Synthase Type II; Nitriles; Nitrites; Promoter Regions, Genetic; Rats; Sulfones; Transcriptional Activation

2005
Further pharmacological evidence of nuclear factor-kappa B pathway involvement in bradykinin B1 receptor-sensitized responses in human umbilical vein.
    The Journal of pharmacology and experimental therapeutics, 2002, Volume: 301, Issue:3

    Bradykinin (BK) B(1) receptors are thought to exert a pivotal role in maintaining and modulating inflammatory processes. They are not normally present under physiological situations but are induced under physiopathological conditions. In isolated human umbilical vein (HUV), a spontaneous BK B(1) receptor up-regulation and sensitization process has been demonstrated. Based on pyrrolidine-dithiocarbamate inhibition, it has been proposed that this phenomenon is dependent on nuclear factor-kappaB (NF-kappaB) activation. The aim of this study was to further evaluate the NF-kappaB pathway involvement on BK B(1) receptor sensitization in isolated HUV, using several pharmacological tools. In 5-h incubated rings, either the I-kappaB kinase inhibitor 3-(4-methylphenylsulfonyl)-2-propenenitrile (Bay 11-7082) or the proteasome activity inhibitor Z-Leu-Leu-Leu-CHO (MG-132) inhibited the development of the BK B(1) receptor-sensitized contractile responses. Furthermore, pro-inflammatory cytokine interleukin-6 (IL-6) produced a leftward shift of the concentration-response curve to the BK B(1) receptor agonist, whereas anti-inflammatory cytokines interleukin-4 (IL-4) and tumor growth factor-beta1 (TGF-beta1) produced a rightward shift of the responses to des-Arg(9)-BK in our preparations. Taken together, these results point to NF-kappaB as a key intermediary in the activation of the expression of BK B(1) receptor-sensitized responses in HUV and support the role of inflammatory mediators in the modulation of this process.

    Topics: Antineoplastic Agents; Bradykinin; Cysteine Proteinase Inhibitors; Cytokines; Drug Evaluation, Preclinical; Enzyme Inhibitors; Free Radical Scavengers; Humans; Infant, Newborn; Interleukin-4; Interleukin-6; Leupeptins; NF-kappa B; Nitriles; Organic Chemicals; Receptor, Bradykinin B1; Receptors, Bradykinin; Recombinant Proteins; Serotonin; Signal Transduction; Sulfones; Transforming Growth Factor beta; Transforming Growth Factor beta1; Umbilical Veins

2002
Expression of kinin B(1) receptor in fresh or cultured rabbit aortic smooth muscle: role of NF-kappa B.
    American journal of physiology. Heart and circulatory physiology, 2002, Volume: 283, Issue:1

    Kinin B(1) receptor (B(1)R) expression and the importance of the transcription factor nuclear factor (NF)-kappa B in this process were evaluated in models based on the rabbit aorta: freshly isolated tissue (postisolation induction) and cultured smooth muscle cells (SMCs). A 3-h incubation of freshly isolated tissues determined a sharp B(1)R mRNA increase (RT-PCR). Coincubation of tissues with a stimulus (interleukin-1 beta, fetal bovine serum, epidermal growth factor, or cycloheximide) further increased mRNA levels. Cultured SMCs possessed a basal population of surface B(1)Rs ([(3)H]Lys-des-Arg(9)-bradykinin binding) that was upregulated by treatments with the same set of stimuli (binding, mRNA, nuclear runon). Pharmacological inhibitors of NF-kappa B (MG-132, BAY 11-7082, dexamethasone) or actinomycin D reduced the postisolation induction of B(1)Rs in fresh aortic tissue (contractility or mRNA) and the cytokine effect on cells (mRNA, binding). NF-kappa B may be a common mediator of various stimuli that increase B(1)R gene transcription in the rabbit aorta, including tissue isolation, but cycloheximide also stabilizes B(1)R mRNA. The SMC models faithfully mimic the in vivo situation with regard to B(1)R regulation.

    Topics: Animals; Antineoplastic Agents; Aorta; Blood Proteins; Cells, Cultured; Dexamethasone; Dose-Response Relationship, Drug; Epidermal Growth Factor; Interleukin-1; Leupeptins; Male; Muscle, Smooth, Vascular; NF-kappa B; Nitriles; Nucleic Acid Synthesis Inhibitors; Organic Chemicals; Protein Synthesis Inhibitors; Rabbits; Receptor, Bradykinin B1; Receptors, Bradykinin; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Sulfones; Up-Regulation; Vasoconstrictor Agents; Vasodilator Agents

2002