Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Biomol Screen ; 21(9): 989-97, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27461835

ABSTRACT

The RAS-MAPK pathway controls many cellular programs, including cell proliferation, differentiation, and apoptosis. In colorectal cancers, recurrent mutations in this pathway often lead to increased cell signaling that may contribute to the development of neoplasms, thereby making this pathway attractive for therapeutic intervention. To this end, we developed a 26-member gene signature of RAS-MAPK pathway activity utilizing the Affymetrix QuantiGene Plex 2.0 reagent system and performed both primary and confirmatory gene expression-based high-throughput screens (GE-HTSs) using KRAS mutant colon cancer cells (SW837) and leveraging a highly annotated chemical library. The screen achieved a hit rate of 1.4% and was able to enrich for hit compounds that target RAS-MAPK pathway members such as MEK and EGFR. Sensitivity and selectivity performance measurements were 0.84 and 1.00, respectively, indicating high true-positive and true-negative rates. Active compounds from the primary screen were confirmed in a dose-response GE-HTS assay, a GE-HTS assay using 14 additional cancer cell lines, and an in vitro colony formation assay. Altogether, our data suggest that this GE-HTS assay will be useful for larger unbiased chemical screens to identify novel compounds and mechanisms that may modulate the RAS-MAPK pathway.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Small Molecule Libraries/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mutation , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Small Molecule Libraries/pharmacology
2.
PLoS One ; 10(12): e0145151, 2015.
Article in English | MEDLINE | ID: mdl-26670328

ABSTRACT

Glucocorticoid signaling regulates target genes by multiple mechanisms, including the repression of transcriptional activities of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) though direct protein-protein interactions and subsequent O-GlcNAcylation of RNA polymerase II (pol II). Recent studies have shown that overexpression of O-linked ß-N-acetylglucosamine transferase (OGT), which adds an O-linked ß-N-acetylglucosamine (O-GlcNAc) group to the C-terminal domain of RNA pol II, increases the transrepression effects of glucocorticoids (GC). As O-GlcNAcase (OGA) is an enzyme that removes O-GlcNAc from O-GlcNAcylated proteins, we hypothesized that the potentiation of GC effects following OGT overexpression could be similarly observed via the direct inhibition of OGA, inhibiting O-GlcNAc removal from pol II. Here we show that despite pharmacological evidence of target engagement by a selective small molecule inhibitor of OGA, there is no evidence for a sensitizing effect on glucocorticoid-mediated effects on TNF-α promoter activity, or gene expression generally, in human cells. Furthermore, inhibition of OGA did not potentiate glucocorticoid-induced apoptosis in several cancer cell lines. Thus, despite evidence for O-GlcNAc modification of RNA pol II in GR-mediated transrepression, our data indicate that pharmacological inhibition of OGA does not potentiate or enhance glucocorticoid-mediated transrepression.


Subject(s)
Enzyme Inhibitors/pharmacology , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Pyrans/pharmacology , Receptors, Glucocorticoid/metabolism , Thiazoles/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Dexamethasone/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inflammation/genetics , Inhibitory Concentration 50 , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , N-Acetylglucosaminyltransferases/metabolism , Prednisolone/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
3.
Mol Cancer Ther ; 14(7): 1532-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931518

ABSTRACT

KRAS is activated by mutation in the vast majority of cases of pancreatic cancer; unfortunately, therapeutic attempts to inhibit KRAS directly have been unsuccessful. Our previous studies showed that inhibition of cyclin-dependent kinase 5 (CDK5) reduces pancreatic cancer growth and progression, through blockage of the centrally important RAL effector pathway, downstream of KRAS. In the current study, the therapeutic effects of combining the CDK inhibitor dinaciclib (SCH727965; MK-7965) with the pan-AKT inhibitor MK-2206 were evaluated using orthotopic and subcutaneous patient-derived human pancreatic cancer xenograft models. The combination of dinaciclib (20 mg/kg, i.p., three times a week) and MK-2206 (60 mg/kg, orally, three times a week) dramatically blocked tumor growth and metastasis in all eight pancreatic cancer models examined. Remarkably, several complete responses were induced by the combination treatment of dinaciclib and MK-2206. The striking results obtained in these models demonstrate that the combination of dinaciclib with the pan-AKT inhibitor MK-2206 is promising for therapeutic evaluation in pancreatic cancer, and strongly suggest that blocking RAL in combination with other effector pathways downstream from KRAS may provide increased efficacy in pancreatic cancer. Based on these data, an NCI-CTEP-approved multicenter phase I clinical trial for pancreatic cancer of the combination of dinaciclib and MK-2206 (NCT01783171) has now been opened.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods , Administration, Oral , Animals , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Proliferation/drug effects , Cyclic N-Oxides , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Drug Administration Schedule , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunohistochemistry , Indolizines , Injections, Intraperitoneal , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/pharmacology , Retinoblastoma Protein/metabolism , Treatment Outcome
4.
PLoS One ; 9(10): e108371, 2014.
Article in English | MEDLINE | ID: mdl-25289887

ABSTRACT

Dinaciclib is a potent CDK1, 2, 5 and 9 inhibitor being developed for the treatment of cancer. Additional understanding of antitumor mechanisms and identification of predictive biomarkers are important for its clinical development. Here we demonstrate that while dinaciclib can effectively block cell cycle progression, in vitro and in vivo studies, coupled with mouse and human pharmacokinetics, support a model whereby induction of apoptosis is a main mechanism of dinaciclib's antitumor effect and relevant to the clinical duration of exposure. This was further underscored by kinetics of dinaciclib-induced downregulation of the antiapoptotic BCL2 family member MCL1 and correlation of sensitivity with the MCL1-to-BCL-xL mRNA ratio or MCL1 amplification in solid tumor models in vitro and in vivo. This MCL1-dependent apoptotic mechanism was additionally supported by synergy with the BCL2, BCL-xL and BCL-w inhibitor navitoclax (ABT-263). These results provide the rationale for investigating MCL1 and BCL-xL as predictive biomarkers for dinaciclib antitumor response and testing combinations with BCL2 family member inhibitors.


Subject(s)
Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Pyridinium Compounds/pharmacology , bcl-X Protein/metabolism , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cyclic N-Oxides , Disease Models, Animal , Diterpenes/pharmacology , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epoxy Compounds/pharmacology , Female , Gene Dosage , Humans , Indolizines , Male , Mice , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasms/genetics , Phenanthrenes/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays , bcl-X Protein/genetics
5.
Mol Cancer Res ; 12(7): 1055-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752990

ABSTRACT

UNLABELLED: TBK1 (TANK-binding kinase 1) is a noncanonical IκB protein kinase that phosphorylates and activates downstream targets such as IRF3 and c-Rel and, mediates NF-κB activation in cancer. Previous reports demonstrated synthetic lethality of TBK1 with mutant KRAS in non-small cell lung cancer (NSCLC); thus, TBK1 could be a novel target for treatment of KRAS-mutant NSCLC. Here, the effect of TBK1 on proliferation in a panel of cancer cells by both genetic and pharmacologic approaches was evaluated. In KRAS-mutant cancer cells, reduction of TBK1 activity by knockdown or treatment with TBK1 inhibitors did not correlate with reduced proliferation in a two-dimensional viability assay. Verification of target engagement via reduced phosphorylation of S386 of IRF3 (pIRF3(S386)) was difficult to assess in NSCLC cells due to low protein expression. However, several cell lines were identified with high pIRF3(S386) levels after screening a large panel of cell lines, many of which also harbor KRAS mutations. Specifically, a large subset of KRAS-mutant pancreatic cancer cell lines was uncovered with high constitutive pIRF3(S386) levels, which correlated with high levels of phosphorylated S172 of TBK1 (pTBK1(S172)). Finally, TBK1 inhibitors dose-dependently inhibited pIRF3(S386) in these cell lines, but this did not correlate with inhibition of cell growth. Taken together, these data demonstrate that the regulation of pathways important for cell proliferation in some NSCLC, pancreatic, and colorectal cell lines is not solely dependent on TBK1 activity. IMPLICATIONS: TBK1 has therapeutic potential under certain contexts and phosphorylation of its downstream target IRF3 is a biomarker of TBK1 activity.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Interferon Regulatory Factor-3/antagonists & inhibitors , Lung Neoplasms/therapy , Neoplasms/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy , Neoplasms/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
6.
Anal Biochem ; 434(1): 96-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23149234

ABSTRACT

Homologous recombination (HR) is a cellular mechanism for accurate repair of double-strand DNA breaks, often deregulated in cancer. Development of novel cancer therapeutics targeting HR pathways would benefit from a quantitative and rapid means of measuring HR. Here, we describe a zinc finger nuclease (ZFN) assay that can quantify HR. Knockdown of BRCA1 or inactivation of BRCA2 decreased HR activity in cells, whereas gene restoration induced activity 8-fold. HR activity was also reflective of BRCA1/2 status in cells with known endogenous mutations.


Subject(s)
Endonucleases/metabolism , Enzyme Assays , Homologous Recombination , BRCA1 Protein/antagonists & inhibitors , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Cell Line, Tumor , HeLa Cells , Humans , RNA Interference , RNA, Small Interfering/metabolism , Zinc Fingers
7.
BMJ Open ; 2(5)2012.
Article in English | MEDLINE | ID: mdl-23065444

ABSTRACT

OBJECTIVES: Anti-phosphorylated histone H3 (pHH3) antibodies specifically detect the core protein histone H3 only when phosphorylated at serine 10 (Ser10) or serine 28 (Ser28). Measurement of pHH3 levels can be used for quantifying mitosis and the effectiveness of mitotic inhibitors in early drug development. However, data on the expression level of pHH3 (Ser10) and pHH3 (Ser28) among different cancers are limited. This study was designed to investigate the expression levels of pHH3 across different types of cancers, using uniform techniques and assay platforms in a single laboratory. DESIGN: Retrospective study. SETTING: Single laboratory. SPECIMENS: Formalin-fixed, paraffin-embedded various human cancer specimens were provided by Mosaic Laboratories Tissue Bank. PRIMARY AND SECONDARY OUTCOME MEASURES: Using immunohistochemistry, pHH3 levels were measured using both pHH3 (Ser10) and (Ser28) antibodies among 10 human melanoma and 10 ovarian tumour samples. The samples were reviewed blindly by two reviewers. pHH3 (Ser10) was then selected to measure the pHH3 levels in cancers of breast, colorectal, oesophageal, gastric, head and neck and lung (n=5 for each cancer). RESULTS: The pHH3 (Ser10) expression was higher than pHH3 (Ser28) in both melanoma and ovarian cancers (p<0.01), with the mean (SD) levels of 1.28% (0.47%) for Ser10 and 0.53% (0.44%) for Ser28 among melanoma and 3.47% (3.51%) for Ser10 and 0.62% (0.68%) for Ser28 among ovarian cancers, respectively. No statistically significant differences were observed among different cancer types tested for pHH3 using Ser10 (p=0.197). No reviewer effect was identified. CONCLUSIONS: The pHH3 Ser10 was significantly higher than Ser28 and may serve as the more robust of two pHH3 assays for measuring mitotic index.

8.
Genes Cancer ; 1(8): 822-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21127729

ABSTRACT

Glioblastoma (GBM) is the most common malignant brain tumor that is characterized by high proliferative rate and invasiveness. Since dysregulation of Notch signaling is implicated in the pathogenesis of many human cancers, here we investigated the role of Notch signaling in GBM. We found that there is aberrant activation of Notch signaling in GBM cell lines and human GBM-derived neurospheres. Inhibition of Notch signaling via the expression of a dominant negative form of the Notch coactivator, mastermind-like 1 (DN-MAML1), or the treatment of a γ-secretase inhibitor, (GSI) MRK-003, resulted in a significant reduction in GBM cell growth in vitro and in vivo. Knockdown of individual Notch receptors revealed that Notch1 and Notch2 receptors differentially contributed to GBM cell growth, with Notch2 having a predominant role. Furthermore, blockade of Notch signaling inhibited the proliferation of human GBM-derived neurospheres in vitro and in vivo. Our overall data indicate that Notch signaling contributes significantly to optimal GBM growth, strongly supporting that the Notch pathway is a promising therapeutic target for GBM.

9.
Anticancer Res ; 30(10): 3853-67, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036696

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) are believed to be responsible for breast cancer formation and recurrence; therefore, therapeutic strategies targeting CSCs must be developed. One approach may be targeting signaling pathways, like Notch, that are involved in stem cell self-renewal and survival. MATERIALS AND METHODS: Breast cancer stem-like cells derived from cell lines and patient samples were examined for Notch expression and activation. The effect of Notch inhibition on sphere formation, proliferation, and colony formation was determined. RESULTS: Breast cancer stem-like cells consistently expressed elevated Notch activation compared with bulk tumor cells. Blockade of Notch signaling using pharmacologic and genomic approaches prevented sphere formation, proliferation, and/or colony formation in soft agar. Interestingly, a gamma-secretase inhibitor, MRK003, induced apoptosis in these cells. CONCLUSION: Our findings support a crucial role for Notch signaling in maintenance of breast cancer stem-like cells, and suggest Notch inhibition may have clinical benefits in targeting CSCs.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptors, Notch/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cyclic S-Oxides/pharmacology , Female , Humans , Neoplastic Stem Cells/drug effects , Receptor, ErbB-2/biosynthesis , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Spheroids, Cellular , Thiadiazoles/pharmacology
10.
Cancer Res ; 70(6): 2476-84, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20197467

ABSTRACT

ERBB2/neu and Notch signaling are known to be deregulated in many human cancers. However, pathway cross-talk and dependencies are not well understood. In this study, we use an ERBB2-transgenic mouse model of breast cancer (neuT) to show that Notch signaling plays a critical role in tumor maintenance. Inhibition of the Notch pathway with a gamma-secretase inhibitor (GSI) decreased both the Notch and the mammalian target of rapamycin/AKT pathways. Antitumor activity resulting from GSI treatment was associated with decreased cell proliferation as measured by Ki67 and decreased expression of glucose transporter Glut1. Positron emission tomography (PET) imaging showed that the functional consequences of decreased Glut1 translated to reduced glucose uptake and correlated with antitumor effects as measured by micro-computed tomography imaging. The decrease of Glut1 in neuT tumors was also observed in several human breast cancer cell lines following GSI treatment. We provide evidence that approximately 27% of ERBB2-positive human breast cancer specimens display high expression of HES1, phospho-S6RP, and GLUT1. Together, these results suggest that pathways downstream of Notch signaling are, at least in part, responsible for promoting tumor growth in neuT and also active in both neuT and a subset of human breast cancers. These findings suggest that GSI may provide therapeutic benefit to a subset of ERBB2-positive breast cancers and that [(18)F]FDG-PET imaging may be useful in monitoring clinical response.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Glucose/pharmacokinetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cyclic S-Oxides/pharmacology , Down-Regulation/drug effects , Female , Fluorodeoxyglucose F18/pharmacokinetics , Glucose Transporter Type 1/biosynthesis , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Inbred BALB C , Mice, Transgenic , Oncogene Protein v-akt/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Thiadiazoles/pharmacology
11.
Blood ; 115(9): 1735-45, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20007543

ABSTRACT

To identify dysregulated pathways in distinct phases of NOTCH1-mediated T-cell leukemogenesis, as well as small-molecule inhibitors that could synergize with or substitute for gamma-secretase inhibitors (GSIs) in T-cell acute lymphoblastic leukemia (T-ALL) therapy, we compared gene expression profiles in a Notch1-induced mouse model of T-ALL with those in human T-ALL. The overall patterns of NOTCH1-mediated gene expression in human and mouse T-ALLs were remarkably similar, as defined early in transformation in the mouse by the regulation of MYC and its target genes and activation of nuclear factor-kappaB and PI3K/AKT pathways. Later events in murine Notch1-mediated leukemogenesis included down-regulation of genes encoding tumor suppressors and negative cell cycle regulators. Gene set enrichment analysis and connectivity map algorithm predicted that small-molecule inhibitors, including heat-shock protein 90, histone deacetylase, PI3K/AKT, and proteasome inhibitors, could reverse the gene expression changes induced by NOTCH1. When tested in vitro, histone deacetylase, PI3K and proteasome inhibitors synergized with GSI in suppressing T-ALL cell growth in GSI-sensitive cells. Interestingly, alvespimycin, a potent inhibitor of the heat-shock protein 90 molecular chaperone, markedly inhibited the growth of both GSI-sensitive and -resistant T-ALL cells, suggesting that its loss disrupts signal transduction pathways crucial for the growth and survival of T-ALL cells.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic/genetics , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Genes, myc , Humans , In Vitro Techniques , Leukemia, Experimental/drug therapy , Leukemia, Experimental/etiology , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1/genetics , Signal Transduction , Species Specificity
12.
Cancer Res ; 69(7): 3060-8, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19318552

ABSTRACT

NOTCH signaling is deregulated in the majority of T-cell acute lymphoblastic leukemias (T-ALL) as a result of activating mutations in NOTCH1. Gamma secretase inhibitors (GSI) block proteolytic activation of NOTCH receptors and may provide a targeted therapy for T-ALL. We have investigated the mechanisms of GSI sensitivity across a panel of T-ALL cell lines, yielding an approach for patient stratification based on pathway activity and also providing a rational combination strategy for enhanced response to GSI. Whereas the NOTCH1 mutation status does not serve as a predictor of GSI sensitivity, a gene expression signature of NOTCH pathway activity does correlate with response, and may be useful in the selection of patients more likely to respond to GSI. Furthermore, inhibition of the NOTCH pathway activity signature correlates with the induction of the cyclin-dependent kinase inhibitors CDKN2D (p19(INK4d)) and CDKN1B (p27(Kip1)), leading to derepression of RB and subsequent exit from the cell cycle. Consistent with this evidence of cell cycle exit, short-term exposure of GSI resulted in sustained molecular and phenotypic effects after withdrawal of the compound. Combination treatment with GSI and a small molecule inhibitor of CDK4 produced synergistic growth inhibition, providing evidence that GSI engagement of the CDK4/RB pathway is an important mechanism of GSI action and supports further investigation of this combination for improved efficacy in treating T-ALL.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cyclic S-Oxides/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protease Inhibitors/pharmacology , Receptor, Notch1/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Thiadiazoles/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p19/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27 , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Profiling , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , S Phase/drug effects , S Phase/genetics , Signal Transduction/drug effects , Transcription, Genetic , Transfection
13.
Cancer Res ; 68(23): 9678-85, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047145

ABSTRACT

Malignant mesothelioma (MM) is a cancer of the lining of the lungs, heart, and intestine and is known to respond poorly to chemotherapy. Here we show that malignant mesothelial cells have an elevated Notch signaling pathway compared with normal human mesothelial cells. We studied the role of Notch in MM under normoxic and hypoxic conditions, the latter condition best recapitulating the MM microenvironment. Genetic and chemical modulation of the Notch pathway indicated that MM cells are dependent on Notch signaling. More specifically, this signaling was Notch-1 dependent as the result of its negative transcriptional regulation on phosphatase and tensin homologue (PTEN), which led to activation of the prosurvival phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway. Our study also provides evidence that whereas Notch-1 is elevated in the malignant setting, Notch-2 is diminished. This differential expression of the two Notch isoforms benefits cancer cell survival because reexpression of Notch-2 was toxic to MM cells. The mechanism of Notch-2 toxicity to MM cells countered that of Notch-1, as it was the result of positive transcriptional regulation of PTEN and inhibition of the PI3K/Akt/mTOR signaling pathway. These results provide new insight into the role of Notch in MM and suggest that Notch pathway inhibitors may be useful in the treatment of this deadly disease.


Subject(s)
Mesothelioma/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Cell Growth Processes/physiology , Cell Hypoxia , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA, Neoplasm/biosynthesis , Humans , Mesothelioma/genetics , Mesothelioma/pathology , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/biosynthesis , Receptor, Notch2/biosynthesis , Signal Transduction , Tumor Cells, Cultured
14.
Clin Cancer Res ; 14(18): 5735-42, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794082

ABSTRACT

PURPOSE: For many tumor cells, de novo lipogenesis is a requirement for growth and survival. A considerable body of work suggests that inhibition of this pathway may be a powerful approach to antineoplastic therapy. It has recently been shown that inhibition of various steps in the lipogenic pathway individually can induce apoptosis or loss of viability in tumor cells. However, it is not clear whether quantitative differences exist in the ability of lipogenic enzymes to control tumor cell survival. We present a systematic approach that allows for a direct comparison of the control of lipogenic pathway enzymes over tumor cell growth and apoptosis using different cancer cells. EXPERIMENTAL DESIGN: RNA interference-mediated, graded down-regulation of fatty acid synthase (FAS) pathway enzymes was employed in combination with measurements of lipogenesis, apoptosis, and cell growth. RESULTS: In applying RNA interference titrations to two lipogenic enzymes, acetyl-CoA carboxylase 1 (ACC1) and FAS, we show that ACC1 and FAS both significantly control cell growth and apoptosis in HCT-116 cells. These results also extend to PC-3 and A2780 cancer cells. CONCLUSIONS: Control of tumor cell survival by different steps in de novo lipogenesis can be quantified. Because ACC1 and FAS both significantly control tumor cell growth and apoptosis, we propose that pharmacologic inhibitors of either enzyme might be useful agents in targeting cancer cells that critically rely on fatty acid synthesis. The experimental approach described here may be extended to other targets or disease-relevant pathways to identify steps suitable for therapeutic intervention.


Subject(s)
Acetyltransferases/metabolism , Colonic Neoplasms/enzymology , Fatty Acid Synthases/metabolism , Cell Line , Cell Proliferation , Cell Survival , Enzyme Inhibitors/pharmacology , HCT116 Cells , Humans , Lipogenesis , Signal Transduction , Transfection
15.
Cancer Res ; 67(17): 7954-9, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17804701

ABSTRACT

NOTCH signaling is an evolutionarily conserved signaling pathway that regulates cell fate during development and postnatal life. It has been increasingly linked to carcinogenesis, although its role in cancer seems to be highly context and tissue specific. Although NOTCH signaling is required for lung development, little is known about its role in lung cancer. In this study, we show that NOTCH signaling, as measured by the gamma-secretase cleavage product N(IC)-1, is active in both normal human and lung tumor samples; however, downstream NOTCH readouts (i.e., HES-1 and HES-5) are elevated in lung tumors. Levels of NOTCH signaling components in primary human lung cells reflect observations in tissue samples, yet lung tumor cell lines showed little NOTCH signaling. Because oxygen concentrations are important in normal lung physiology and lung tumors are hypoxic, the effect of low oxygen on these lung tumor cell lines was evaluated. We found that hypoxia dramatically elevates NOTCH signaling (especially NOTCH-1) in lung tumor cell lines and concomitantly sensitizes them to inhibition via small-molecule gamma-secretase inhibitors or NOTCH-1 RNA interference. gamma-Secretase inhibitor-induced apoptosis of lung tumor cells grown under hypoxic conditions could be rescued by reintroduction of active NOTCH-1. Our data strengthen the role of NOTCH in lung cancer and as a therapeutic target for the treatment of lung and other hypoxic tumor types.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/genetics , Oxygen/pharmacology , Receptor, Notch1/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/genetics , Cell Hypoxia/physiology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oxygen/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured
16.
J Exp Med ; 204(8): 1813-24, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-17646409

ABSTRACT

gamma-secretase inhibitors (GSIs) can block NOTCH receptor signaling in vitro and therefore offer an attractive targeted therapy for tumors dependent on deregulated NOTCH activity. To clarify the basis for GSI resistance in T cell acute lymphoblastic leukemia (T-ALL), we studied T-ALL cell lines with constitutive expression of the NOTCH intracellular domain (NICD), but that lacked C-terminal truncating mutations in NOTCH1. Each of the seven cell lines examined and 7 of 81 (8.6%) primary T-ALL samples harbored either a mutation or homozygous deletion of the gene FBW7, a ubiquitin ligase implicated in NICD turnover. Indeed, we show that FBW7 mutants cannot bind to the NICD and define the phosphodegron region of the NICD required for FBW7 binding. Although the mutant forms of FBW7 were still able to bind to MYC, they do not target it for degradation, suggesting that stabilization of both NICD and its principle downstream target, MYC, may contribute to transformation in leukemias with FBW7 mutations. In addition, we show that all seven leukemic cell lines with FBW7 mutations were resistant to the MRK-003 GSI. Most of these resistant lines also failed to down-regulate the mRNA levels of the NOTCH targets MYC and DELTEX1 after treatment with MRK-003, implying that residual NOTCH signaling in T-ALLs with FBW7 mutations contributes to GSI resistance.


Subject(s)
Cell Cycle Proteins/genetics , Enzyme Inhibitors/pharmacology , F-Box Proteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia/genetics , Leukemia/metabolism , Mutation , Receptors, Notch/genetics , Ubiquitin-Protein Ligases/genetics , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Apoptosis , Cell Cycle , Cell Line, Tumor , F-Box-WD Repeat-Containing Protein 7 , Humans , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Messenger/metabolism , Receptors, Notch/metabolism
17.
Chem Biol ; 14(2): 209-19, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17317574

ABSTRACT

In this report, inhibitors of the gamma-secretase enzyme have been exploited to characterize the antiproliferative relationship between target inhibition and cellular responses in Notch-dependent human T cell acute lymphoblastic leukemia (T-ALL) cell lines. Inhibition of gamma-secretase led to decreased Notch signaling, measured by endogenous NOTCH intracellular domain (NICD) formation, and was associated with decreased cell viability. Flow cytometry revealed that decreased cell viability resulted from a G(0)/G(1) cell cycle block, which correlated strongly to the induction of apoptosis. These effects associated with inhibitor treatment were rescued by exogenous expression of NICD and were not mirrored when a markedly less active enantiomer was used, demonstrating the gamma-secretase dependency and specificity of these responses. Together, these data strengthen the rationale for using gamma-secretase inhibitors therapeutically and suggest that programmed cell death may contribute to reduction of tumor burden in the clinic.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/pathology , Receptors, Notch/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic S-Oxides/pharmacology , Flow Cytometry , Humans , Leukemia-Lymphoma, Adult T-Cell/enzymology , Leukemia-Lymphoma, Adult T-Cell/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Thiadiazoles/pharmacology
19.
Cancer Res ; 65(17): 7533-40, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140914

ABSTRACT

The response of eukaryotic cells to DNA damage includes the activation of phosphatidylinositol-3 kinase-related kinases (PIKK), such as ATM, ATR, and DNA-dependent protein kinase (DNA-PK). These three kinases have very similar substrate specificities in vitro, but in vivo, their substrates overlap only partially. Several in vivo substrates of ATM and ATR have been identified and almost all of them are involved in DNA damage-induced cell cycle arrest and/or apoptosis. In contrast, few in vivo substrates of DNA-PK have been identified. These include histone H2AX and DNA-PK itself. We identify here valosin-containing protein (VCP) as a novel substrate of DNA-PK and other PIKK family members. VCP is phosphorylated at Ser784 within its COOH terminus, a region previously shown to target VCP to specific intracellular compartments. Furthermore, VCP phosphorylated at Ser784 accumulated at sites of DNA double-strand breaks (DSBs). VCP is a protein chaperone that unfolds and translocates proteins. Its phosphorylation in response to DNA damage and its recruitment to sites of DNA DSBs could indicate a role of VCP in DNA repair.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/physiology , Adenosine Triphosphatases , Amino Acid Sequence , Antibodies/pharmacology , Cell Line, Tumor , Checkpoint Kinase 2 , DNA, Neoplasm/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Transfection , Valosin Containing Protein
20.
Proc Natl Acad Sci U S A ; 99(23): 14734-9, 2002 Nov 12.
Article in English | MEDLINE | ID: mdl-12407176

ABSTRACT

The oncoprotein hdm2 ubiquitinates p53, resulting in the rapid degradation of p53 through the ubiquitin (Ub)-proteasome pathway. Hdm2-mediated destabilization and inactivation of p53 are thought to play a critical role in a number of human cancers. We have used an in vitro enzyme assay, monitoring hdm2-catalyzed Ub transfer from preconjugated Ub-Ubc4 to p53, to identify small molecule inhibitors of this enzyme. Three chemically distinct types of inhibitors were identified this way, each with potency in the micromolar range. All three types of compounds display selective inhibition of hdm2 E3 ligase activity, with little or no effect on other Ub-using enzymes. Most strikingly, these compounds do not inhibit the autoubiquitination activity of hdm2. Steady-state analysis reveals that all three classes behave as simple reversible inhibitors of the enzyme and that they are noncompetitive with respect to both substrates, Ub-Ubc4 and p53. Studies of the effects of combinations of two inhibitory molecules on hdm2 activity indicate that the three types of compounds bind in a mutually exclusive fashion, suggesting a common binding site on hdm2 for all of these inhibitors. These compounds establish the feasibility of selectively blocking hdm2-mediated ubiquitination of p53 by small molecule inhibitors. Selective inhibitors of hdm2 E3 ligase activity could provide a novel mechanism for the development of new chemotherapeutics for the treatment of human cancers.


Subject(s)
Nuclear Proteins , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases , Ubiquitin/metabolism , Biotinylation , Calcium-Binding Proteins/metabolism , Catalysis , Endosomal Sorting Complexes Required for Transport , Humans , Kinetics , Ligases/metabolism , Models, Biological , Nedd4 Ubiquitin Protein Ligases , Peptide Synthases/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2
SELECTION OF CITATIONS
SEARCH DETAIL
...