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.
J Biol Chem ; 295(24): 8214-8226, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32350115

ABSTRACT

Epithelial cell-transforming sequence 2 (ECT2) is a guanine nucleotide exchange factor for Rho GTPases that is overexpressed in many cancers and involved in signal transduction pathways that promote cancer cell proliferation, invasion, and tumorigenesis. Recently, we demonstrated that a significant pool of ECT2 localizes to the nucleolus of non-small-cell lung cancer (NSCLC) cells, where it binds the transcription factor upstream binding factor 1 (UBF1) on the promoter regions of ribosomal DNA (rDNA) and activates rDNA transcription, transformed cell growth, and tumor formation. Here, we investigated the mechanism by which ECT2 engages UBF1 on rDNA promoters. Results from ECT2 mutagenesis indicated that the tandem BRCT domain of ECT2 mediates binding to UBF1. Biochemical and MS-based analyses revealed that protein kinase Cι (PKCι) directly phosphorylates UBF1 at Ser-412, thereby generating a phosphopeptide-binding epitope that binds the ECT2 BRCT domain. Lentiviral shRNA knockdown and reconstitution experiments revealed that both a functional ECT2 BRCT domain and the UBF1 Ser-412 phosphorylation site are required for UBF1-mediated ECT2 recruitment to rDNA, elevated rRNA synthesis, and transformed growth. Our findings provide critical molecular insight into ECT2-mediated regulation of rDNA transcription in cancer cells and offer a rationale for therapeutic targeting of UBF1- and ECT2-stimulated rDNA transcription for the management of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/metabolism , DNA, Ribosomal/metabolism , Isoenzymes/metabolism , Lung Neoplasms/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Ribosomal/metabolism , Amino Acid Motifs , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Humans , Lung Neoplasms/pathology , Models, Biological , Phosphopeptides/metabolism , Phosphorylation , Protein Binding , Protein Domains , Proto-Oncogene Proteins/chemistry
2.
Cancer Cell ; 31(2): 256-269, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28110998

ABSTRACT

The guanine nucleotide exchange factor (GEF) epithelial cell transforming sequence 2 (Ect2) has been implicated in cancer. However, it is not clear how Ect2 causes transformation and whether Ect2 is necessary for tumorigenesis in vivo. Here, we demonstrate that nuclear Ect2 GEF activity is required for Kras-Trp53 lung tumorigenesis in vivo and that Ect2-mediated transformation requires Ect2-dependent rDNA transcription. Ect2 activates rRNA synthesis by binding the nucleolar transcription factor upstream binding factor 1 (UBF1) on rDNA promoters and recruiting Rac1 and its downstream effector nucleophosmin (NPM) to rDNA. Protein kinase Cι (PKCι)-mediated Ect2 phosphorylation stimulates Ect2-dependent rDNA transcription. Thus, Ect2 regulates rRNA synthesis through a PKCι-Ect2-Rac1-NPM signaling axis that is required for lung tumorigenesis.


Subject(s)
Adenocarcinoma/etiology , Lung Neoplasms/etiology , Proto-Oncogene Proteins p21(ras)/physiology , Proto-Oncogene Proteins/physiology , RNA, Ribosomal/biosynthesis , Tumor Suppressor Protein p53/physiology , Adenocarcinoma of Lung , Animals , Auranofin/pharmacology , Cell Line, Tumor , Cell Nucleolus/metabolism , Cytokinesis , Humans , Isoenzymes/physiology , Mice , Nuclear Proteins/physiology , Nucleophosmin , Protein Kinase C/physiology , Signal Transduction/physiology , rac1 GTP-Binding Protein/physiology
3.
Cancer Cell ; 29(3): 367-378, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26977885

ABSTRACT

We report that the protein kinase Cι (PKCι) oncogene controls expression of NOTCH3, a key driver of stemness, in KRAS-mediated lung adenocarcinoma (LADC). PKCι activates NOTCH3 expression by phosphorylating the ELF3 transcription factor and driving ELF3 occupancy on the NOTCH3 promoter. PKCι-ELF3-NOTCH3 signaling controls the tumor-initiating cell phenotype by regulating asymmetric cell division, a process necessary for tumor initiation and maintenance. Primary LADC tumors exhibit PKCι-ELF3-NOTCH3 signaling, and combined pharmacologic blockade of PKCι and NOTCH synergistically inhibits tumorigenic behavior in vitro and LADC growth in vivo demonstrating the therapeutic potential of PKCι-ELF3-NOTCH3 signal inhibition to more effectively treat KRAS LADC.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Cell Transformation, Neoplastic/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Notch/metabolism , Stem Cells/metabolism , Adenocarcinoma of Lung , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/metabolism , Humans , Mutation/genetics , Neoplastic Stem Cells/metabolism , Phenotype , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-ets/metabolism , Receptor, Notch3 , Signal Transduction/genetics , Transcription Factors/metabolism
4.
Cancer Res ; 66(3): 1767-74, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452237

ABSTRACT

We recently showed that atypical protein kinase Ciota (PKCiota) is required for transformed growth of human non-small-cell lung cancer (NSCLC) cells by activating Rac1. Genetic disruption of PKCiota signaling blocks Rac1 activity and transformed growth, indicating that PKCiota is a viable target for development of novel therapeutics for NSCLC. Here, we designed and implemented a novel fluorescence resonance energy transfer-based assay to identify inhibitors of oncogenic PKCiota signaling. This assay was used to identify compounds that disrupt the interaction between PKCiota and its downstream effector Par6, which links PKCiota to Rac1. We identified aurothioglucose (ATG), a gold compound used clinically to treat rheumatoid arthritis, and the related compound, aurothiomalate (ATM), as potent inhibitors of PKCiota-Par6 interactions in vitro (IC(50) approximately 1 micromol/L). ATG blocks PKCiota-dependent signaling to Rac1 and inhibits transformed growth of NSCLC cells. ATG-mediated inhibition of transformation is relieved by expression of constitutively active Rac1, consistent with a mechanism at the level of the interaction between PKCiota and Par6. ATG inhibits A549 cell tumor growth in nude mice, showing efficacy against NSCLC in a relevant preclinical model. Our data show the utility of targeting protein-protein interactions involving PKCiota for antitumor drug development and provide proof of concept that chemical disruption of PKCiota signaling can be an effective treatment for NSCLC. ATG and ATM will be useful reagents for studying PKCiota function in transformation and represent promising new agents for the clinical treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Isoenzymes/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Aurothioglucose/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Fluorescence Resonance Energy Transfer , Gold Sodium Thiomalate/pharmacology , Humans , Isoenzymes/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Protein Kinase C/metabolism , Protein Structure, Tertiary , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/metabolism
5.
Cancer Res ; 65(19): 8905-11, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204062

ABSTRACT

Protein kinase C (PKC) isozymes have long been implicated in carcinogenesis. However, little is known about the functional significance of these enzymes in human cancer. We recently showed that the atypical PKC (aPKC) isozyme PKCiota is overexpressed in human non-small cell lung cancer (NSCLC) cells and that PKCiota plays a critical role in the transformed growth of the human lung adenocarcinoma A549 cell line in vitro and tumorigenicity in vivo. Here we provide compelling evidence that PKCiota is an oncogene in NSCLC based on the following criteria: (a) aPKCiota is overexpressed in the vast majority of primary NSCLC tumors; (b) tumor PKCiota expression levels predict poor survival in patients with NSCLC; (c) the PKCiota gene is frequently amplified in established NSCLC cell lines and primary NSCLC tumors; (d) gene amplification drives PKCiota expression in NSCLC cell lines and primary NSCLC tumors; and (e) disruption of PKCiota signaling with a dominant negative PKCiota allele blocks the transformed growth of human NSCLC cells harboring PKCiota gene amplification. Taken together, our data provide conclusive evidence that PKCiota is required for the transformed growth of NSCLC cells and that the PKCiota gene is a target for tumor-specific genetic alteration by amplification. Interestingly, PKCiota expression predicts poor survival in NSCLC patients independent of tumor stage. Therefore, PKCiota expression profiling may be useful in identifying early-stage NSCLC patients at elevated risk of relapse. Our functional data indicate that PKCiota is an attractive target for development of novel, mechanism-based therapeutics to treat NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Isoenzymes/genetics , Lung Neoplasms/genetics , Oncogenes , Protein Kinase C/genetics , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Amplification , Humans , Isoenzymes/biosynthesis , Lung Neoplasms/enzymology , Male , Middle Aged , Protein Kinase C/biosynthesis
6.
J Biol Chem ; 280(35): 31109-15, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-15994303

ABSTRACT

Atypical protein kinase C (aPKC) isozymes function in epithelial cell polarity, proliferation, and survival and have been implicated in cellular transformation. However, the role of these enzymes in human cancer is largely unexplored. Here, we report that aPKCiota is highly expressed in human non-small cell lung cancer cell lines, whereas the closely related aPKC isozyme PKCzeta is undetectable in these cells. Disruption of PKCiota signaling reveals that PKCiota is dispensable for adherent growth of non-small cell lung cancer cells but is required for transformed growth in soft agar in vitro and for tumorigenicity in vivo. Molecular dissection of signaling down-stream of PKCiota demonstrates that Rac1 is a critical molecular target for PKCiota-dependent transformation, whereas PKCiota is not necessary for NFkappaB activation in vitro or in vivo. Expression of the PB1 domain of PKCiota (PKCiota-(1-113)) blocks PKCiota-dependent Rac1 activity and inhibits cellular transformation indicating a role for this domain in the transforming activity of PKCiota. Taken together, our data demonstrate that PKCiota is a critical lung cancer gene that activates a Rac1-->Pak-->Mek1,2-->Erk1,2 signaling pathway required for transformed growth. Our data indicate that PKCiota may be an attractive molecular target for mechanism-based therapies for treatment of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Transformation, Neoplastic , Isoenzymes/metabolism , Lung Neoplasms , Protein Kinase C/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Humans , Isoenzymes/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Transplantation , Protein Kinase C/genetics , Signal Transduction/physiology , rac1 GTP-Binding Protein/metabolism
7.
J Cell Biol ; 164(6): 797-802, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15024028

ABSTRACT

Protein kinase C iota (PKCiota) has been implicated in Ras signaling, however, a role for PKCiota in oncogenic Ras-mediated transformation has not been established. Here, we show that PKCiota is a critical downstream effector of oncogenic Ras in the colonic epithelium. Transgenic mice expressing constitutively active PKCiota in the colon are highly susceptible to carcinogen-induced colon carcinogenesis, whereas mice expressing kinase-deficient PKCiota (kdPKCiota) are resistant to both carcinogen- and oncogenic Ras-mediated carcinogenesis. Expression of kdPKCiota in Ras-transformed rat intestinal epithelial cells blocks oncogenic Ras-mediated activation of Rac1, cellular invasion, and anchorage-independent growth. Constitutively active Rac1 (RacV12) restores invasiveness and anchorage-independent growth in Ras-transformed rat intestinal epithelial cells expressing kdPKCiota. Our data demonstrate that PKCiota is required for oncogenic Ras- and carcinogen-mediated colon carcinogenesis in vivo and define a procarcinogenic signaling axis consisting of Ras, PKCiota, and Rac1.


Subject(s)
Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Azoxymethane/pharmacology , Carcinogens/pharmacology , Colonic Neoplasms/pathology , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/enzymology , Epithelial Cells/physiology , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase C/genetics , Rats , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , ras Proteins/genetics
8.
J Biol Chem ; 279(10): 9400-8, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14670960

ABSTRACT

The protein kinase C (PKC) family of serine/threonine kinases plays an important role in numerous cancer signaling pathways, including those downstream of the bcr-abl oncogene. We demonstrated previously that atypical PKCiota is required for Bcr-Abl-mediated resistance of human K562 chronic myelogenous leukemia (CML) cells to Taxol-induced apoptosis. Here, we report that the pattern of PKC isozyme expression characteristic of CML cells is regulated by Bcr-Abl. When Bcr-Abl was expressed in Bcr-Abl-negative HL-60 promyelocytic leukemia cells, expression of the PKCbetaI, PKCbetaII, and PKCiota genes was induced, whereas expression of the PKCdelta gene was reduced to levels similar to those found in CML cells. Given the importance of PKCiota in Bcr-Abl-mediated transformation, we characterized the mechanism by which Bcr-Abl regulates PKCiota expression. A 1200-bp PKCiota promoter construct isolated from genomic DNA was highly active in Bcr-Abl-positive K562 cells and was activated when Bcr-Abl-negative cells were transfected with Bcr-Abl. Bcr-Abl-mediated induction of the PKCiota promoter was dependent upon MEK1/2 activity, but not phosphatidylinositol 3-kinase or p38 MAPK activity. Mutational analysis of the PKCiota promoter revealed a region between 97 and 114 bp upstream of the transcriptional start site that is responsible for Bcr-Abl-mediated regulation. Mutation of a consensus Elk1-binding site within this region abolished Bcr-Abl-mediated regulation. We conclude that Bcr-Abl regulates PKCiota expression through the MEK-dependent activation of an Elk1 element within the proximal PKCiota promoter. Our results indicate that Bcr-Abl-mediated transformation involves transcriptional activation of the PKCiota gene, which in turn is required for Bcr-Abl-mediated chemoresistance.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation, Enzymologic , Genes, abl/genetics , Isoenzymes/genetics , Protein Kinase C/genetics , Transcription Factors , Cell Transformation, Neoplastic/genetics , HL-60 Cells , Humans , K562 Cells , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Transcription, Genetic , ets-Domain Protein Elk-1
9.
J Cell Biol ; 157(6): 915-20, 2002 Jun 10.
Article in English | MEDLINE | ID: mdl-12058013

ABSTRACT

Increasing evidence demonstrates that protein kinase C betaII (PKCbetaII) promotes colon carcinogenesis. We previously reported that colonic PKCbetaII is induced during colon carcinogenesis in rodents and humans, and that elevated expression of PKCbetaII in the colon of transgenic mice enhances colon carcinogenesis. Here, we demonstrate that PKCbetaII represses transforming growth factor beta receptor type II (TGFbetaRII) expression and reduces sensitivity to TGF-beta-mediated growth inhibition in intestinal epithelial cells. Transgenic PKCbetaII mice exhibit hyperproliferation, enhanced colon carcinogenesis, and marked repression of TGFbetaRII expression. Chemopreventive dietary omega-3 fatty acids inhibit colonic PKCbetaII activity in vivo and block PKCbetaII-mediated hyperproliferation, enhanced carcinogenesis, and repression of TGFbetaRII expression in the colonic epithelium of transgenic PKCbetaII mice. These data indicate that dietary omega-3 fatty acids prevent colon cancer, at least in part, through inhibition of colonic PKCbetaII signaling and restoration of TGF-beta responsiveness.


Subject(s)
Colonic Neoplasms/etiology , Fatty Acids, Omega-3/metabolism , Isoenzymes/physiology , Protein Kinase C/physiology , Receptors, Transforming Growth Factor beta/physiology , Animals , Cell Division , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Intestinal Mucosa/cytology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Transgenic , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C beta , Rats , Rats, Sprague-Dawley , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...