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










Publication year range
1.
Cell Chem Biol ; 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38537632

ABSTRACT

This study describes the identification and target deconvolution of small molecule inhibitors of oncogenic Yes-associated protein (YAP1)/TAZ activity with potent anti-tumor activity in vivo. A high-throughput screen (HTS) of 3.8 million compounds was conducted using a cellular YAP1/TAZ reporter assay. Target deconvolution studies identified the geranylgeranyltransferase-I (GGTase-I) complex as the direct target of YAP1/TAZ pathway inhibitors. The small molecule inhibitors block the activation of Rho-GTPases, leading to subsequent inactivation of YAP1/TAZ and inhibition of cancer cell proliferation in vitro. Multi-parameter optimization resulted in BAY-593, an in vivo probe with favorable PK properties, which demonstrated anti-tumor activity and blockade of YAP1/TAZ signaling in vivo.

2.
Clin Exp Med ; 23(8): 5445-5461, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37935952

ABSTRACT

The PI3K pathway is one of the most frequently altered signaling pathways in human cancer. In addition to its function in cancer cells, PI3K plays a complex role in modulating anti-tumor immune responses upon immune checkpoint inhibition (ICI). Here, we evaluated the effects of the pan-Class I PI3K inhibitor copanlisib on different immune cell types in vitro and on tumor growth and immune cell infiltration in syngeneic murine cancer models. Intermittent treatment with copanlisib resulted in a strong in vivo anti-tumor efficacy, increased tumor infiltration of activated T cells and macrophages, and increased CD8+ T cell/regulatory T cell and M1/M2 macrophage ratios. The strong in vivo efficacy was at least partially due to immunomodulatory activity of copanlisib, as in vitro these murine cancer cells were resistant to PI3K inhibition. Furthermore, the combination of copanlisib with the ICI antibody anti-PD-1 demonstrated enhanced anti-tumor efficacy in both ICI-sensitive and insensitive syngeneic mouse tumor models. Importantly, in an ICI-sensitive model, combination therapy resulted in complete remission and prevention of tumor recurrence. Thus, the combination of ICIs with PI3K inhibition by intermittently dosed copanlisib represents a promising new strategy to increase sensitivity to ICI therapies and to treat human solid cancers.


Subject(s)
Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Animals , Mice , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/metabolism , Neoplasms/drug therapy , Immunity , Tumor Microenvironment
3.
Nat Cancer ; 3(4): 471-485, 2022 04.
Article in English | MEDLINE | ID: mdl-35484422

ABSTRACT

Aberrant expression of MYC transcription factor family members predicts poor clinical outcome in many human cancers. Oncogenic MYC profoundly alters metabolism and mediates an antioxidant response to maintain redox balance. Here we show that MYCN induces massive lipid peroxidation on depletion of cysteine, the rate-limiting amino acid for glutathione (GSH) biosynthesis, and sensitizes cells to ferroptosis, an oxidative, non-apoptotic and iron-dependent type of cell death. The high cysteine demand of MYCN-amplified childhood neuroblastoma is met by uptake and transsulfuration. When uptake is limited, cysteine usage for protein synthesis is maintained at the expense of GSH triggering ferroptosis and potentially contributing to spontaneous tumor regression in low-risk neuroblastomas. Pharmacological inhibition of both cystine uptake and transsulfuration combined with GPX4 inactivation resulted in tumor remission in an orthotopic MYCN-amplified neuroblastoma model. These findings provide a proof of concept of combining multiple ferroptosis targets as a promising therapeutic strategy for aggressive MYCN-amplified tumors.


Subject(s)
Ferroptosis , Neuroblastoma , Cell Death , Child , Cysteine/therapeutic use , Ferroptosis/genetics , Glutathione/therapeutic use , Humans , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics
4.
J Med Chem ; 64(21): 15883-15911, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34699202

ABSTRACT

PIP4K2A is an insufficiently studied type II lipid kinase that catalyzes the conversion of phosphatidylinositol-5-phosphate (PI5P) into phosphatidylinositol 4,5-bisphosphate (PI4,5P2). The involvement of PIP4K2A/B in cancer has been suggested, particularly in the context of p53 mutant/null tumors. PIP4K2A/B depletion has been shown to induce tumor growth inhibition, possibly due to hyperactivation of AKT and reactive oxygen species-mediated apoptosis. Herein, we report the identification of the novel potent and highly selective inhibitors BAY-091 and BAY-297 of the kinase PIP4K2A by high-throughput screening and subsequent structure-based optimization. Cellular target engagement of BAY-091 and BAY-297 was demonstrated using cellular thermal shift assay technology. However, inhibition of PIP4K2A with BAY-091 or BAY-297 did not translate into the hypothesized mode of action and antiproliferative activity in p53-deficient tumor cells. Therefore, BAY-091 and BAY-297 serve as valuable chemical probes to study PIP4K2A signaling and its involvement in pathophysiological conditions such as cancer.


Subject(s)
Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Naphthyridines/chemistry , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
5.
J Med Chem ; 62(2): 928-940, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30563338

ABSTRACT

The availability of a chemical probe to study the role of a specific domain of a protein in a concentration- and time-dependent manner is of high value. Herein, we report the identification of a highly potent and selective ERK5 inhibitor BAY-885 by high-throughput screening and subsequent structure-based optimization. ERK5 is a key integrator of cellular signal transduction, and it has been shown to play a role in various cellular processes such as proliferation, differentiation, apoptosis, and cell survival. We could demonstrate that inhibition of ERK5 kinase and transcriptional activity with a small molecule did not translate into antiproliferative activity in different relevant cell models, which is in contrast to the results obtained by RNAi technology.


Subject(s)
Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Apoptosis/drug effects , Binding Sites , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , Half-Life , Humans , Mitogen-Activated Protein Kinase 7/metabolism , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyridines/metabolism , Pyridines/pharmacology , Pyrimidines/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription, Genetic/drug effects
6.
Cell Death Dis ; 9(2): 192, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416006

ABSTRACT

Cancer stem cells (CSCs) are involved in metastasis and resistance development, thus affecting anticancer therapy efficacy. The underlying pathways required for CSC maintenance and survival are not fully understood and only a limited number of treatment strategies to specifically target CSCs have been identified. To identify novel CSC targeting compounds, we here set-up an aldehyde dehydrogenase (ALDH)-based phenotypic screening system that allows for an automated and standardized identification of CSCs. By staining cancer cells for ALDH activity and applying high-content-based single-cell population analysis, the proportion of a potential CSC subpopulation with significantly higher ALDH activity (ALDHhigh) can be quantified in a heterogeneous cell population. We confirmed high ALDH activity as surrogate marker for the CSC subpopulation in vitro and validated Wnt signaling as an essential factor for the maintenance of CSCs in SUM149 breast cancer cells. In a small molecule screen, we identified phosphodiesterase type 5 (PDE5) inhibition as potential strategy to target CSC maintenance and survival in multiple cancer cell lines. CSC elimination by PDE5 inhibition was not dependent on PKG signaling, and we suggest a novel mechanism in which PDE5 inhibition leads to elevated cGMP levels that stimulate cAMP/PKA signaling to eliminate CSCs.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Phosphodiesterase 5 Inhibitors/pharmacology , Aldehyde Dehydrogenase/metabolism , Cell Line, Tumor , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Humans , Mastodynia/drug therapy , Mastodynia/enzymology , Mastodynia/pathology , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Wnt Signaling Pathway
7.
Handb Exp Pharmacol ; 232: 25-41, 2016.
Article in English | MEDLINE | ID: mdl-26330261

ABSTRACT

Targeted therapies in personalized medicine require the knowledge about the molecular changes within the patient that cause the disease. With the beginning of the new century, a plethora of new technologies became available to detect these changes and use this information as starting point for drug development. Next-generation genome sequencing and sophisticated genome-wide functional genomics' methods have led to a significant increase in the identification of novel drug target candidates and understanding of the relevance of these genomic and molecular changes for the diseases. As functional genomic tool for target identification, high-throughput gene silencing through RNA interference screening has become the established method. RNAi is discussed with its advantages and challenges in this chapter. Furthermore the potential of CRISPR/Cas9, a gene-editing method that has recently been adapted for use as functional screening tool, will be briefly reviewed.


Subject(s)
Drug Discovery , Genomics , High-Throughput Nucleotide Sequencing , RNA Interference
8.
PLoS One ; 10(9): e0137640, 2015.
Article in English | MEDLINE | ID: mdl-26361354

ABSTRACT

Functional RNAi based screening is affected by large numbers of false positive and negative hits due to prevalent sequence based off-target effects. We performed a druggable genome targeting siRNA screen intended to identify novel regulators of E-cadherin (CDH1) expression, a known key player in epithelial mesenchymal transition (EMT). Analysis of primary screening results indicated a large number of false-positive hits. To address these crucial difficulties we developed an analysis method, SENSORS, which, similar to published methods, is a seed enrichment strategy for analyzing siRNA off-targets in RNAi screens. Using our approach, we were able to demonstrate that accounting for seed based off-target effects stratifies primary screening results and enables the discovery of additional screening hits. While traditional hit detection methods are prone to false positive results which are undetected, we were able to identify false positive hits robustly. Transcription factor MYBL1 was identified as a putative novel target required for CDH1 expression and verified experimentally. No siRNA pool targeting MYBL1 was present in the used siRNA library. Instead, MYBL1 was identified as a putative CDH1 regulating target solely based on the SENSORS off-target score, i.e. as a gene that is a cause for off-target effects down regulating E-cadherin expression.


Subject(s)
Cadherins/metabolism , High-Throughput Screening Assays/methods , RNA, Small Interfering/genetics , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Genome, Human , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Sensitivity and Specificity , Trans-Activators/genetics , Trans-Activators/metabolism
9.
J Pathol ; 226(3): 482-94, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21953249

ABSTRACT

Chromosomal instability (CIN) has been implicated in multidrug resistance and the silencing of the ceramide transporter, CERT, promotes sensitization to diverse cytotoxics. An improved understanding of mechanisms governing multidrug sensitization might provide insight into pathways contributing to the death of CIN cancer cells. Using an integrative functional genomics approach, we find that CERT-specific multidrug sensitization is associated with enhanced autophagosome-lysosome flux, resulting from the expression of LAMP2 following CERT silencing in colorectal and HER2(+) breast cancer cell lines. Live cell microscopy analysis revealed that CERT depletion induces LAMP2-dependent death of polyploid cells following exit from mitosis in the presence of paclitaxel. We find that CERT is relatively over-expressed in HER2(+) breast cancer and CERT protein expression acts as an independent prognostic variable and predictor of outcome in adjuvant chemotherapy-treated patients with primary breast cancer. These data suggest that the induction of LAMP2-dependent autophagic flux through CERT targeting may provide a rational approach to enhance multidrug sensitization and potentiate the death of polyploid cells following paclitaxel exposure to limit the acquisition of CIN and intra-tumour heterogeneity.


Subject(s)
Autophagy/physiology , Breast Neoplasms/drug therapy , Chromosomal Instability/physiology , Protein Serine-Threonine Kinases/deficiency , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/genetics , Ceramides/metabolism , Ceramides/pharmacology , Cisplatin/pharmacology , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Female , Gene Expression , Gene Silencing/physiology , Humans , Lysosomal-Associated Membrane Protein 2 , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/physiology , Middle Aged , Mitosis Modulators/pharmacology , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptor, ErbB-2 , Tumor Cells, Cultured
10.
Genome Med ; 2(8): 53, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20701793

ABSTRACT

The European Union multi-disciplinary Personalised RNA interference to Enhance the Delivery of Individualised Cytotoxic and Targeted therapeutics (PREDICT) consortium has recently initiated a framework to accelerate the development of predictive biomarkers of individual patient response to anti-cancer agents. The consortium focuses on the identification of reliable predictive biomarkers to approved agents with anti-angiogenic activity for which no reliable predictive biomarkers exist: sunitinib, a multi-targeted tyrosine kinase inhibitor and everolimus, a mammalian target of rapamycin (mTOR) pathway inhibitor. Through the analysis of tumor tissue derived from pre-operative renal cell carcinoma (RCC) clinical trials, the PREDICT consortium will use established and novel methods to integrate comprehensive tumor-derived genomic data with personalized tumor-derived small hairpin RNA and high-throughput small interfering RNA screens to identify and validate functionally important genomic or transcriptomic predictive biomarkers of individual drug response in patients. PREDICT's approach to predictive biomarker discovery differs from conventional associative learning approaches, which can be susceptible to the detection of chance associations that lead to overestimation of true clinical accuracy. These methods will identify molecular pathways important for survival and growth of RCC cells and particular targets suitable for therapeutic development. Importantly, our results may enable individualized treatment of RCC, reducing ineffective therapy in drug-resistant disease, leading to improved quality of life and higher cost efficiency, which in turn should broaden patient access to beneficial therapeutics, thereby enhancing clinical outcome and cancer survival. The consortium will also establish and consolidate a European network providing the technological and clinical platform for large-scale functional genomic biomarker discovery. Here we review our current understanding of molecular mechanisms driving resistance to anti-angiogenesis agents, the current limitations of laboratory and clinical trial strategies and how the PREDICT consortium will endeavor to identify a new generation of predictive biomarkers.

11.
Proc Natl Acad Sci U S A ; 106(21): 8671-6, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19458043

ABSTRACT

Microtubule-stabilizing (MTS) agents, such as taxanes, are important chemotherapeutics with a poorly understood mechanism of action. We identified a set of genes repressed in multiple cell lines in response to MTS agents and observed that these genes are overexpressed in tumors exhibiting chromosomal instability (CIN). Silencing 22/50 of these genes, many of which are involved in DNA repair, caused cancer cell death, suggesting that these genes are involved in the survival of aneuploid cells. Overexpression of these "CIN-survival" genes is associated with poor outcome in estrogen receptor-positive breast cancer and occurs frequently in basal-like and Her2-positive cases. In diploid cells, but not in chromosomally unstable cells, paclitaxel causes repression of CIN-survival genes, followed by cell death. In the OV01 ovarian cancer clinical trial, a high level of CIN was associated with taxane resistance but carboplatin sensitivity, indicating that CIN may determine MTS response in vivo. Thus, pretherapeutic assessment of CIN may optimize treatment stratification and clinical trial design using these agents.


Subject(s)
Bridged-Ring Compounds/pharmacology , Chromosomal Instability/drug effects , Chromosomal Instability/genetics , Taxoids/pharmacology , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Microtubules/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Paclitaxel/toxicity , Polymerase Chain Reaction , Prognosis
13.
Cancer Cell ; 12(6): 514-27, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18068629

ABSTRACT

The extracellular matrix (ECM) can induce chemotherapy resistance via AKT-mediated inhibition of apoptosis. Here, we show that loss of the ECM protein TGFBI (transforming growth factor beta induced) is sufficient to induce specific resistance to paclitaxel and mitotic spindle abnormalities in ovarian cancer cells. Paclitaxel-resistant cells treated with recombinant TGFBI protein show integrin-dependent restoration of paclitaxel sensitivity via FAK- and Rho-dependent stabilization of microtubules. Immunohistochemical staining for TGFBI in paclitaxel-treated ovarian cancers from a prospective clinical trial showed that morphological changes of paclitaxel-induced cytotoxicity were restricted to areas of strong expression of TGFBI. These data show that ECM can mediate taxane sensitivity by modulating microtubule stability.


Subject(s)
Extracellular Matrix Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Transforming Growth Factor beta/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Adhesion/drug effects , Cell Death/drug effects , Cell Line, Tumor , Centrosome/drug effects , Centrosome/metabolism , Drug Resistance, Neoplasm/drug effects , Extracellular Matrix Proteins/deficiency , Female , Fibronectins/metabolism , Gene Silencing/drug effects , Humans , Integrins/metabolism , Mitosis/drug effects , Models, Biological , Ovarian Neoplasms/pathology , Protein Transport/drug effects , Recombinant Proteins/metabolism , Transforming Growth Factor beta/deficiency , Tubulin/metabolism
14.
Cell Cycle ; 6(16): 2001-4, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17721083

ABSTRACT

The sequential use of non cross-resistant cytotoxic agents is the standard of care for advanced solid tumors in order to enhance survival and optimise quality of life. Nevertheless, drug resistance to non cross-resistant agents is commonly witnessed, with clinical response rates to non cross-resistant regimens declining as the disease advances. Expression of ABC transporters is unlikely to fully explain this phenomenon, and a clear molecular explanation for this process remains uncertain. A statistical analysis of a recently published RNA interference screen targeting 779 kinases in three cell lines deriving from different tumor types reveals a significant correlation between resistance to paclitaxel and a non cross-resistant cytotoxic agent. Furthermore, 20% of kinases that promote resistance to paclitaxel when targeted by RNAi also promote resistance to a non cross-resistant agent within that same cell line, consistent with a tissue-type dependence of multi-drug resistance. Conversely, paclitaxel-specific resistance and sensitising kinases occur less frequently than expected. This indicates that several cell line specific kinases may regulate multi-drug resistance and provide a potential explanation for the phenomenon of drug resistance to non cross-resistant agents witnessed in oncology practice. Furthermore, this work supports efforts to identify common pathways of drug response for future drug discovery programmes.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/genetics , Phosphotransferases/genetics , RNA, Small Interfering/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , HCT116 Cells , Humans , Paclitaxel/pharmacology , Phosphotransferases/metabolism , Transfection
15.
Cell ; 129(5): 957-68, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17540175

ABSTRACT

Ras proteins signal through direct interaction with a number of effector enzymes, including type I phosphoinositide (PI) 3-kinases. Although the ability of Ras to control PI 3-kinase has been well established in manipulated cell culture models, evidence for a role of the interaction of endogenous Ras with PI 3-kinase in normal and malignant cell growth in vivo has been lacking. Here we generate mice with mutations in the Pi3kca gene encoding the catalytic p110alpha isoform that block its interaction with Ras. Cells from these mice show proliferative defects and selective disruption of signaling from growth factors to PI 3-kinase. The mice display defective development of the lymphatic vasculature, resulting in perinatal appearance of chylous ascites. Most importantly, they are highly resistant to endogenous Ras oncogene-induced tumorigenesis. The interaction of Ras with p110alpha is thus required in vivo for certain normal growth factor signaling and for Ras-driven tumor formation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Cattle , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphatic Abnormalities/genetics , Lymphatic Abnormalities/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/genetics , Point Mutation , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Sequence Alignment , Signal Transduction
16.
Cancer Cell ; 11(6): 498-512, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17560332

ABSTRACT

Cytotoxic drug resistance is a major cause of cancer treatment failure. We report an RNA interference screen to identify genes influencing sensitivity of different cancer cell types to chemotherapeutic agents. A set of genes whose targeting leads to resistance to paclitaxel is identified, many of which are involved in the spindle assembly checkpoint. Silencing these genes attenuates paclitaxel-induced mitotic arrest and induces polyploidy in the absence of drug. We also identify a ceramide transport protein, COL4A3BP or CERT, whose downregulation sensitizes cancer cells to multiple cytotoxic agents, potentiating endoplasmic reticulum stress. COL4A3BP expression is increased in drug-resistant cell lines and in residual tumor following paclitaxel treatment of ovarian cancer, suggesting that it could be a target for chemotherapy-resistant cancers.


Subject(s)
Ceramides/metabolism , Drug Resistance, Neoplasm , Mitosis , Paclitaxel/pharmacology , Protein Serine-Threonine Kinases/physiology , Cell Line, Tumor , Cell Survival/drug effects , Chromosomal Instability , Down-Regulation , Drug Resistance, Multiple/genetics , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Paclitaxel/therapeutic use , Polyploidy , Protein Kinases/metabolism , RNA, Small Interfering/pharmacology
17.
Mol Cell ; 20(5): 673-85, 2005 Dec 09.
Article in English | MEDLINE | ID: mdl-16337592

ABSTRACT

The ability of activated Ras to induce growth arrest of human ovarian surface epithelial (HOSE) cells via induction of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) has been used to screen for Ras pathway signaling components using a library of RNA interference (RNAi) vectors targeting the kinome. Two known Ras-regulated kinases were identified, phosphoinositide 3-kinase p110alpha and ribosomal protein S6 kinase p70(S6K1), plus the MAP kinase kinase kinase kinase MINK, which had not previously been implicated in Ras signaling. MINK is activated after Ras induction via a mechanism involving reactive oxygen species and mediates stimulation of the stress-activated protein kinase p38 MAPK downstream of the Raf/ERK pathway. p38 MAPK activation is essential for Ras-induced p21(WAF1/CIP1) upregulation and cell cycle arrest. MINK is thus a distal target of Ras signaling in the induction of a growth-arrested, senescent-like phenotype that may act to oppose oncogenic transformation in HOSE cells.


Subject(s)
Epithelial Cells/enzymology , Ovarian Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/physiology , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases , Phenotype , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/pharmacology , RNA Interference/physiology , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae Proteins/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Mol Biol Cell ; 15(7): 3450-63, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15090615

ABSTRACT

The Raf protein kinases are major effectors of Ras GTPases and key components of the transcriptional response to serum factors, acting at least in part through the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway. It has recently been suggested that Raf also may trigger other as yet uncharacterized signaling pathways. Here, we have used cDNA microarrays to dissect changes in gene expression induced by activation of inducible c-Raf-1 constructs in human mammary epithelial and ovarian epithelial cells. The majority of Raf-induced transcriptional responses are shown to be blocked by pharmacological inhibition of the Raf substrate mitogen-activated protein kinase kinase, indicating that potential mitogen-activated protein kinase kinase-independent Raf signaling pathways have no significant influence on gene expression. In addition, we used epidermal growth factor receptor inhibitory drugs to address the contribution of autocrine signaling by Raf-induced EGF family proteins to the Raf transcriptional response. At least one-half of the transcription induced by Raf activation requires epidermal growth factor (EGF) receptor function The EGF receptor-independent component of the Raf transcriptional response is entirely up-regulation of gene expression, whereas the EGF receptor-dependent component is an equal mixture of up- and down-regulation. The use of transcriptional profiling in this way allows detailed analysis of the architecture of signaling pathways to be undertaken.


Subject(s)
Autocrine Communication , Gene Expression Regulation/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Cell Line , Enzyme Activation/genetics , ErbB Receptors/antagonists & inhibitors , Gene Expression Profiling , Humans , Hydroxytestosterones/pharmacology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-raf/genetics , Transcription, Genetic
20.
Cancer Res ; 63(17): 5344-51, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500366

ABSTRACT

RASSF1A is a recently identified 3p21.3 tumor suppressor gene. The high frequency of epigenetic inactivation of this gene in a wide range of human sporadic cancers including non-small cell lung cancer (NSCLC) and neuroblastoma suggests that RASSF1A inactivation is important for tumor development. Although little is known about the function of RASSF1A, preliminary data suggests that it may have multiple functions. To gain insight into RASSF1A functions in an unbiased manner, we have characterized the expression profile of a lung cancer cell line (A549) transfected with RASSF1A. Initially we demonstrated that transient expression of RASSF1A into the NSCLC cell line A549 induced G(1) cell cycle arrest, as measured by propidium iodide staining. Furthermore, annexin-V staining showed that RASSF1A-expressing cells had an increased sensitivity to staurosporine-induced apoptosis. We then screened a cDNA microarray containing more than 6000 probes to identify genes differentially regulated by RASSF1A. Sixty-six genes showed at least a 2-fold change in expression. Among these were many genes with relevance to tumorigenesis involved in transcription, cytoskeleton, signaling, cell cycle, cell adhesion, and apoptosis. For 22 genes we confirmed the microarray results by real-time RT-PCR and/or Northern blotting. In silico, we were able to confirm the majority of these genes in other NSCLC cell lines using published data on gene expression profiles. Furthermore, we confirmed 10 genes at the RNA level in two neuroblastoma cell lines, indicating that these RASSF1A target genes have relevance in non-lung cell backgrounds. Protein analysis of six genes (ETS2, Cyclin D3, CDH2, DAPK1, TXN, and CTSL) showed that the changes induced by RASSF1A at the RNA level correlated with changes in protein expression in both non-small cell lung cancer and neuroblastoma cell lines. Finally, we have used a transient assay to demonstrate the induction of CDH2 and TGM2 by RASSF1A in NSCLC cell lines. We have identified several novel targets for RASSF1A tumor suppressor gene both at the RNA and the protein levels in two different cellular backgrounds. The identified targets are involved in diverse cellular processes; this should help toward understanding mechanisms that contribute to RASSF1A biological activity.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Neuroblastoma/genetics , Tumor Suppressor Proteins , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclin D1/biosynthesis , Cyclin D1/genetics , Down-Regulation , G1 Phase/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , Humans , Lung Neoplasms/metabolism , Neoplasm Proteins/physiology , Neuroblastoma/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Staurosporine/pharmacology , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...