Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Carcinogenesis ; 35(7): 1469-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24413684

ABSTRACT

PDCD4 is a tumor suppressor induced by apoptotic stimuli that regulates both translation and transcription. Previously, we showed that overexpression of PDCD4 leads to decreased anchorage-independent growth in glioblastoma (GBM)-derived cell lines and decreased tumor growth in a GBM xenograft model. In inflammatory cells, PDCD4 stimulates tumor necrosis factor-induced activation of the transcription factor NF-κB, an oncogenic driver in many cancer sites. However, the effect of PDCD4 on NF-κB transcriptional activity in most cancers including GBM is still unknown. We studied the effect of PDCD4 on NF-κB-dependent transcriptional activity in GBM by stably overexpressing PDCD4 in U251 and LN229 cells. Stable PDCD4 expression inhibits NF-κB transcriptional activation measured by a luciferase reporter. The molecular mechanism by which PDCD4 inhibits NF-κB transcriptional activation does not involve inhibited expression of NF-κB p65 or p50 proteins. PDCD4 does not inhibit pathways upstream of NF-κB including the activation of IKKα and IKKß kinases or degradation of IκBα, events needed for nuclear transport of p65 and p50. PDCD4 overexpression does inhibit localization of p65 but not p50 in the nucleus. PDCD4 protein interacts preferentially with p65 protein as shown by co-immunoprecipitation and confocal imaging. PDCD4 overexpression inhibits the mRNA expression of two NF-κB target genes in a p65-dependent manner. These results suggest that PDCD4 can significantly inhibit NF-κB activity in GBM cells by a mechanism that involves direct or indirect protein-protein interaction independent of the expected mRNA-selective translational inhibition. These findings offer novel opportunities for NF-κB-targeted interventions to prevent or treat cancer.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , NF-kappa B p50 Subunit/metabolism , RNA-Binding Proteins/genetics , Transcription Factor RelA/metabolism , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Immunoprecipitation , NF-kappa B p50 Subunit/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcription, Genetic , Tumor Cells, Cultured
2.
Genes Cancer ; 3(1): 37-50, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22893789

ABSTRACT

The transcription factor AP-1 (activator protein-1) regulates a number of genes that drive tumor promotion and progression. While basal levels of AP-1 activity are important for normal cell proliferation and cell survival, overactivated AP-1-dependent gene expression stimulates inflammation, angiogenesis, invasion, and other events that propel carcinogenesis. We seek to discover genes targeted by carcinogenesis inhibitors that do not also inhibit cell proliferation or survival. Transgenic TAM67 (dominant-negative c-Jun) inhibits mouse skin tumorigenesis and tumor progression without inhibiting cell proliferation or induced hyperproliferation. Expression profiling of wild-type and K14-TAM67 mouse epidermis has revealed a number of functionally significant genes that are induced by tumor promoters in wild-type mice but not in those expressing the AP-1 blocker. The current study now identifies Wnt5a signaling as a new target of TAM67 when it inhibits DMBA/TPA-induced carcinogenesis. Wnt5a is required to maintain the tumor phenotype in tumorigenic mouse JB6 cells and Ras-transformed human squamous carcinoma HaCaT-II4 cells, as Wnt5a knockdown suppresses anchorage-independent and tumor xenograft growth. The oncogenic Wnt5a-mediated pathway signals through activation of the protein kinase PKCα and oncogenic transcription factor STAT3 phosphorylation and not through the canonical Wnt/ß-catenin pathway. Similar to Wnt5a knockdown, inhibitors of PKCα blocked STAT3 activation in both mouse JB6 and human HaCaT-II4 tumor cells. Moreover, expression of STAT3-regulated genes FAS, MMP3, IRF1, and cyclin D1 was suppressed with Wnt5a knockdown. Treatment of mouse Wnt5a knockdown cells with a PKCα-specific activator rescued phosphorylation of STAT3. Thus, Wnt5a signaling is required for maintaining the tumor phenotype in squamous carcinoma cells, Wnt5a targeting by the AP-1 blockade contributes to inhibition of skin carcinogenesis, and the signaling pathway traverses PKCα and STAT3 activation. Coordinate overactivation of Wnt5a expression and STAT3 signaling is observed in human skin and colon cancers as well as glioblastoma.

3.
Carcinogenesis ; 32(10): 1427-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21771721

ABSTRACT

The tumor suppressor programmed cell death 4 (Pdcd4) is lost in various tumor tissues. Loss of Pdcd4 has been associated with increased tumorigenic potential and tumor progression. While various mechanisms of Pdcd4 regulation have been described, the effect of an inflammatory tumor microenvironment on Pdcd4 protein expression has not been characterized so far. In the present study, we aimed to elucidate the molecular mechanisms of Pdcd4 protein regulation in tumor cells under inflammatory conditions. 12-O-tetradecanoylphorbol 13-acetate-induced differentiation of human U937 monocytes increased the expression and secretion of inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)-6 and IL-8. Exposure to conditioned medium (CM) of these activated macrophages markedly decreased Pdcd4 protein expression in various tumor cells. Similarly, indirect coculture with such activated U937 monocyte-derived macrophages resulted in the loss of Pdcd4 protein in tumor cells. Decreased Pdcd4 protein levels were attributable to enhanced proteasomal degradation, diminishing Pdcd4 protein half-life. Proteasomal degradation required activation of phosphatidylinositol-3-kinase (PI3K)-mammalian target of rapamycin (mTOR) signaling. Since macrophage-CM sufficed to induce Pdcd4 degradation, Pdcd4 downregulation was determined to be an indirect unidirectional effect of the macrophages on the tumor cells. Pdcd4 protein expression was also attenuated in vivo in mouse colon tissue in response to dextran sodium sulfate-induced colitis. In summary, we characterized PI3K-mTOR-dependent proteasome-mediated Pdcd4 degradation in tumor cells in the inflammatory tumor microenvironment. Consequently, stabilization of Pdcd4 protein could provide a promising novel avenue for therapeutics targeting inflammation-associated tumors.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Genes, Tumor Suppressor , Inflammation/etiology , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Breast Neoplasms/genetics , Carcinogens/pharmacology , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Female , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Interleukin-8/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , U937 Cells
4.
J Biomol Screen ; 15(1): 21-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19901084

ABSTRACT

The novel tumor suppressor Pdcd4 affects tumorigenesis by inhibiting translation. Pdcd4 is phosphorylated and subsequently lost by proteasomal degradation in response to tumor-promoting conditions. Here, the authors describe the development of a reporter cell system to monitor the stability of Pdcd4. The phosphorylation-dependent degradation domain ("target") or an adjacent ("off-target") region of Pdcd4 was cloned into a luciferase expression system. The target constructs were responsive to Pdcd4 degrading conditions (e.g., TPA, p70(S6K1) overactivation), whereas the off-target constructs remained stable. The system was optimized for and shown to be reliable in a high-throughput compatible 384-well format. Screening of 15,275 pure compounds resulted in a hit rate of 0.30% (>50% inhibition of TPA-induced loss of signal, confirmed by reassay). Among the hits were inhibitors of previously identified critical signaling events for TPA-induced Pdcd4 degradation. One compound was identified to be nonspecific using the off-target control cell line. Screening of 135,678 natural product extracts yielded 42 confirmed, specific hits. Z' averaged 0.58 across 446 plates. Further characterization of active natural products and synthetic compounds is expected to identify novel Pdcd4 stabilizers that may be useful in targeting translation to prevent or treat cancers.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Genes, Reporter , High-Throughput Screening Assays/methods , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/chemistry , Biological Products/pharmacology , Cell Line , Humans , Protein Stability/drug effects , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Reproducibility of Results , Small Molecule Libraries/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/chemistry
5.
Mol Cancer Ther ; 8(3): 571-81, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19258426

ABSTRACT

NSC 676914 has been identified as a selective nuclear factor-kappaB (NF-kappaB) inhibitor that does not inhibit cell proliferation. This compound was originally identified in a high-throughput cell-based assay for activator protein-1 (AP-1) inhibitors using synthetic compound libraries and the National Cancer Institute natural product repository. NSC 676914 shows activity against NF-kappaB in luciferase reporter assays at concentrations much less than the IC50 for AP-1. A serum response element reporter used as a specificity control and indicator of cell proliferation was relatively insensitive to the compound. Pretreatment with NSC 676914 is here shown to repress 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced IkappaB-alpha phosphorylation and translocation of p65/50 to the nucleus but not the processing of p52 from p100, suggesting the inhibition of NF-kappaB regulator IKKbeta rather than IKKalpha. Inhibition of NF-kappaB activation occurred as a consequence of blocking phosphorylation of IKK. Induction of IkappaB-alpha phosphorylation by TPA was diminished by pretreatment of NSC 676914 even at 1.1 mumol/L. In contrast, kinases c-Jun-NH2-kinase and extracellular signal-regulated kinases 1 and 2, important for AP-1 activation, showed no significant repression by this compound. Furthermore, a Matrigel invasion assay with breast cancer cell lines and a transformation assay in mouse JB6 cells revealed that TPA-induced invasion and transformation responses were completely repressed by this compound. These results suggest that NSC 676914 could be a novel inhibitor having potential therapeutic activity to target NF-kappaB for cancer treatment or prevention.


Subject(s)
Antineoplastic Agents/isolation & purification , Breast Neoplasms/pathology , NF-kappa B/antagonists & inhibitors , Small Molecule Libraries/analysis , Transcription Factor AP-1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Azo Compounds/pharmacology , Azo Compounds/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , I-kappa B Proteins/metabolism , Models, Biological , NF-kappa B/genetics , Phosphorylation/drug effects , Substrate Specificity , Sulfonic Acids/pharmacology , Sulfonic Acids/therapeutic use , Transcription Factor AP-1/genetics , Transfection
6.
Cancer Res ; 68(5): 1254-60, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18296647

ABSTRACT

Inactivation of tumor suppressors is among the rate-limiting steps in carcinogenesis that occur during the tumor promotion stage. The translation inhibitor programmed cell death 4 (Pdcd4) suppresses tumorigenesis and invasion. Although Pdcd4 is not mutationally inactivated in human cancer, the mechanisms controlling Pdcd4 inactivation during tumorigenesis remain elusive. We report that tumor promoter 12-O-tetradecanoylphorbol-13-acetate exposure decreases protein levels of Pdcd4 in mouse skin papillomas and keratinocytes as well as in human HEK293 cells. This decrease is attributable to increased proteasomal degradation of Pdcd4 and is mediated by protein kinase C-dependent activation of phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin-p70(S6K) and mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK signaling. Both Akt and p70(S6K) phosphorylate Pdcd4, allowing for binding of the E3-ubiquitin ligase beta-TrCP and consequently ubiquitylation. MEK-ERK signaling on the other hand facilitates the subsequent proteasomal degradation. We further show that Pdcd4 protein levels in vivo are limiting for tumor formation, establishing Pdcd4 as a haploinsufficient tumor suppressor in Pdcd4-deficient mice. Thus, because endogenous Pdcd4 levels are limiting for tumorigenesis, inhibiting signaling to Pdcd4 degradation may prove a valid strategy for cancer prevention and intervention.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/physiology , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Skin Neoplasms/metabolism , Animals , Disease Progression , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, Transgenic , Proteasome Endopeptidase Complex/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate
7.
Cancer Res ; 67(6): 2430-8, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363560

ABSTRACT

Activation of activator protein 1 (AP-1) and nuclear factor kappaB (NFkappaB)-dependent transcription is required for tumor promotion in cell culture models and transgenic mice. Dominant-negative c-Jun (TAM67) blocks AP-1 activation by dimerizing with Jun or Fos family proteins and blocks NFkappaB activation by interacting with NFkappaB p65. Two-stage [7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA)] skin carcinogenesis experiments in a model relevant to human cancer risk, transgenic mice expressing human papillomavirus 16 E7 oncogene (K14-HPV16-E7), show E7-enhanced tumor promotion. A cross to K14-TAM67-expressing mice results in dramatic inhibition of tumor promoter-induced AP-1 luciferase reporter activation and papillomagenesis. Epithelial specific TAM67 expression inhibits tumorigenesis without affecting TPA- or E7-induced hyperproliferation of the skin. Thus, the mouse model enriches for TAM67 targets relevant to tumorigenesis rather than to general cell proliferation or hyperplasia, implicating a subset of AP-1- and/or NFkappaB-dependent genes. The aim of the present study was to identify target genes responsible for TAM67 inhibition of DMBA-TPA-induced tumorigenesis. Microarray expression analysis of epidermal tissues revealed small sets of genes in which expression is both up-regulated by tumor promoter and down-regulated by TAM67. Among these, cyclooxygenase-2 (Cox-2/Ptgs2) and osteopontin (Opn/Spp1) are known to be functionally significant in driving carcinogenesis. Results identify both Cox-2 and Opn as transcriptional targets of TAM67 with CRE, but not NFkappaB sites important in the Cox-2 promoter and an AP-1 site important in the Opn promoter.


Subject(s)
Cyclooxygenase 2/genetics , Gene Expression Regulation, Neoplastic/genetics , Osteopontin/genetics , Peptide Fragments/genetics , Proto-Oncogene Proteins c-jun/genetics , Skin Neoplasms/genetics , Transcription Factor AP-1/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins, Viral/genetics , Osteopontin/biosynthesis , Papillomavirus E7 Proteins , Peptide Fragments/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/biosynthesis , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate , Transcription, Genetic , Transfection
8.
Mol Cell Biol ; 27(1): 147-56, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17060447

ABSTRACT

The tumor suppressor function of Programmed Cell Death 4 (Pdcd4) is achieved through interactions between Pdcd4 and components of the translation initiation complex, namely, the RNA helicase eIF4A and the scaffolding protein eIF4G. These interactions are mediated through two MA3 domains on the Pdcd4 molecule and result in inhibition of protein synthesis. We have solved the high-resolution crystal structure of the C-terminal MA3 (cMA3) domain of Pdcd4 in several crystal forms and demonstrated its similarity to the MA3 domain of eIF4G. As predicted by the structure, the cMA3 domain competes with eIF4Gc for binding to eIF4A and surprisingly is sufficient to inhibit translation initiation. Mutations that abolish eIF4A binding negate both functions of the cMA3. Interestingly mutations in the Akt phosphorylation site influenced neither cMA3 binding to eIF4A nor its ability to inhibit translation initiation. Finally, our structural analysis reveals MA3 domains to be a novel subfamily of VHS domains.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/physiology , Gene Expression Regulation , Protein Biosynthesis , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Amino Acid Sequence , Eukaryotic Initiation Factor-4A/physiology , Eukaryotic Initiation Factor-4G/physiology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Two-Hybrid System Techniques
9.
Mol Cell Biol ; 26(4): 1297-306, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449643

ABSTRACT

Programmed cell death 4 (Pdcd4) suppresses neoplastic transformation by inhibiting the activation of c-Jun and consequently AP-1-dependent transcription. We report that Pdcd4 blocks c-Jun activation by inhibiting the expression of mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1)/hematopoietic progenitor kinase 1, a kinase upstream of Jun N-terminal kinase (JNK). cDNA microarray analysis of Pdcd4-overexpressing RKO human colon carcinoma cells revealed MAP4K1 as the sole target of Pdcd4 on the JNK activation pathway. Cotransfection of a MAP4K1 promoter-reporter with Pdcd4 demonstrated inhibition of transcription from the MAP4K1 promoter. Ectopic expression of Pdcd4 in metastatic RKO cells suppressed invasion. MAP4K1 activity is functionally significant in invasion, as overexpression of a dominant negative MAP4K1 (dnMAP4K1) mutant in RKO cells inhibited not only c-Jun activation but also invasion. Overexpression of a MAP4K1 cDNA in Pdcd4-transfected cells rescued the kinase activity of JNK. Thus, Pdcd4 suppresses tumor progression in human colon carcinoma cells by the novel mechanism of down-regulating MAP4K1 transcription, with consequent inhibition of c-Jun activation and AP-1-dependent transcription.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Colonic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Movement , Cloning, Molecular , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA, Neoplasm/genetics , Down-Regulation , Enzyme Activation , Extracellular Matrix/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Oligonucleotide Array Sequence Analysis , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...