Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Cell Biol ; 149(2): 307-16, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10769024

ABSTRACT

Individual members of the serine-arginine (SR) and heterogeneous nuclear ribonucleoprotein (hnRNP) A/B families of proteins have antagonistic effects in regulating alternative splicing. Although hnRNP A1 accumulates predominantly in the nucleus, it shuttles continuously between the nucleus and the cytoplasm. Some but not all SR proteins also undergo nucleo-cytoplasmic shuttling, which is affected by phosphorylation of their serine/arginine (RS)-rich domain. The signaling mechanisms that control the subcellular localization of these proteins are unknown. We show that exposure of NIH-3T3 and SV-40 transformed green monkey kidney (COS) cells to stress stimuli such as osmotic shock or UVC irradiation, but not to mitogenic activators such as PDGF or EGF, results in a marked cytoplasmic accumulation of hnRNP A1, concomitant with an increase in its phosphorylation. These effects are mediated by the MKK(3/6)-p38 pathway, and moreover, p38 activation is necessary and sufficient for the induction of hnRNP A1 cytoplasmic accumulation. The stress-induced increase in the cytoplasmic levels of hnRNP A/B proteins and the concomitant decrease in their nuclear abundance are paralleled by changes in the alternative splicing pattern of an adenovirus E1A pre-mRNA splicing reporter. These results suggest the intriguing possibility that signaling mechanisms regulate pre-mRNA splicing in vivo by influencing the subcellular distribution of splicing factors.


Subject(s)
Alternative Splicing , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Ribonucleoproteins/metabolism , 3T3 Cells , Animals , COS Cells , Cell Line, Transformed , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoproteins , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mice , Osmolar Concentration , Phosphorylation , RNA-Binding Proteins/metabolism , Recombinant Proteins/biosynthesis , Signal Transduction , Simian virus 40 , Transfection , Ultraviolet Rays
2.
Science ; 238(4826): 533-6, 1987 Oct 23.
Article in English | MEDLINE | ID: mdl-2821623

ABSTRACT

The p21 products of ras proto-oncogenes are thought to be important components in pathways regulating normal cell proliferation and differentiation. These proteins acquire transforming properties as a result of activating lesions that convert ras genes to oncogenes in a wide array of malignancies. In Xenopus laevis oocytes, microinjection of transforming ras p21 is a potent inducer of maturation, whereas microinjection of a monoclonal antibody to ras p21 inhibits normal maturation induced by hormones. The phosphoinositide pathway is a ubiquitous system that appears to play a key role in diverse cellular functions. By use of the Xenopus oocyte system, it was possible to quantitate the effects of ras p21 microinjection on individual components of the phosphoinositide pathway. Within 20 minutes of microinjection, levels of phosphatidylinositol 4,5-bisphosphate, inositol 1-phosphate, and inositol bisphosphate increased 1.5- to 2-fold. The most striking effects were on diacylglycerol, which increased 5-fold under the same conditions. In contrast, the normal ras p21 protein induced no detectable alteration in any of the metabolites analyzed. The earliest effects of the transforming p21 on phosphoinositol turnover were observable within 2 minutes, implying a very rapid effect of ras p21 on the enzymes involved in phospholipid metabolism.


Subject(s)
Diglycerides/biosynthesis , Glycerides/biosynthesis , Oocytes/metabolism , Phosphatidylinositols/metabolism , Proto-Oncogene Proteins/pharmacology , Animals , Female , Glycerol/metabolism , Inositol/metabolism , Inositol Phosphates/biosynthesis , Kinetics , Microinjections , Oocytes/drug effects , Phosphatidylinositol 4,5-Diphosphate , Phosphatidylinositols/biosynthesis , Proto-Oncogene Proteins p21(ras) , Xenopus laevis
3.
Nat Commun ; 10(1): 46, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30610188

ABSTRACT

Epigenetically regulated transcriptional plasticity has been proposed as a mechanism of differentiation arrest and resistance to therapy. BCR-ABL leukemias result from leukemic stem cell/progenitor transformation and represent an opportunity to identify epigenetic progress contributing to lineage leukemogenesis. Primary human and murine BCR-ABL+ leukemic progenitors have increased activation of Cdc42 and the downstream atypical protein kinase C (aPKC). While the isoform aPKCζ behaves as a leukemic suppressor, aPKCλ/ι is critically required for oncogenic progenitor proliferation, survival, and B-cell differentiation arrest, but not for normal B-cell lineage differentiation. In vitro and in vivo B-cell transformation by BCR-ABL requires the downregulation of key genes in the B-cell differentiation program through an aPKC λ/ι-Erk dependent Etv5/Satb2 chromatin repressive signaling complex. Genetic or pharmacological targeting of aPKC impairs human oncogenic addicted leukemias. Therefore, the aPKCλ/ι-SATB2 signaling cascade is required for leukemic BCR-ABL+ B-cell progenitor transformation and is amenable to non-tyrosine kinase inhibition.


Subject(s)
Leukemia/pathology , Protein Kinase C/metabolism , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/metabolism , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Epigenesis, Genetic , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic , Humans , Leukemia/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Matrix Attachment Region Binding Proteins/physiology , Mice , Precursor Cells, B-Lymphoid/metabolism , Protein Kinase C/physiology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism
4.
Cell Death Differ ; 13(5): 702-11, 2006 May.
Article in English | MEDLINE | ID: mdl-16322752

ABSTRACT

The atypical protein kinase C (PKC) isoforms (aPKC) have been implicated in the regulation of a number of essential signaling events. Early studies using dominant-negative mutants suggested that they are important intermediaries in the activation of the canonical nuclear factor (NF)-kappaB pathway. More recent data using knockout mice genetically demonstrate that in fact the PKCzeta isoform is essential for the adequate activation of this cascade both upstream and downstream the IkappaB kinase complex. In this review, we summarize the mechanistic details whereby the aPKC pathway regulates important cellular functions and how this is achieved by the ability of these kinases to interact with different protein regulators and adapters, as well as to impinge in NF-kappaB-independent signaling cascades such as the Janus kinase-1/signal transducer and activator of transcription 6 system, which plays a critical role in T-cell-mediated hepatitis and asthma.


Subject(s)
NF-kappa B/metabolism , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , STAT6 Transcription Factor/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Apoptosis , Cell Proliferation , Drosophila , Immunity , Interleukin-4/pharmacology , Janus Kinase 1 , Liver/immunology , Mice , Molecular Sequence Data , Protein Kinase C/genetics , Sequence Homology, Amino Acid , Th2 Cells/metabolism , Transcriptional Activation
5.
Mol Cell Biol ; 21(4): 1218-27, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11158308

ABSTRACT

The MEK5-extracellular signal-regulated kinase (ERK5) tandem is a novel mitogen-activated protein kinase cassette critically involved in mitogenic activation by the epidermal growth factor (EGF). The atypical protein kinase C isoforms (aPKCs) have been shown to be required for cell growth and proliferation and have been reported to interact with the adapter protein p62 through a short stretch of acidic amino acids termed the aPKC interaction domain. This region is also present in MEK5, suggesting that it may be an aPKC-binding partner. Here we demonstrate that the aPKCs interact in an EGF-inducible manner with MEK5 and that this interaction is required and sufficient for the activation of MEK5 in response to EGF. Consistent with the role of the aPKCs in the MEK5-ERK5 pathway, we show that zetaPKC and lambda/iotaPKC activate the Jun promoter through the MEF2C element, a well-established target of ERK5. From all these results, we conclude that MEK5 is a critical target of the aPKCs during mitogenic signaling.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase C/metabolism , Cell Division , Cell Line , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Genes, jun , HeLa Cells , Humans , In Vitro Techniques , Isoenzymes/metabolism , MAP Kinase Kinase 5 , Mitosis , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Signal Transduction
6.
Mol Cell Biol ; 15(6): 3164-70, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7760812

ABSTRACT

Stromelysins, which are the metalloproteinases with the widest substrate specificities, play a critical role in tumor invasion and metastasis. We have previously reported an element (SPRE) of the stromelysin promoter located between nucleotides -1221 and -1203 that is necessary and sufficient for the control of stromelysin gene expression by mitogenic activation, which induces a nuclear activity that binds to this sequence. Using a concatenated probe with several copies of this element to screen a lambda gt11 cDNA expression library from mouse Swiss 3T3 fibroblasts, we report here the molecular cloning of a cDNA coding for a novel protein (SPBP) of 937 amino acids that binds to this element and has several features of a transcription factor, such as a putative leucine zipper region, a nuclear localization signal, and a basic domain with homology to the DNA-binding domains of Fos and Jun. Evidence that SPBP is at least a critical component of the mitogen-induced SPRE nuclear binding activity is presented here. Furthermore, the transfection of an expression plasmid for SPBP transactivates reporter chloramphenicol acetyltransferase plasmids containing either the full-length stromelysin promoter or a single copy of the SPRE cloned upstream of the herpes simplex virus thymidine kinase minimal promoter. Therefore, the results presented here identify a novel transcription factor critically involved in the control of stromelysin expression.


Subject(s)
Metalloendopeptidases/biosynthesis , Transcription Factors/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Gene Expression Regulation, Enzymologic , Matrix Metalloproteinase 3 , Metalloendopeptidases/genetics , Mice , Molecular Sequence Data , Transcription Factors/chemistry , Transcription Factors/isolation & purification
7.
Mol Cell Biol ; 16(1): 105-14, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8524286

ABSTRACT

The members of the atypical subfamily of protein kinase C (PKC) show dramatic structural and functional differences from other PKC isotypes. Thus, in contrast to the classical or novel PKCs, they are not activated by diacylglycerol or phorbol esters. However, the atypical PKCs are the target of important lipid second messengers such as ceramide, phosphatidic acid, and 3'-phosphoinositides. The catalytic and pseudosubstrate sequences in the two atypical PKCs (lambda/iota PKC and zeta PKC) are identical but are significantly different from those of conventional or novel PKCs. It has been shown that microinjection of a peptide with the sequence of the pseudosubstrate of the atypical PKC isotypes but not of alpha PKC or epsilon PKC dramatically inhibited maturation and NF-kappa B activation in Xenopus oocytes, as well as reinitiation of DNA synthesis in quiescent mouse fibroblasts. This indicates that either or both atypical isoforms are important in cell signalling. Besides the pseudosubstrate, the major differences in the sequence between lambda/iota PKC and zeta PKC are located in the regulatory domain. Therefore, any functional divergence between the two types of atypical PKCs will presumably reside in that region. We report here the molecular characterization of lambda-interacting protein (LIP), a novel protein that specifically interacts with the zinc finger of lambda/iota PKC but not zeta PKC. We show in this paper that this interaction is detected not only in vitro but also in vivo, that LIP activates lambda/iota PKC but not zeta PKC in vitro and in vivo, and that this interaction is functionally relevant. Thus, expression of LIP leads to the transactivation of a kappa B-dependent promoter in a manner that is dependent on lambda/iota PKC. To our knowledge, this is the first report on the cloning and characterization of a protein activator of a PKC that binds to the zinc finger domain, which has so far been considered a site for binding of lipid modulators. The fact that LIP binds to lambda/iota PKC but not to the highly related zeta PKC isoform suggests that the specificity of the activation of the members of the different PKC subfamilies will most probably be accounted for by proteins like LIP rather than by lipid activators.


Subject(s)
Carrier Proteins/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Carrier Proteins/genetics , DNA Primers/genetics , DNA, Complementary/genetics , Enzyme Activation , Female , Humans , In Vitro Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , NF-kappa B/genetics , NF-kappa B/metabolism , Oocytes/growth & development , Oocytes/metabolism , Protein Kinase C/genetics , Transcriptional Activation , Xenopus laevis , Zinc Fingers
8.
Mol Cell Biol ; 13(2): 1290-5, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8423794

ABSTRACT

Nuclear factor kappa B (NF-kappa B) plays a critical role in the regulation of a large variety of cellular genes. However, the mechanism whereby this nuclear factor is activated remains to be determined. In this report, we present evidence that in oocytes from Xenopus laevis, (i) ras p21- and phospholipase C (PLC)-mediated phosphatidylcholine (PC) hydrolysis activates NF-kappa B and (ii) protein kinase C zeta subspecies is involved in the activation of NF-kappa B in response to insulin/ras p21/PC-PLC. Thus, the microinjection of either ras p21 or PC-PLC, or the exposure of oocytes to insulin, promotes a significant translocation to the nucleus of an NF-kappa B-like activity. This effect is not observed when oocytes are incubated with phorbol myristate acetate or progesterone, both of which utilize a ras p21-independent pathway for oocyte activation. These data strongly suggest a critical role of the insulin/ras p21/PC-PLC/protein kinase C zeta pathway in the control of NF-kappa B activation.


Subject(s)
NF-kappa B/metabolism , Protein Kinase C/antagonists & inhibitors , Amino Acid Sequence , Animals , Cells, Cultured , Cytosol/metabolism , Enhancer Elements, Genetic , HIV-1/genetics , Insulin/pharmacology , Kinetics , Molecular Sequence Data , Oocytes/drug effects , Oocytes/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins p21(ras)/pharmacology , Transcriptional Activation , Xenopus laevis
9.
Mol Cell Biol ; 14(1): 646-54, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8264633

ABSTRACT

In order to determine whether chronic elevation of intracellular diacylglycerol levels generated by hydrolysis of phosphatidylcholine (PC) by PC-hydrolyzing phospholipase C (PC-PLC) is oncogenic, we generated stable transfectants of NIH 3T3 cells expressing the gene encoding PC-PLC from Bacillus cereus. We found that constitutive expression of this gene (plc) led to transformation of NIH 3T3 cells as evidenced by anchorage-independent growth in soft agar, formation of transformed foci in tissue culture, and loss of contact inhibition. The plc transfectants displayed increased intracellular levels of diacylglycerol and phosphocholine. Expression of B. cereus PC-PLC was confirmed by immunoperoxidase and immunofluorescence staining with an affinity-purified anti-PC-PLC antibody. The NIH 3T3 clones expressing plc induced DNA synthesis, progressed through the cell cycle in the absence of added mitogens, and showed significant growth in low-concentration serum. Transfection with an antisense plc expression vector led to a loss of PC-PLC expression accompanied by a complete reversion of the transformed phenotype, suggesting that plc expression was required for maintenance of the transformed state. Taken together, our results show that chronic stimulation of PC hydrolysis by an unregulated PC-PLC enzyme is oncogenic to NIH 3T3 cells.


Subject(s)
Bacillus cereus/enzymology , Bacillus cereus/genetics , Type C Phospholipases/genetics , 3T3 Cells/cytology , 3T3 Cells/metabolism , Animals , Base Sequence , Cell Cycle , DNA/biosynthesis , DNA, Antisense/genetics , DNA, Bacterial/genetics , Diglycerides/metabolism , Genetic Vectors , Hydrolysis , Mice , Molecular Sequence Data , Phenotype , Phosphatidylcholines/metabolism , Transfection
10.
Mol Cell Biol ; 19(3): 2180-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10022904

ABSTRACT

The atypical protein kinase C (PKC) isotypes (lambda/iotaPKC and zetaPKC) have been shown to be critically involved in important cell functions such as proliferation and survival. Previous studies have demonstrated that the atypical PKCs are stimulated by tumor necrosis factor alpha (TNF-alpha) and are required for the activation of NF-kappaB by this cytokine through a mechanism that most probably involves the phosphorylation of IkappaB. The inability of these PKC isotypes to directly phosphorylate IkappaB led to the hypothesis that zetaPKC may use a putative IkappaB kinase to functionally inactivate IkappaB. Recently several groups have molecularly characterized and cloned two IkappaB kinases (IKKalpha and IKKbeta) which phosphorylate the residues in the IkappaB molecule that serve to target it for ubiquitination and degradation. In this study we have addressed the possibility that different PKCs may control NF-kappaB through the activation of the IKKs. We report here that alphaPKC as well as the atypical PKCs bind to the IKKs in vitro and in vivo. In addition, overexpression of zetaPKC positively modulates IKKbeta activity but not that of IKKalpha, whereas the transfection of a zetaPKC dominant negative mutant severely impairs the activation of IKKbeta but not IKKalpha in TNF-alpha-stimulated cells. We also show that cell stimulation with phorbol 12-myristate 13-acetate activates IKKbeta, which is entirely dependent on the activity of alphaPKC but not that of the atypical isoforms. In contrast, the inhibition of alphaPKC does not affect the activation of IKKbeta by TNF-alpha. Interestingly, recombinant active zetaPKC and alphaPKC are able to stimulate in vitro the activity of IKKbeta but not that of IKKalpha. In addition, evidence is presented here that recombinant zetaPKC directly phosphorylates IKKbeta in vitro, involving Ser177 and Ser181. Collectively, these results demonstrate a critical role for the PKC isoforms in the NF-kappaB pathway at the level of IKKbeta activation and IkappaB degradation.


Subject(s)
Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line, Transformed , Enzyme Activation , Gene Expression Regulation , Humans , I-kappa B Kinase , Isoenzymes/genetics , Isoenzymes/metabolism , Mitogens/pharmacology , Promoter Regions, Genetic , Protein Kinase C/genetics , Protein Kinase C-alpha , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
11.
Mol Cell Biol ; 12(12): 5329-35, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1448068

ABSTRACT

We have used a dominant inhibitory ras mutant (Ha-ras Asn-17) to investigate the relationship of Ras proteins to hydrolysis of phosphatidylcholine (PC) in the transduction of mitogenic signals. Expression of Ha-Ras Asn-17 inhibited NIH 3T3 cell proliferation induced by polypeptide growth factors or phorbol esters. In contrast, the mitogenic activity of PC-specific phospholipase C (PC-PLC) was not inhibited by Ha-Ras Asn-17 expression. Similarly, cotransfection with a cloned PC-PLC gene bypassed the block to NIH 3T3 cell proliferation resulting from expression of the inhibitory ras mutant. Hydrolysis of PC can therefore induce cell proliferation in the absence of normal Ras activity, suggesting that PC-derived second messengers may act downstream of Ras in mitogenic signal transduction. This was substantiated by the finding that Ha-Ras Asn-17 expression inhibited growth factor-stimulated hydrolysis of PC. Taken together, these results indicate that PC hydrolysis is a target of Ras during the transduction of growth factor-initiated mitogenic signals.


Subject(s)
Mitogens/pharmacology , Phosphatidylcholines/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Type C Phospholipases/metabolism , 3T3 Cells , Animals , Blotting, Northern , Cell Division , Cloning, Molecular , Hydrolysis , Mice , Second Messenger Systems , Transfection , Type C Phospholipases/genetics
12.
Mol Cell Biol ; 18(5): 3069-80, 1998 May.
Article in English | MEDLINE | ID: mdl-9566925

ABSTRACT

An increasing number of independent studies indicate that the atypical protein kinase C (PKC) isoforms (aPKCs) are critically involved in the control of cell proliferation and survival. The aPKCs are targets of important lipid mediators such as ceramide and the products of the PI 3-kinase. In addition, the aPKCs have been shown to interact with Ras and with two novel proteins, LIP (lambda-interacting protein; a selective activator of lambda/iotaPKC) and the product of par-4 (a gene induced during apoptosis), which is an inhibitor of both lambda/iotaPKC and zetaPKC. LIP and Par-4 interact with the zinc finger domain of the aPKCs where the lipid mediators have been shown to bind. Here we report the identification of p62, a previously described phosphotyrosine-independent p56(lck) SH2-interacting protein, as a molecule that interacts potently with the V1 domain of lambda/iotaPKC and, albeit with lower affinity, with zetaPKC. We also show in this study that ectopically expressed p62 colocalizes perfectly with both lambda/iotaPKC and zetaPKC. Interestingly, the endogenous p62, like the ectopically expressed protein, displays a punctate vesicular pattern and clearly colocalizes with endogenous lambda/iotaPKC and endogenous zetaPKC. P62 colocalizes with Rab7 and partially with lamp-1 and limp-II as well as with the epidermal growth factor (EGF) receptor in activated cells, but not with Rab5 or the transferrin receptor. Of functional relevance, expression of dominant negative lambda/iotaPKC, but not of the wild-type enzyme, severely impairs the endocytic membrane transport of the EGF receptor with no effect on the transferrin receptor. These findings strongly suggest that the aPKCs are anchored by p62 in the lysosome-targeted endosomal compartment, which seems critical for the control of the growth factor receptor trafficking. This is particularly relevant in light of the role played by the aPKCs in mitogenic cell signaling events.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , Immediate-Early Proteins , Isoenzymes/metabolism , Lysosomes/metabolism , Protein Kinase C/metabolism , Proteins , Adaptor Proteins, Signal Transducing , Biological Transport , Cell Compartmentation , Endocytosis , ErbB Receptors/metabolism , HeLa Cells , Humans , Mitosis , Protein Binding , Sequestosome-1 Protein , Signal Transduction
13.
Mol Cell Biol ; 17(8): 4346-54, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9234692

ABSTRACT

Recent studies have documented the involvement of the atypical protein kinase C (aPKC) isoforms in important cellular functions such as cell proliferation and survival. Exposure of cells to a genotoxic stimulus that induces apoptosis, such as UV irradiation, leads to a profound inhibition of the atypical PKC activity in vivo. In this study, we addressed the relationship between this phenomenon and different proteins involved in the apoptotic response. We show that (i) the inhibition of the aPKC activity precedes UV-induced apoptosis; (ii) UV-induced aPKC inhibition and apoptosis are independent of p53; (iii) Bcl-2 proteins are potent modulators of aPKC activity; and (iv) the aPKCs are located upstream of the interleukin-converting enzyme-like protease system, which is required for the induction of apoptosis by both Par-4 (a selective aPKC inhibitor) and UV irradiation. We also demonstrate here that inhibition of aPKC activity leads to a decrease in mitogen-activated protein (MAP) kinase activity and simultaneously an increase in p38 activity. Both effects are critical for the induction of apoptosis in response to Par-4 expression and UV irradiation. Collectively, these results clarify the position of the aPKCs in the UV-induced apoptotic pathway and strongly suggest that MAP kinases play a role in this signaling cascade.


Subject(s)
Apoptosis/physiology , Intracellular Signaling Peptides and Proteins , Protein Kinase C/metabolism , 3T3 Cells , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspase 1 , Cysteine Endopeptidases/metabolism , Humans , Mice , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/radiation effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Suppressor Protein p53/physiology , Ultraviolet Rays
14.
Mol Cell Biol ; 12(1): 302-8, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309592

ABSTRACT

Cell growth and tumor transformation can be restrained in certain cell systems by the action of transforming growth factor beta (TGF-beta). It has been established that the mechanism whereby TGF-beta 1 inhibits cell growth does not interfere with the triggering of early mitogenic signal transduction mechanisms. Phospholipase C-catalyzed hydrolysis of phosphatidylcholine (PC) is a relatively late step in the cascade activated by growth factors. Therefore, conceivably activation of phospholipase C-catalyzed hydrolysis of PC could be the target of TGF-beta 1 action. In the study reported here, we demonstrate that TGF-beta 1 inhibits the coupling of ras p21 to the activation of PC hydrolysis, which appears to be critical for the antiproliferative effects of TGF-beta 1.


Subject(s)
Phosphatidylcholines/metabolism , Transforming Growth Factor beta/physiology , Type C Phospholipases/metabolism , Animals , Bacillus cereus/enzymology , Cell Line , Enzyme Activation , Genes, myc , Hydrolysis , Insulin/physiology , Keratinocytes/enzymology , Kinetics , Mice , Microinjections , Oocytes/enzymology , Progesterone/physiology , Protamine Kinase/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/metabolism , Type C Phospholipases/antagonists & inhibitors , Xenopus laevis
15.
Mol Cell Biol ; 17(2): 732-41, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9001227

ABSTRACT

The Raf protein kinases function downstream of Ras guanine nucleotide-binding proteins to transduce intracellular signals from growth factor receptors. Interaction with Ras recruits Raf to the plasma membrane, but the subsequent mechanism of Raf activation has not been established. Previous studies implicated hydrolysis of phosphatidylcholine (PC) in Raf activation; therefore, we investigated the role of the epsilon isotype of protein kinase C (PKC), which is stimulated by PC-derived diacylglycerol, as a Raf activator. A dominant negative mutant of PKC epsilon inhibited both proliferation of NIH 3T3 cells and activation of Raf in COS cells. Conversely, overexpression of active PKC epsilon stimulated Raf kinase activity in COS cells and overcame the inhibitory effects of dominant negative Ras in NIH 3T3 cells. PKC epsilon also stimulated Raf kinase in baculovirus-infected Spodoptera frugiperda Sf9 cells and was able to directly activate Raf in vitro. Consistent with its previously reported activity as a Raf activator in vitro, PKC alpha functioned similarly to PKC epsilon in both NIH 3T3 and COS cell assays. In addition, constitutively active mutants of both PKC alpha and PKC epsilon overcame the inhibitory effects of dominant negative mutants of the other PKC isotype, indicating that these diacylglycerol-regulated PKCs function as redundant activators of Raf-1 in vivo.


Subject(s)
Epidermal Growth Factor/pharmacology , Isoenzymes/physiology , Protein Kinase C/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , COS Cells , Cell Division , Cell Line , Diglycerides/metabolism , Enzyme Activation/drug effects , Isoenzymes/genetics , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Isoenzymes/pharmacology , Mice , Mitogens/pharmacology , Mutation , Protein Kinase C/genetics , Protein Kinase C/isolation & purification , Protein Kinase C/metabolism , Protein Kinase C/pharmacology , Protein Kinase C-alpha , Protein Kinase C-epsilon , Proto-Oncogene Proteins c-raf , Recombinant Fusion Proteins , Spodoptera , Tetradecanoylphorbol Acetate/pharmacology , Transfection
16.
Mol Cell Biol ; 12(9): 3776-83, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1508183

ABSTRACT

A number of studies have demonstrated the activation of phospholipase C-mediated hydrolysis of phosphatidylcholine (PC-PLC) both by growth factors and by the product of the ras oncogene, p21ras. Evidence has been presented indicating that the stimulation of this phospholipid degradative pathway is sufficient to activate mitogenesis in fibroblasts as well as that it is sufficient and necessary for induction of maturation in Xenopus laevis oocytes. However, the mechanism whereby PC-PLC transduces mitogenic signals triggered by growth factors or oncogenes remains to be elucidated. In this study, data are presented that show the involvement of protein kinase C zeta subspecies in the channelling of the mitogenic signal activated by insulin-p21ras-PC-PLC in Xenopus oocytes as well as the lack of a critical role of protein kinase C isotypes alpha, beta, gamma, delta, and epsilon in these pathways.


Subject(s)
Oocytes/cytology , Protein Kinase C/physiology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , DNA , Immunoblotting , Maturation-Promoting Factor/physiology , Molecular Sequence Data , Oocytes/enzymology , Oogenesis/physiology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , RNA, Antisense/pharmacology , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
17.
Mol Cell Biol ; 13(12): 7645-51, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246981

ABSTRACT

We have investigated the relationship between hydrolysis of phosphatidylcholine (PC) and activation of the Raf-1 protein kinase in Ras-mediated transduction of mitogenic signals. As previously reported, cotransfection of a PC-specific phospholipase C (PC-PLC) expression plasmid bypassed the block to cell proliferation resulting from expression of the dominant inhibitory mutant Ras N-17. In contrast, PC-PLC failed to bypass the inhibitory effect of dominant negative Raf mutants, suggesting that PC-PLC functions downstream of Ras but upstream of Raf. Consistent with this hypothesis, treatment of quiescent cells with exogenous PC-PLC induced Raf activation, even when normal Ras function was blocked by Ras N-17 expression. Further, activation of Raf in response to mitogenic growth factors was blocked by inhibition of endogenous PC-PLC. Taken together, these results indicate that hydrolysis of PC mediates Raf activation in response to mitogenic growth factors.


Subject(s)
Genes, ras , Phosphatidylcholines/metabolism , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Cell Division , Enzyme Activation , Hydrolysis , Mice , Mutation , Proto-Oncogene Proteins c-raf , Signal Transduction , Transfection , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
18.
Mol Cell Biol ; 13(8): 4770-5, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8336714

ABSTRACT

Nuclear factor kappa B (NF-kappa B) plays a critical role in the regulation of a number of genes. NF-kappa B is a heterodimer of 50- and 65-kDa subunits sequestered in the cytoplasm complexed to inhibitory protein I kappa B. Following stimulation of cells, I kappa B dissociates from NF-kappa B, allowing its translocation to the nucleus, where it carries out the transactivation function. The precise mechanism controlling NF-kappa B activation and the involvement of members of the protein kinase C (PKC) family of isotypes have previously been investigated. It was found that phorbol myristate acetate, (PMA) which is a potent stimulant of phorbol ester-sensitive PKC isotypes, activates NF-kappa B. However, the role of PMA-sensitive PKCs in vivo is not as apparent. It has recently been demonstrated in the model system of Xenopus laevis oocytes that the PMA-insensitive PKC isotype, zeta PKC, is a required step in the activation of NF-kappa B in response to ras p21. We demonstrate here that overexpression of zeta PKC is by itself sufficient to stimulate a permanent translocation of functionally active NF-kappa B into the nucleus of NIH 3T3 fibroblasts and that transfection of a kinase-defective dominant negative mutant of zeta PKC dramatically inhibits the kappa B-dependent transactivation of a chloramphenicol acetyltransferase reporter plasmid in NIH 3T3 fibroblasts. All these results support the notion that zeta PKC plays a decisive role in NF-kappa B regulation in mammalian cells.


Subject(s)
NF-kappa B/metabolism , Protein Kinase C/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Compartmentation , Cloning, Molecular , Enhancer Elements, Genetic , Gene Expression Regulation , Genes, Dominant , In Vitro Techniques , Mice , Molecular Sequence Data , Mutation , Oligodeoxyribonucleotides/chemistry , Transcriptional Activation , Transfection
19.
Mol Endocrinol ; 2(9): 799-805, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2459606

ABSTRACT

BALB/MK is a nontransformed epithelial cell line derived from primary BALB/c mouse keratinocytes that requires epidermal growth factor (EGF) for growth. Using a defined-medium culture system, we investigated the role of physiological concentrations of EGF on phosphoinositide metabolism in these cells. The results show that EGF rapidly activates phospholipase-C mediated phosphoinositide metabolism resulting in the generation of the second messengers inositol 1,4,5-trisphosphate and diacylglycerol. These metabolites control intracellular Ca2+ levels and activate protein kinase C, respectively. Protein kinase C activation in response to EGF was evidenced by the phosphorylation of the acidic 80 kilodalton endogenous protein substrate (p80) specific for this kinase. In contrast, insulin, which acts in concert with EGF to cause BALB/MK cell proliferation, had no effect on phosphoinositide metabolism nor led to any additional stimulation when added in combination with EGF. Taken together, our results show that rapid alterations in phosphoinositide metabolism and protein kinase C activation are associated with the normal mitogenic response of keratinocytes to EGF.


Subject(s)
Epidermal Growth Factor/pharmacology , Epidermis/metabolism , Keratins , Phosphatidylinositols/metabolism , Protein Kinase C/metabolism , Animals , Dose-Response Relationship, Drug , Epidermis/drug effects , Epidermis/enzymology , Mice , Mice, Inbred BALB C , Phosphorylation , Thymidine/analogs & derivatives , Thymidine/metabolism
20.
Endocrinology ; 138(11): 4721-31, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9348199

ABSTRACT

We examined the question of whether insulin activates protein kinase C (PKC)-zeta in L6 myotubes, and the dependence of this activation on phosphatidylinositol (PI) 3-kinase. We also evaluated a number of issues that are relevant to the question of whether diacylglycerol (DAG)-dependent PKCs or DAG-insensitive PKCs, such as PKC-zeta, are more likely to play a role in insulin-stimulated glucose transport in L6 myotubes and other insulin-sensitive cell types. We found that insulin increased the enzyme activity of immunoprecipitable PKC-zeta in L6 myotubes, and this effect was blocked by PI 3-kinase inhibitors, wortmannin and LY294002; this suggested that PKC-zeta operates downstream of PI 3-kinase during insulin action. We also found that treatment of L6 myotubes with 5 microM tetradecanoyl phorbol-13-acetate (TPA) for 24 h led to 80-100% losses of all DAG-dependent PKCs (alpha, beta1, beta2, delta, epsilon) and TPA-stimulated glucose transport (2-deoxyglucose uptake); in contrast, there was full retention of PKC-zeta, as well as insulin-stimulated glucose transport and translocation of GLUT4 and GLUT1 to the plasma membrane. Unlike what has been reported in BC3H-1 myocytes, TPA treatment did not elicit increases in PKCbeta2 messenger RNA or protein in L6 myotubes, and selective retention of this PKC isoform could not explain the retention of insulin effects on glucose transport after prolonged TPA treatment. Of further interest, TPA acutely activated membrane-associated PI 3-kinase in L6 myotubes, and acute effects of TPA on glucose transport were inhibited, not only by the PKC inhibitor, LY379196, but also by both wortmannin and LY294002; this suggested that DAG-sensitive PKCs activate glucose transport through cross-talk with phosphatidylinositol (PI) 3-kinase, rather than directly through PKC. Also, the cell-permeable, myristoylated PKC-zeta pseudosubstrate inhibited insulin-stimulated glucose transport both in non-down-regulated and PKC-depleted (TPA-treated) L6 myotubes; thus, the PKC-zeta pseudosubstrate appeared to inhibit a protein kinase that is required for insulin-stimulated glucose transport but is distinct from DAG-sensitive PKCs. In keeping with the latter dissociation of DAG-sensitive PKCs and insulin-stimulated glucose transport, LY379196, which inhibits PKC-beta (preferentially) and other DAG-sensitive PKCs at relatively low concentrations, inhibited insulin-stimulated glucose transport only at much higher concentrations, not only in L6 myotubes, but also in rat adipocytes, BC3H-1 myocytes, 3T3/L1 adipocytes and rat soleus muscles. Finally, stable and transient expression of a kinase-inactive PKC-zeta inhibited basal and insulin-stimulated glucose transport in L6 myotubes. Collectively, our findings suggest that, whereas PKC-zeta is a reasonable candidate to participate in insulin stimulation of glucose transport, DAG-sensitive PKCs are unlikely participants.


Subject(s)
Diglycerides/pharmacology , Glucose/metabolism , Insulin/physiology , Muscle, Skeletal/metabolism , Protein Kinase C/drug effects , Protein Kinase C/physiology , Protein Serine-Threonine Kinases , Animals , Biological Transport/drug effects , Biological Transport/physiology , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Muscle, Skeletal/cytology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt , Rats , Substrate Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL