Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters











Publication year range
1.
Oncogene ; 36(4): 534-545, 2017 01 26.
Article in English | MEDLINE | ID: mdl-27321186

ABSTRACT

Atypical protein kinase Cι (PKCι) is an oncogene in lung and ovarian cancer. The PKCι gene PRKCI is targeted for frequent tumor-specific copy number gain (CNG) in both lung squamous cell carcinoma (LSCC) and ovarian serous carcinoma (OSC). We recently demonstrated that in LSCC cells PRKCI CNG functions to drive transformed growth and tumorigenicity by activating PKCι-dependent cell autonomous Hedgehog (Hh) signaling. Here, we assessed whether OSC cells harboring PRKCI CNG exhibit similar PKCι-dependent Hh signaling. Surprisingly, we find that whereas PKCι is required for the transformed growth of OSC cells harboring PRKCI CNG, these cells do not exhibit PKCι-dependent Hh signaling or Hh-dependent proliferation. Rather, transformed growth of OSC cells is regulated by PKCι-dependent nuclear localization of the oncogenic transcription factor, YAP1. Lentiviral shRNA-mediated knockdown (KD) of PKCι leads to decreased nuclear YAP1 and increased YAP1 binding to angiomotin (AMOT), which sequesters YAP1 in the cytoplasm. Biochemical analysis reveals that PKCι directly phosphorylates AMOT at a unique site, Thr750, whose phosphorylation inhibits YAP1 binding. Pharmacologic inhibition of PKCι decreases YAP1 nuclear localization and blocks OSC tumor growth in vitro and in vivo. Immunohistochemical analysis reveals a strong positive correlation between tumor PKCι expression and nuclear YAP1 in primary OSC tumor samples, indicating the clinical relevance of PKCι-YAP1 signaling. Our results uncover a novel PKCι-AMOT-YAP1 signaling axis that promotes OSC tumor growth, and provide a rationale for therapeutic targeting of this pathway for treatment of OSC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/metabolism , Isoenzymes/metabolism , Ovarian Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Adaptor Proteins, Signal Transducing/genetics , Angiomotins , Animals , Carcinogenesis/pathology , Cell Cycle Proteins , Cell Line, Tumor , Female , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Isoenzymes/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Microfilament Proteins , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphoproteins/genetics , Protein Kinase C/genetics , Signal Transduction , Transfection , YAP-Signaling Proteins
2.
Oncogene ; 33(16): 2134-44, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-23604119

ABSTRACT

Protein kinase C alpha (PKCα) can activate both pro- and anti-tumorigenic signaling depending upon cellular context. Here, we investigated the role of PKCα in lung tumorigenesis in vivo. Gene expression data sets revealed that primary human non-small lung cancers (NSCLC) express significantly decreased PKCα levels, indicating that loss of PKCα expression is a recurrent event in NSCLC. We evaluated the functional relevance of PKCα loss during lung tumorigenesis in three murine lung adenocarcinoma models (LSL-Kras, LA2-Kras and urethane exposure). Genetic deletion of PKCα resulted in a significant increase in lung tumor number, size, burden and grade, bypass of oncogene-induced senescence, progression from adenoma to carcinoma and a significant decrease in survival in vivo. The tumor promoting effect of PKCα loss was reflected in enhanced Kras-mediated expansion of bronchio-alveolar stem cells (BASCs), putative tumor-initiating cells, both in vitro and in vivo. LSL-Kras/Prkca(-/-) mice exhibited a decrease in phospho-p38 MAPK in BASCs in vitro and in tumors in vivo, and treatment of LSL-Kras BASCs with a p38 inhibitor resulted in increased colony size indistinguishable from that observed in LSL-Kras/Prkca(-/-) BASCs. In addition, LSL-Kras/Prkca(-/-) BASCs exhibited a modest but reproducible increase in TGFß1 mRNA, and addition of exogenous TGFß1 to LSL-Kras BASCs results in enhanced growth similar to untreated BASCs from LSL-Kras/Prkca(-/-) mice. Conversely, a TGFßR1 inhibitor reversed the effects of PKCα loss in LSL-Kras/Prkca(-/-) BASCs. Finally, we identified the inhibitors of DNA binding (Id) Id1-3 and the Wilm's Tumor 1 as potential downstream targets of PKCα-dependent tumor suppressor activity in vitro and in vivo. We conclude that PKCα suppresses tumor initiation and progression, at least in part, through a PKCα-p38MAPK-TGFß signaling axis that regulates tumor cell proliferation and Kras-induced senescence. Our results provide the first direct evidence that PKCα exhibits tumor suppressor activity in the lung in vivo.


Subject(s)
Lung Neoplasms/genetics , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Bronchioles/metabolism , Bronchioles/pathology , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology , Transforming Growth Factor beta/metabolism , WT1 Proteins/genetics , WT1 Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Oncogene ; 32(3): 286-95, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-22349825

ABSTRACT

Alveolar rhabdomyosarcoma is an aggressive pediatric cancer exhibiting skeletal-muscle differentiation. New therapeutic targets are required to improve the dismal prognosis for invasive or metastatic alveolar rhabdomyosarcoma. Protein kinase C iota (PKCι) has been shown to have an important role in tumorigenesis of many cancers, but little is known about its role in rhabdomyosarcoma. Our gene-expression studies in human tumor samples revealed overexpression of PRKCI. We confirmed overexpression of PKCι at the mRNA and protein levels using our conditional mouse model that authentically recapitulates the progression of rhabdomyosarcoma in humans. Inhibition of Prkci by RNA interference resulted in a dramatic decrease in anchorage-independent colony formation. Interestingly, treatment of primary cell cultures using aurothiomalate (ATM), which is a gold-containing classical anti-rheumatic agent and a PKCι-specific inhibitor, resulted in decreased interaction between PKCι and Par6, decreased Rac1 activity and reduced cell viability at clinically relevant concentrations. Moreover, co-treatment with ATM and vincristine (VCR), a microtubule inhibitor currently used in rhabdomyosarcoma treatment regimens, resulted in a combination index of 0.470-0.793 through cooperative accumulation of non-proliferative multinuclear cells in the G2/M phase, indicating that these two drugs synergize. For in vivo tumor growth inhibition studies, ATM demonstrated a trend toward enhanced VCR sensitivity. Overall, these results suggest that PKCι is functionally important in alveolar rhabdomyosarcoma anchorage-independent growth and tumor-cell proliferation and that combination therapy with ATM and microtubule inhibitors holds promise for the treatment of alveolar rhabdomyosarcoma.


Subject(s)
Isoenzymes/metabolism , Molecular Targeted Therapy/methods , Protein Kinase C/metabolism , Rhabdomyosarcoma, Alveolar/drug therapy , Rhabdomyosarcoma, Alveolar/enzymology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant , Drug Synergism , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gold Sodium Thiomalate/pharmacology , Gold Sodium Thiomalate/therapeutic use , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Mice , Protein Kinase C/deficiency , Protein Kinase C/genetics , RNA Interference , RNA, Small Interfering/genetics , Rhabdomyosarcoma, Alveolar/metabolism , Rhabdomyosarcoma, Alveolar/pathology , Vincristine/pharmacology , Vincristine/therapeutic use
4.
Oncogene ; 28(41): 3597-607, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19617897

ABSTRACT

Protein kinase Ciota (PKCiota) promotes non-small cell lung cancer (NSCLC) by binding to Par6alpha and activating a Rac1-Pak-Mek1,2-Erk1,2 signaling cascade. The mechanism by which the PKCiota-Par6alpha complex regulates Rac1 is unknown. Here we show that epithelial cell transforming sequence 2 (Ect2), a guanine nucleotide exchange factor for Rho family GTPases, is coordinately amplified and overexpressed with PKCiota in NSCLC tumors. RNA interference-mediated knockdown of Ect2 inhibits Rac1 activity and blocks transformed growth, invasion and tumorigenicity of NSCLC cells. Expression of constitutively active Rac1 (RacV12) restores transformation to Ect2-deficient cells. Interestingly, the role of Ect2 in transformation is distinct from its well-established role in cytokinesis. In NSCLC cells, Ect2 is mislocalized to the cytoplasm where it binds the PKCiota-Par6alpha complex. RNA interference-mediated knockdown of either PKCiota or Par6alpha causes Ect2 to redistribute to the nucleus, indicating that the PKCiota-Par6alpha complex regulates the cytoplasmic localization of Ect2. Our data indicate that Ect2 and PKCiota are genetically and functionally linked in NSCLC, acting to coordinately drive tumor cell proliferation and invasion through formation of an oncogenic PKCiota-Par6alpha-Ect2 complex.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cytokinesis/genetics , Cytoplasm/metabolism , Enzyme Activation , Gene Amplification , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Protein Transport , Proto-Oncogene Proteins/deficiency
5.
Oncogene ; 27(35): 4841-53, 2008 Aug 14.
Article in English | MEDLINE | ID: mdl-18427549

ABSTRACT

Protein kinase Ciota (PKCiota) drives transformed growth of non-small cell lung cancer (NSCLC) cells through the Rho family GTPase Rac1. We show here that PKCiota activates Rac1 in NSCLC cells by formation of a PKCiota-Par6alpha complex that drives anchorage-independent growth and invasion through activation of matrix metalloproteinase-10 (MMP-10) expression. RNAi-mediated knockdown of PKCiota, Par6alpha or Rac1 expression inhibits NSCLC transformation and MMP-10 expression in vitro. Expression of wild-type Par6alpha in Par6alpha-deficient cells restores transformation and MMP-10 expression, whereas expression of Par6alpha mutants that either cannot bind PKCiota (Par6alpha-K19A) or couple to Rac1 (Par6alpha-DeltaCRIB) do not. Knockdown of MMP-10 expression blocks anchorage-independent growth and invasion of NSCLC cells and addition of catalytically active MMP-10 to PKCiota- or Par6alpha-deficient cells restores anchorage-independent growth and invasion. Dominant-negative PKCiota inhibits tumorigenicity and MMP-10 expression in subcutaneous NSCLC tumors. MMP-10 and PKCiota are coordinately overexpressed in primary NSCLC tumors, and tumor MMP-10 expression predicts poor survival in NSCLC patients. Our data define a PKCiota-Par6alpha-Rac1 signaling axis that drives anchorage-independent growth and invasion of NSCLC cells through induction of MMP-10 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carcinoma, Non-Small-Cell Lung/pathology , Isoenzymes/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinase 10/metabolism , Protein Kinase C/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Division , Cell Line, Tumor , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Protein Binding , RNA Interference
6.
Biochem Soc Trans ; 35(Pt 5): 996-1000, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17956262

ABSTRACT

PKC (protein kinase C) isoenzymes are key signalling components involved in the regulation of normal cell proliferation, differentiation, polarity and survival. The aberrant regulation of PKC isoenzymes has been implicated in the development of many human diseases including cancer [Fields and Gustafson (2003) Methods Mol. Biol. 233, 519-537]. To date, however, only one PKC isoenzyme, the aPKC [atypical PKCiota (protein kinase Ciota)], has been identified as a human oncogene [Regala, Weems, Jamieson, Khoor, Edell, Lohse and Fields (2005) Cancer Res. 65, 8905-8911]. PKCiota has also proven to be a useful prognostic marker and legitimate target for the development of novel pharmacological agents for the treatment of cancer. The PKCiota gene resides at chromosome 3q26 and is a frequent target of tumour-specific gene amplification in multiple forms of human cancer. PKCiota gene amplification in turn drives PKCiota overexpression in these cancers. Genetic disruption of PKCiota expression blocks multiple aspects of the transformed phenotype of human cancer cells including transformed growth in soft agar, invasion through Matrigel and growth of subcutaneous tumours in nude mice. Genetic dissection of oncogenic PKCiota signalling mechanisms demonstrates that PKCiota drives transformed growth by activating a PKCiota --> Rac1 --> PAK (p21-activated kinase) --> MEK [MAPK (mitogen-activated protein kinase) 1,2/ERK (extracellular-signal-regulated kinase) kinase] 1,2 signalling pathway [Regala, Weems, Jamieson, Copland, Thompson and Fields (2005) J. Biol. Chem. 280, 31109-31115]. The transforming activity of PKCiota requires the N-terminal PB1 (Phox-Bem1) domain of PKCiota, which serves to couple PKCiota with downstream effector molecules. Hence, there exists a strong rationale for developing novel cancer therapeutics that target the PB1 domain of PKCiota and thereby disrupt its interactions with effector molecules. Using a novel high-throughput drug screen, we identified compounds that can disrupt PB1-PB1 domain interactions between PKCiota and the adaptor molecule Par6 [Stallings-Mann, Jamieson, Regala, Weems, Murray and Fields (2006) Cancer Res. 66, 1767-1774]. Our screen identified the gold compounds ATG (aurothioglucose) and ATM (aurothiomalate) as specific inhibitors of the PB1-PB1 domain interaction between PKCiota and Par6 that exhibit anti-tumour activity against NSCLC (non-small-cell lung cancer) both in vitro and in vivo. Structural analysis, site-directed mutagenesis and modelling indicate that ATM specifically targets the PB1 domain of PKCiota to mediate its anti-tumour activity [Erdogan, Lamark, Stallings-Mann, Lee, Pellechia, Thompson, Johansen and Fields (2006) J. Biol. Chem. 281, 28450-28459]. Taken together, our recent work demonstrates that PKCiota signalling is required for transformed growth of human tumours and is an attractive target for development of mechanism-based cancer therapies. ATM is currently in Phase I clinical trials for the treatment of NSCLC.


Subject(s)
Isoenzymes/metabolism , Neoplasms/drug therapy , Oncogenes , Protein Kinase C/metabolism , Signal Transduction , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Prognosis , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry
7.
Oncogene ; 25(25): 3565-75, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16462766

ABSTRACT

In hematopoietic cells the transforming potential of the ecotropic viral integration site 1 (Evi1) oncogene is thought to be dependent upon the ability to inhibit TGFbeta signaling. Although Evi1 has recently been implicated in certain epithelial cancers, the effects of Evi1 on transformation and TGFbeta signaling in epithelial cells are not completely understood. Herein, we have determined the effects of Evi1 on TGFbeta signaling in intestinal epithelial cells. Stable expression of Evi1 in non-transformed intestinal epithelial cells inhibited induction of some Smad3-dependent TGFbeta target genes, such as PAI1. However, TGFbeta-mediated induction of cellular adhesion signaling components such as integrin1 and paxillin was not inhibited by Evi1; nor did Evi1 inhibit TGFbeta-mediated epithelial to mesenchymal transition. Likewise, Evi1 did not inhibit TGFbeta-mediated downregulation of cyclin D1 or block TGFbeta-mediated growth inhibition. However, Evi1 did inhibit TGFbeta-mediated apoptosis by a process that involves phosphoinositide-3-kinase (PI3K) and its downstream effector AKT. The ability of Evi1 to suppress apoptosis is not restricted to TGFbeta-mediated cell death, since Evi1 also protects intestinal epithelial cells from taxol-mediated apoptosis. Evi1 is overexpressed in some human colon cancer cell lines, and overexpression is associated with amplification of the Evi1 gene. Knockdown of Evi1 by siRNA inhibited AKT phosphorylation in HT-29 human colon cancer cells and increased their sensitivity to taxol-mediated apoptosis. These data indicate that Evi1 functions as a survival gene in intestinal epithelial cells and colon cancer cells, activating PI3K/AKT and conveying resistance to both physiological and therapeutic apoptotic stimuli.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/physiology , Oncogene Protein v-akt/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogenes/physiology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Movement/physiology , Colonic Neoplasms/metabolism , Humans , Intestinal Mucosa/metabolism , Rats , Signal Transduction/physiology , Transcriptional Activation
8.
Oncogene ; 20(35): 4777-92, 2001 Aug 09.
Article in English | MEDLINE | ID: mdl-11521190

ABSTRACT

In chronic myelogenous leukemia (CML), the oncogene bcr-abl encodes a dysregulated tyrosine kinase that inhibits apoptosis. We showed previously that human erythroleukemia K562 cells are resistant to antineoplastic drug (taxol)-induced apoptosis through the atypical protein kinase C iota isozyme (PKC iota), a kinase downstream of Bcr-Abl. The mechanism(s) by which PKC iota mediates cell survival to taxol is unknown. Here we demonstrate that PKC iota requires the transcription factor nuclear factor-kappaB (NF-kappaB) to confer cell survival. At apoptosis-inducing concentrations, taxol weakly induces IkappaB(alpha) proteolysis and NF-kappaB translocation in K562 cells, but potently induces its transcriptional activity. Inhibition of NF-kappaB activity (by blocking IkappaB(alpha) degradation) significantly sensitizes cells to taxol-induced apoptosis. Likewise, K562 cells expressing antisense PKC iota mRNA or kinase dead PKC iota (PKC iota-KD) are sensitized to taxol; these cells are rescued from apoptosis by NF-kappaB overexpression. Expression of constitutively active PKC iota (PKC iota-CA) upregulates NF-kappaB transactivation and rescues cells from apoptosis in the absence of Bcr-Abl tyrosine kinase activity. Using a chimeric GAL4-RelA transactivator, we find that taxol potently activates GAL4-RelA-dependent transcription. This activation was further upregulated by expression of PKC iota-CA and inhibited by expression of PKC iota-KD. Our results indicate that RelA transactivation is an important downstream target of the PKC iota-mediated Bcr-Abl signaling pathway and is required for resistance to taxol-induced apoptosis.


Subject(s)
I-kappa B Proteins , Isoenzymes/physiology , NF-kappa B/physiology , Protein Kinase C/physiology , Transcriptional Activation , Cell Survival , DNA/metabolism , DNA-Binding Proteins/metabolism , Fusion Proteins, bcr-abl/physiology , Humans , I-kappa B Kinase , K562 Cells , NF-KappaB Inhibitor alpha , Paclitaxel/pharmacology , Protein Serine-Threonine Kinases/physiology , Transcription Factor RelA
9.
J Biol Chem ; 276(25): 22709-14, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11331269

ABSTRACT

We have cloned and characterized a new member of the phosphatidylinositol kinase (PIK)-related kinase family. This gene, which we term human SMG-1 (hSMG-1), is orthologous to Caenorhabditis elegans SMG-1, a protein that functions in nonsense-mediated mRNA decay (NMD). cDNA sequencing revealed that hSMG-1 encodes a protein of 3031 amino acids containing a conserved kinase domain, a C-terminal domain unique to the PIK-related kinases and an FKBP12-rapamycin binding-like domain similar to that found in the PIK-related kinase mTOR. Immunopurified FLAG-tagged hSMG-1 exhibits protein kinase activity as measured by autophosphorylation and phosphorylation of the generic PIK-related kinase substrate PHAS-1. hSMG-1 kinase activity is inhibited by high nanomolar concentrations of wortmannin (IC(50) = 105 nm) but is not inhibited by a FKBP12-rapamycin complex. Mutation of conserved residues within the kinase domain of hSMG-1 abolishes both autophosphorylation and substrate phosphorylation, demonstrating that hSMG-1 exhibits intrinsic protein kinase activity. hSMG-1 phosphorylates purified hUpf1 protein, a phosphoprotein that plays a critical role in NMD, at sites that are also phosphorylated in whole cells. Based on these data, we conclude that hSMG-1 is the human orthologue to C. elegans SMG-1. Our data indicate that hSMG-1 may function in NMD by directly phosphorylating hUpf1 protein at physiologically relevant sites.


Subject(s)
Protein Kinases/genetics , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , DNA, Complementary , Humans , Metalloendopeptidases , Molecular Sequence Data , Protein Kinases/chemistry , Protein Kinases/metabolism , Sequence Homology, Amino Acid
10.
Cancer Res ; 61(4): 1375-81, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245437

ABSTRACT

Protein kinase C (PKC) has been implicated in colon carcinogenesis in humans and in rodent models. However, little is known about the specific role of individual PKC isozymes in this process. We recently demonstrated that elevated expression of PKC betaII in the colonic epithelium induces hyperproliferation in vivo (N. R. Murray et al., J. Cell Biol., 145: 699-711, 1999). Because hyperproliferation is a major risk factor for colon cancer, we assessed whether specific alterations in PKC betaII expression occur during azoxymethane-induced colon carcinogenesis in mice. An increase in PKC betaII expression was observed in preneoplastic lesions (aberrant crypt foci, 3.7-fold) compared with saline-treated animals, and in colon tumors (7.8-fold; P = 0.011) compared with uninvolved colonic epithelium. In contrast, PKC alpha and PKC betaI (a splicing variant of PKC betaII) expression was slightly decreased in aberrant crypt foci and dramatically reduced in colon tumors. Quantitative reverse transcription-PCR analysis revealed that PKC mRNA levels do not directly correlate with PKC protein levels, indicating that PKC isozyme expression is likely regulated at the posttranscriptional/translational level. Finally, transgenic mice expressing elevated PKC betaII in the colonic epithelium exhibit a trend toward increased colon tumor formation after exposure to azoxymethane. Taken together, our results demonstrate that elevated expression of PKC betaII is an important early, promotive event that plays a role in colon cancer development.


Subject(s)
Colonic Neoplasms/enzymology , Isoenzymes/biosynthesis , Precancerous Conditions/enzymology , Protein Kinase C/biosynthesis , Animals , Azoxymethane/toxicity , Carcinogens/toxicity , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Female , Genetic Predisposition to Disease , Immunohistochemistry , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precancerous Conditions/chemically induced , Precancerous Conditions/genetics , Protein Kinase C/genetics , Protein Kinase C beta , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
J Biol Chem ; 276(21): 17754-61, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11278304

ABSTRACT

Highly purified liver nuclei incorporated radiolabeled phosphate into phosphatidylinositol 4-phosphate (PtdIns(4)P), PtdIns(4,5)P(2), and PtdIns(3,4,5)P(3). When nuclei were depleted of their membrane, no radiolabeling of PtdIns(3,4,5)P(3) could be detected showing that within the intranuclear region there are no class I phosphoinositide 3-kinases (PI3K)s. In membrane-depleted nuclei harvested 20 h after partial hepatectomy, the incorporation of radiolabel into PtdIns(3)P was observed together with an increase in immunoprecipitable PI3K-C2beta activity, which is sensitive to wortmannin (10 nm) and shows strong preference for PtdIns over PtdIns(4)P as a substrate. On Western blots PI3K-C2beta revealed a single immunoreactive band of 180 kDa, whereas 20 h after partial hepatectomy gel shift of 18 kDa was noticed, suggesting that observed activation of enzyme is achieved by proteolysis. When intact membrane-depleted nuclei were subjected to short term (20 min) exposure to micro-calpain, similar gel shift together with an increase in PI3K-C2beta activity was observed, when compared with the nuclei harvested 20 h after partial hepatectomy. Moreover, the above-mentioned gel shift and increase in PI3K-C2beta activity could be prevented by the calpain inhibitor calpeptin. The data presented in this report show that, in the membrane-depleted nuclei during the compensatory liver growth, there is an increase in PtdIns(3)P formation as a result of PI3K-C2beta activation, which may be a calpain-mediated event.


Subject(s)
Liver Regeneration , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Nucleus/enzymology , Enzyme Activation , Male , Rats , Rats, Wistar , Signal Transduction
12.
J Biol Chem ; 275(50): 39727-33, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10995778

ABSTRACT

The transcription factor serum amyloid A (SAA)-activating factor (SAF), a family of zinc finger proteins, plays a significant role in the induced expression of the SAA gene. Activity of SAF is regulated by a phosphorylation event involving serine/threonine protein kinase (Ray, A., Schatten, H., and Ray, B. K. (1999) J. Biol. Chem. 274, 4300-4308; Ray, A., and Ray, B. K. (1998) Mol. Cell. Biol. 18, 7327-7335). However, the identity of the protein kinase has so far remained unknown. Induction of SAA by phorbol 12-myristate 13-acetate, a known agonist of protein kinase C (PKC), suggested a potential role of the PKC signaling pathway in the activation process. The DNA binding activity of endogenous SAF was increased by agonists of PKC. In vitro phosphorylation of SAF-1 by PKC-beta markedly increased its DNA binding ability. Consistent with these findings, treatment of cells with activators of PKC or overexpression of PKC-betaII in transfected cells increased expression of an SAF-regulated promoter. Further analysis with a GAL4 reporter system indicated that PKC-mediated phosphorylation mostly increases the DNA binding activity of SAF-1. Together these data indicated that the PKC signaling pathway plays a major role in controlling expression of SAF-regulated genes by increasing the interaction between promoter DNA and phosphorylated SAF.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Protein Kinase C/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Chloramphenicol O-Acetyltransferase/metabolism , DNA-Binding Proteins/genetics , Genes, Reporter , Isoenzymes/metabolism , Phosphorylation , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Kinase C beta , Rabbits , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors , Transfection , Zinc Fingers
13.
Am J Physiol Lung Cell Mol Physiol ; 279(3): L429-38, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10956616

ABSTRACT

Individual protein kinase C (PKC) isozymes have been implicated in many cellular responses important in lung health and disease, including permeability, contraction, migration, hypertrophy, proliferation, apoptosis, and secretion. New ideas on mechanisms that regulate PKC activity, including the identification of a novel PKC kinase, 3-phosphoinositide-dependent kinase-1 (PDK-1), that regulates phosphorylation of PKC, have been advanced. The importance of targeted translocation of PKC and isozyme-specific binding proteins (like receptors for activated C-kinase and caveolins) is well established. Phosphorylation state and localization are now thought to be key determinants of isozyme activity and specificity. New concepts on the role of individual PKC isozymes in proliferation and apoptosis are emerging. Opposing roles for selected isozymes in the same cell system have been defined. Coupling to the Wnt signaling pathway has been described. Phenotypes for PKC knockout mice have recently been reported. More specific approaches for studying PKC isozymes and their role in cell responses have been developed. Strengths and weaknesses of different experimental strategies are reviewed. Future directions for investigation are identified.


Subject(s)
Isoenzymes/physiology , Lung/physiology , Protein Kinase C/physiology , Animals , Apoptosis/physiology , Cell Division/physiology , Humans , Ischemia/pathology , Ischemia/physiopathology , Lung/cytology , Pulmonary Circulation
14.
J Cell Biol ; 145(4): 699-711, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10330400

ABSTRACT

Protein kinase C betaII (PKC betaII) has been implicated in proliferation of the intestinal epithelium. To investigate PKC betaII function in vivo, we generated transgenic mice that overexpress PKC betaII in the intestinal epithelium. Transgenic PKC betaII mice exhibit hyperproliferation of the colonic epithelium and an increased susceptibility to azoxymethane-induced aberrant crypt foci, preneoplastic lesions in the colon. Furthermore, transgenic PKC betaII mice exhibit elevated colonic beta-catenin levels and decreased glycogen synthase kinase 3beta activity, indicating that PKC betaII stimulates the Wnt/adenomatous polyposis coli (APC)/beta-catenin proliferative signaling pathway in vivo. These data demonstrate a direct role for PKC betaII in colonic epithelial cell proliferation and colon carcinogenesis, possibly through activation of the APC/beta-catenin signaling pathway.


Subject(s)
Colon/pathology , Colonic Neoplasms/etiology , Isoenzymes/physiology , Protein Kinase C/physiology , Trans-Activators , Animals , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Gene Expression , Intestinal Mucosa/cytology , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C beta , Signal Transduction , beta Catenin
15.
J Biol Chem ; 274(7): 3927-30, 1999 Feb 12.
Article in English | MEDLINE | ID: mdl-9933579

ABSTRACT

K562 chronic myelogenous leukemia cells are highly resistant to chemotherapeutic drugs, such as taxol, that induce cell death by apoptosis. This resistance is mediated by the chimeric tyrosine kinase oncogene Bcr-Abl. However, little is known about the mechanism by which Bcr-Abl protects K562 cells from apoptosis. We recently demonstrated that expression of PKCiota is necessary for the resistance of K562 cells to taxol-induced apoptosis (Murray, N. R., and Fields, A. P. (1997) J. Biol. Chem. 272, 27521-27524). We now demonstrate that treatment of K562 cells with taxol leads to sustained activation of PKCiota. In contrast, Bcr-Abl-negative HL60 myeloid leukemia cells, which are sensitive to taxol-induced apoptosis, do not exhibit sustained PKCiota activation in response to taxol. Treatment of K562 cells with tyrphostin AG957, a selective Bcr-Abl inhibitor, blocks taxol-induced PKCiota activation and sensitizes these cells to taxol-induced apoptosis, indicating that PKCiota is a relevant downstream target of Bcr-Abl-mediated resistance. Furthermore, expression of constitutively active PKCiota by adenovirus-mediated gene transfer rescues AG957-treated K562 cells from taxol-induced apoptosis. Taken together, these results demonstrate that both Bcr-Abl and PKCiota activity are necessary for apoptotic resistance in K562 cells. Furthermore, they identify PKCiota as a critical downstream target of Bcr-Abl that is sufficient to mediate the anti-apoptotic effects of Bcr-Abl.


Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , 3T3 Cells , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Mice , Paclitaxel/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Tyrphostins/pharmacology
16.
J Biol Chem ; 274(6): 3764-71, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9920929

ABSTRACT

Our previous studies have shown that human native low density lipoprotein (LDL) can be oxidized by activated human monocytes. In this process, both activation of protein kinase C (PKC) and induction of superoxide anion (O-2) production are required. PKC is a family of isoenzymes, and the functional roles of individual PKC isoenzymes are believed to differ based on subcellular location and distinct responses to regulatory signals. We have shown that the PKC isoenzyme that is required for both monocyte O-2 production and oxidation of LDL is a member of the conventional PKC group of PKC isoenzymes (Li, Q., and Cathcart, M. K. (1994) J. Biol. Chem. 269, 17508-17515). The conventional PKC group includes PKCalpha, PKCbetaI, PKCbetaII, and PKCgamma. With the exception of PKCgamma, each of these isoenzymes was detected in human monocytes. In these studies, we investigated the requirement for select PKC isoenzymes in the process of monocyte-mediated LDL lipid oxidation. Our data indicate that PKC activity was rapidly induced upon monocyte activation with the majority of the activity residing in the membrane/particulate fraction. This enhanced PKC activity was sustained for up to 24 h after activation. PKCalpha, PKCbetaI, and PKCbetaII protein levels were induced upon monocyte activation, and PKCalpha and PKCbetaII substantially shifted their location from the cytosol to the particulate/membrane fraction. To distinguish between these isoenzymes for regulating monocyte O-2 production and LDL oxidation, PKCalpha or PKCbeta isoenzyme-specific antisense oligonucleotides were used to selectively suppress isoenzyme expression. We found that suppression of PKCalpha expression inhibited both monocyte-mediated O-2 production and LDL lipid oxidation by activated human monocytes. In contrast, inhibition of PKCbeta expression (including both PKCbetaI and PKCbetaII) did not affect O-2 production or LDL lipid oxidation. Further studies demonstrated that the respiratory burst oxidase responsible for O-2 production remained functionally intact in monocytes with depressed levels of PKCalpha because O-2 production could be restored by treating the monocytes with arachidonic acid. Taken together, our data reveal that PKCalpha, and not PKCbetaI or PKCbetaII, is the predominant isoenzyme required for O-2 production and maximal oxidation of LDL by activated human monocytes.


Subject(s)
Isoenzymes/metabolism , Lipoproteins, LDL/metabolism , Monocytes/metabolism , Protein Kinase C/metabolism , Superoxides/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Oligonucleotides, Antisense/pharmacology , Oxidation-Reduction , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-alpha
17.
J Biol Chem ; 274(2): 939-47, 1999 Jan 08.
Article in English | MEDLINE | ID: mdl-9873035

ABSTRACT

The IkappaB inhibitors regulate the activity of the potent transcription factor nuclear factor-kappaB (NF-kappaB). Following signal-induced IkappaB proteolysis, NF-kappaB translocates into the nucleus to activate transcription of target genes, including IkappaBalpha itself, initiating the "NF-kappaB-IkappaBalpha autoregulatory feedback loop." Upon IkappaBalpha resynthesis, NF-kappaB is subsequently inactivated and redistributed back into the cytoplasm. We have previously reported a robust NF-kappaB-IkappaBalpha autoregulatory feedback loop in HepG2 hepatocytes. Sixty minutes after tumor necrosis factor (TNF-alpha) stimulation, IkappaBalpha is resynthesized to approximately 2-fold greater level than in control cells and completely inhibits NF-kappaB binding. Here we investigate the mechanism for IkappaBalpha resynthesis comparing the effect of stimulation of TNF-alpha with that of interleukin-1 (IL-1alpha). Although either TNF-alpha or IL-1alpha stimulation of protein kinase C (PKC)-down-regulated cells equivalently induces NF-kappaB translocation, the kinetics of IkappaBalpha resynthesis is slowed. Moreover, pretreatment with selective calcium-dependent PKC inhibitors selectively slowed the kinetics of the IL-1alpha-induced overshoot without affecting that produced by TNF-alpha. Down-regulation of PKCalpha by antisense phosphorothioate oligonucleotides and expression vectors selectively blocked the IL-1alpha-induced IkappaBalpha overshoot. In the absence of PKCalpha, although IL-1alpha induced similar amounts of IkappaBalpha transcription and changes in steady-state mRNA, a greater component of IkappaBalpha mRNA was retained in the nucleus. These data indicate a selective role for PKCalpha in IL-1alpha-induced IkappaBalpha resynthesis, which is mediated, at least in part, by post-transcriptional control of mRNA export.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , Interleukin-1/pharmacology , Isoenzymes/metabolism , Liver/metabolism , NF-kappa B/metabolism , Protein Kinase C/metabolism , RNA Processing, Post-Transcriptional , Base Sequence , DNA Primers , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Down-Regulation , Enzyme Inhibitors/pharmacology , Feedback , Humans , Isoenzymes/antagonists & inhibitors , Ligands , Liver/cytology , NF-KappaB Inhibitor alpha , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , RNA, Messenger/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
18.
J Neuroendocrinol ; 10(8): 569-76, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9725708

ABSTRACT

Protein kinase C (PKC) is a critical regulator of signal transduction and cell function in many tissues, including pituitary. Although PKC influences pituitary hormone secretion in adults, its role in determining characteristic perinatal patterns of hormone secretion and synthesis is not known, and the expression of major PKC isotypes in perinatal pituitary is poorly defined. We therefore determined the developmental, cell-specific expression of the major PKC isotypes, using Western analysis and double label immunohistochemistry, in pituitaries of perinatal and mature rats. Expression of specific PKC isotypes was strikingly age-dependent. Pituitary expression of PKC alpha was particularly high in neonates and declined significantly with age, with levels in adult rats approximately half those of neonates as assessed by Western analysis. Similarly, immunohistochemistry indicated that PKC alpha was less abundant in adult than in neonatal pituitaries; the most intensely staining cells of both age groups were identified as somatotrophs and gonadotrophs. In contrast to PKC alpha, pituitary expression of PKC epsilon increased approximately two-fold with advancing age as assessed by Western analysis; this age-dependent pattern was confirmed by immunohistochemistry. Perinatal pituitaries expressed PKC epsilon in some somatotrophs and in all gonadotrophs, whereas PKC epsilon expression was limited to gonadotrophs in the mature pituitary. Pituitary expression of PKC betaII, delta, and zeta did not differ with age, and PKC gamma was not detected in pituitaries of any age group. These results indicate that expression of PKC isotypes within the pituitary is developmentally regulated in a cell-specific and isotype-specific manner, and are consistent with the concept that PKC contributes to the regulation of pituitary function during early development.


Subject(s)
Animals, Newborn , Isoenzymes/metabolism , Pituitary Gland/enzymology , Pituitary Gland/growth & development , Protein Kinase C/metabolism , Aging , Animals , Blotting, Western , Female , Immunohistochemistry , Isoenzymes/analysis , Male , Protein Kinase C/analysis , Rats , Rats, Sprague-Dawley
19.
J Biol Chem ; 273(32): 20261-6, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685375

ABSTRACT

In human erythroleukemia (K562) cells, the highly related protein kinase C (PKC) alpha and PKC betaII isozymes serve distinct functions in cellular differentiation and proliferation, respectively. Previous studies using two domain switch PKC chimera revealed that the catalytic domains of PKC alpha and betaII contain molecular determinants important for isozyme-specific function (Walker, S. D., Murray, N. R., Burns, D. J., and Fields, A. P. (1995) Proc. Natl. Acad. Sci. U.S.A. 92, 9156-9160). We have now analyzed a panel of PKC chimeras to determine the specific region within the catalytic domain important for PKC betaII function. A cellular assay for PKC betaII function was devised based on the finding that PKC betaII selectively translocates to the nucleus and phosphorylates nuclear lamin B in response to the PKC activator bryostatin. This response is strictly dependent upon expression of PKC betaII or a PKC chimera that functions like PKC betaII. We demonstrate that a PKC alpha/betaII chimera containing only the carboxyl-terminal 13 amino acids from PKC betaII (betaII V5) is capable of nuclear translocation and lamin B phosphorylation. These results are consistent with our recent observation that the PKC betaII V5 region binds to phosphatidylglycerol (PG), a potent and selective PKC betaII activator present in the nuclear membrane (Murray, N. R., and Fields, A. P. (1998) J. Biol. Chem. 273, 11514-11520). Soluble betaII V5 peptide selectively inhibits PG-stimulated PKC betaII activity in a dose-dependent fashion, indicating that PG-mediated activation of PKC betaII involves interactions with the betaII V5 region of the enzyme. We conclude that betaII V5 is a major determinant for PKC betaII nuclear function and suggest a model in which binding of PG to the betaII V5 region stimulates nuclear PKC betaII activity during G2 phase of the cell cycle.


Subject(s)
Isoenzymes/chemistry , Protein Kinase C/chemistry , Base Sequence , Enzyme Activation/physiology , Gene Expression Regulation, Enzymologic/genetics , Humans , Lamin Type B , Lamins , Leukemia, Erythroblastic, Acute/enzymology , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/metabolism , Peptide Fragments/pharmacology , Phosphatidylglycerols/metabolism , Phosphorylation , Protein Binding/physiology , Protein Kinase C beta , Protein Kinase C-alpha , Recombinant Fusion Proteins/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
20.
J Biol Chem ; 273(19): 11514-20, 1998 May 08.
Article in English | MEDLINE | ID: mdl-9565565

ABSTRACT

A major mechanism by which protein kinase C (PKC) function is regulated is through the selective targeting and activation of individual PKC isotypes at distinct subcellular locations. PKC betaII is selectively activated at the nucleus during G2 phase of cell cycle where it is required for entry into mitosis. Selective nuclear activation of PKC betaII is conferred by molecular determinants within the carboxyl-terminal catalytic domain of the kinase (Walker, S. D., Murray, N. R., Burns, D. J., and Fields, A. P. (1995) Proc. Natl. Acad. Sci. U. S. A. 92, 9156-9160). We previously described a lipid-like PKC activator in nuclear membranes, termed nuclear membrane activation factor (NMAF), that potently stimulates PKC betaII activity through interactions involving this domain (Murray, N. R., Burns, D. J., and Fields, A. P. (1994) J. Biol. Chem. 269, 21385-21390). We have now identified NMAF as phosphatidylglycerol (PG), based on several lines of evidence. First, NMAF cofractionates with PG as a single peak of activity through multiple chromatographic separations and exhibits phospholipase sensitivity identical to that of PG. Second, purified PG, but not other phospholipids, exhibits dose-dependent NMAF activity. Third, defined molecular species of PG exhibit different abilities to stimulate PKC betaII activity. 1,2-Dioleoyl-PG possesses significantly higher activity than other PG species, suggesting that both fatty acid side chain composition and the glycerol head group are important determinants for activity. Fourth, in vitro binding studies demonstrate that PG binds to the carboxyl-terminal region of PKC betaII, the same region we previously implicated in NMAF-mediated activation of PKC betaII. Taken together, our results indicate that specific molecular species of nuclear PG function to physiologically regulate PKC betaII activity at the nucleus.


Subject(s)
Phosphatidylglycerols/physiology , Protein Kinase C/physiology , Binding Sites , Cell Nucleus/enzymology , Chromatography, Thin Layer , Enzyme Activation , HL-60 Cells , Humans , Intracellular Membranes/enzymology , Nuclear Envelope/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL