Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters








Publication year range
1.
Oncogene ; 29(45): 6004-15, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20818427

ABSTRACT

Overexpression of Ras(V12) in MCF10A cells, an immortalized mammary epithelial cell line, leads to transformation of these cells. We demonstrate that this is accompanied by degradation of C/EBPbeta1. C/EBPbeta is a transcription factor in which three protein isoforms exist because of alternative translation at three in-frame methionines. When C/EBPbeta1 is expressed in MCF10A-Ras(V12) cells, immunoblot analysis reveals that C/EBPbeta1 is degraded in these cells. Treatment of MCF10A-Ras(V12)-C/EBPbeta1 cells with the cdk inhibitor roscovitine leads to stabilization of C/EBPbeta1. It has been previously shown that cdk2 phosphorylates C/EBPbeta on Thr235. We demonstrate that mutation of Thr235 to alanine in C/EBPbeta1 is sufficient to restore the stability of C/EBPbeta1 expression in MCF10A-Ras(V12) cells. Overexpression of Ras(V12) in primary cells induces senescence rather than transformation, thus suppressing tumorigenesis. C/EBPbeta is required for Ras(V12)-induced senescence in primary mouse embryonic fibroblasts. Upregulation of interleukin-6 (IL6) by C/EBPbeta has been shown to be necessary for oncogene-induced senescence, but the specific isoform of C/EBPbeta has not been investigated. We show that the C/EBPbeta1 isoform upregulates IL6 when introduced into normal fibroblasts. In addition, we show that C/EBPbeta1 induces senescence. Taken together, degradation of C/EBPbeta1 by Ras activation may represent a mechanism to bypass OIS.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Cellular Senescence/drug effects , Oncogene Protein p21(ras)/pharmacology , ras Proteins/pharmacology , Breast , CCAAT-Enhancer-Binding Protein-beta , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Cyclin-Dependent Kinases/antagonists & inhibitors , Epithelial Cells , Fibroblasts , Gene Expression Regulation , Humans , Infant , Interleukin-6/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Roscovitine , Up-Regulation
2.
Methods Enzymol ; 407: 660-76, 2006.
Article in English | MEDLINE | ID: mdl-16757360

ABSTRACT

The Ras gene family has been implicated in the development of many human epithelial cancers. Mutations in K-ras or its downstream mediator BRAF have been detected in about two thirds of low-grade serous carcinomas and borderline serous tumors; mutations in K-ras are also often present in benign and invasive mucinous ovarian cancers. Although the oncogenic allele H-ras(V12) is present in only approximately 6% of ovarian cancers, physiologically activated H-ras protein is commonly detected in human ovarian cancer, presumably because of an increase in upstream signals from tyrosine kinase growth factor receptors such as Her-2/neu, despite the lack of a Ras mutation. The mechanisms by which ras oncogenes transform human epithelial cells are not clear. The methods described here are what we use to culture human ovarian surface epithelial cells, to immortalize those cells, and to transform the immortalized cells with oncogenic H-ras or K-ras. These Ras-transformed human ovarian surface epithelial cells form tumors in nude mice and recapitulate many features of human ovarian cancer, thus providing an excellent model system for studying the initiation and progression of human ovarian cancer.


Subject(s)
Cell Transformation, Neoplastic , Oncogene Protein p21(ras)/pharmacology , Ovarian Neoplasms/physiopathology , Ovary/cytology , Animals , Antigens, Viral, Tumor/pharmacology , Cells, Cultured , Epithelial Cells/drug effects , Female , Humans , Immunohistochemistry , Mice , Mice, Nude , Simian virus 40/immunology , Telomerase/pharmacology , Transfection
3.
J Protein Chem ; 22(7-8): 625-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14714729

ABSTRACT

We have previously found that the protein kinase C (PKC) inhibitor, CGP 41 251, blocks oncogenic ras-p21 protein- and beta-PKC-induced oocyte maturation, but only weakly inhibits insulin-induced oocyte maturation (which requires activation of wild-type endogenous ras-p21). Because the dose-response curves for inhibition of oncogenic p21- and beta-PKC-induced oocyte maturation by CGP 41 251 superimpose and because the ras-p21-inactivating antibody, Y13-259, does not inhibit beta-PKC-induced oocyte maturation, we concluded that the oncogenic, but not wild-type, protein requires beta-PKC as a downstream target. Because multiple isoforms of PKC exist and several of these, such as epsilon-PKC, have been found to be important on ras signal transduction pathways, we have investigated which PKC isoforms are critical to each ras protein. For this purpose, we used PKC-isoform-specific inhibitors, which have been shown to inhibit selectively the function and translocation of PKC isoforms in vitro and in vivo. Specifically, the peptides KLFIMN, QEVIRN, and EAVSLKPT each inhibit beta-1, beta-2, and epsilon-PKC, respectively, but do not cross-inhibit other PKC isoforms. We find that the epsilon-PKC inhibitory peptide strongly blocks insulin- but not oncogenic ras-p21-induced oocyte maturation whereas the beta-2 inhibitory peptide more strongly inhibits oncogenic ras-p21-induced oocyte maturation, corroborating our previous studies. The beta-1 inhibitory peptide has little effect on either protein. We conclude that selective inhibition of individual PKC isoforms permits the distinction between signal transduction initiated by oncogenic and activated wild-type p21 proteins and implicate different specific PKC isoforms in mitogenic signal transduction by each of these proteins. The ability to dissect the role of individual PKC isozymes in this regulation is of therapeutic significance.


Subject(s)
Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Protein Kinase C/metabolism , ras Proteins/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Insulin/pharmacology , Isoenzymes/biosynthesis , Mitosis , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/physiology , Oocytes/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Kinase C/biosynthesis , Signal Transduction , Transfection , Xenopus laevis , ras Proteins/antagonists & inhibitors , ras Proteins/physiology
4.
J Protein Chem ; 21(5): 361-6, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12206510

ABSTRACT

In the accompanying article, using molecular dynamics calculations, we found that the 66-77 and 122-138 domains in ras-p21 and the 821-827, 832-845, 917-924, 943-953, and 1003-1020 domains in GAP have different conformations in complexes of GAP with wild-type and oncogenic ras-p21. We have now synthesized peptides corresponding to each of these domains and coinjected them into oocytes with oncogenic p21, which induces oocyte maturation, or injected them into oocytes incubated with insulin that induces maturation by activating wild-type cellular ras-p21. We find that all of these peptides inhibit both agents but do not inhibit progesterone-induced maturation that occurs by a ras-independent pathway. The p21 66-77 and 122-138 peptides cause greater inhibition of oncogenic p21. On the other hand, the GAP 832-845 and 1003-1021 peptides inhibit insulin-induced maturation to a significantly greater extent. Since we have found that activated wild-type and oncogenic p21 activate downstream targets like raf differently, these GAP peptides may be useful probes for identifying elements unique to the wild-type ras-p21 pathway.


Subject(s)
Enzyme Inhibitors/pharmacology , GTPase-Activating Proteins/pharmacology , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Oocytes/growth & development , Peptide Fragments/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Escherichia coli , GTPase-Activating Proteins/chemistry , Insulin/pharmacology , Molecular Sequence Data , Oncogene Protein p21(ras)/chemistry , Oncogene Protein p21(ras)/metabolism , Peptide Fragments/chemistry , Progesterone/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/pharmacology , Signal Transduction/drug effects , Time Factors , Xenopus laevis
5.
J Protein Chem ; 21(5): 367-70, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12206511

ABSTRACT

We have previously found that a ras switch 1 domain peptide (PNC-7, residues 35-47) selectively blocks oocyte maturation induced by oncogenic (Val 12-containing) ras-p21 protein and also blocks c-raf-induced oocyte maturation. We now find that oncogenic ras-p21 does not inhibit oocyte maturation of a constitutively activated raf protein (raf BXB) that is lacking most of the first 301 amino terminal amino acids, including the major ras binding domain and accessory ras-binding regions. We also find that a dominant negative raf that completely blocks c-raf-induced maturation likewise does not block raf-BXB-induced maturation. We conclude that PNC-7 blocks ras by binding to the amino-terminal domain of raf and that raf BXB must initiate signal transduction in the cytosol.


Subject(s)
Mitogens/pharmacology , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/pharmacology , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction/drug effects , Animals , Binding Sites , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Mitogens/chemistry , Mitogens/metabolism , Oncogene Protein p21(ras)/chemistry , Oncogene Protein p21(ras)/metabolism , Oocytes/drug effects , Oocytes/growth & development , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics , Xenopus laevis
6.
Circulation ; 105(8): 968-74, 2002 Feb 26.
Article in English | MEDLINE | ID: mdl-11864927

ABSTRACT

BACKGROUND: Reactive oxygen species play a critical role in inducing apoptosis. The small GTPase p21 Ras and the ERK1/2 MAPK have been proposed as key regulators of the signaling cascade triggered by oxidative stress (H2O2). Harvey-Ras (Ha-Ras) and Kirsten-Ras (Ki-Ras) isoforms are so far functionally indistinguishable, because they activate the same downstream effectors, including ERK1/2. Moreover, ERK1/2 signaling has been involved in both protection and induction of apoptosis. METHODS AND RESULTS: Human umbilical vein endothelial cells (HUVECs) were subjected to H2O2, and apoptosis was detected by fluorescence-activated cell sorting analysis, fluorescence microscopy, and caspase-3 activation. Transfection of Ha-Ras and Ki-Ras genes in HUVECs was performed to evaluate the response to H2O2. We have found that, whereas Ha-Ras decreases tolerance to oxidative stress, Ki-Ras has a potent antiapoptotic activity. Both effects are mediated by ERK1/2. Tolerance to H2O2 is encoded by a unique stretch of lysines at the COOH terminus of the Ki-Ras, lacking in Ha-Ras, and it is relatively independent of the farnesylated anchor. Inhibition of p21 Ras signaling by farnesylation inhibitors increased the resistance to apoptosis in Ha-Ras-expressing cells. CONCLUSIONS: These findings explain the opposite effects of ERK1/2 stimulation on apoptosis found in different cell types and suggest that local activation of ERK1/2 signaling may account for the opposing response to oxidative stress by Ha-Ras or Ki-Ras-expressing cells. Modulation of cell reactivity to oxidative stress by p21 Ras points to the specific and predictive effects of Ras inhibitors in vivo as potential therapeutic drugs in disorders produced by increase of reactive oxygen species inside the cells.


Subject(s)
Endothelium, Vascular/metabolism , Methionine/analogs & derivatives , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein p21(ras)/metabolism , Oxidative Stress/physiology , Amino Acid Substitution , Apoptosis/drug effects , Cells, Cultured , Cytoprotection/drug effects , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Genes, Dominant , Genes, ras , Humans , Hydrogen Peroxide/pharmacology , Methionine/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/pharmacology , Oxidative Stress/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Protein Prenylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Structure-Activity Relationship , Transfection
7.
FEBS Lett ; 474(2-3): 184-8, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10838082

ABSTRACT

Nerve growth factor and activated Ras can induce differentiation of rat pheochromocytoma cells (PC12 cells) [Greene and Tischler (1976) Proc. Natl. Acad. Sci. USA 73, 2424-2428] from a chromaffin cell-like morphology into one that resembles sympathetic neurones. We developed a special treatment of PC12 cells which apparently synchronizes these cells such that they are more useful for semi-quantitative microinjection studies for signal transduction pathways. This treatment leads to a faster and more reproducible differentiation which faithfully reproduces the involvement of Ras in the process and allows a comparison of the biological activity of different Ras mutants. It shows that G12V and Q61L oncogenic mutants are not equally potent in inducing differentiation. Partial loss-of-function mutations T35S, E37G and Y40C are inactive and even a triple combination of these does not restore full biological activity.


Subject(s)
Microinjections , Nerve Growth Factor/pharmacology , PC12 Cells/cytology , ras Proteins/pharmacology , Amino Acid Substitution/genetics , Animals , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Size/drug effects , Dactinomycin/pharmacology , Dose-Response Relationship, Drug , Mutation/genetics , Nerve Growth Factor/antagonists & inhibitors , Neurites/drug effects , Neurites/metabolism , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Oncogene Protein p21(ras)/pharmacology , PC12 Cells/drug effects , PC12 Cells/metabolism , Rats , Reproducibility of Results , Signal Transduction/drug effects , Time Factors , Transcription, Genetic/drug effects , ras Proteins/genetics , ras Proteins/metabolism
8.
J Protein Chem ; 18(8): 875-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10839624

ABSTRACT

In the preceding paper we performed molecular dynamics calculations of the average structures of the SOS protein bound to wild-type and oncogenic ras-p21. Based on these calculations, we have identified four major domains of the SOS protein, consisting of residues 631-641, 676-691, 718-729, and 994-1004, which differ in structure between the two complexes. We have now microinjected synthetic peptides corresponding to each of these domains into Xenopus laevis oocytes either together with oncogenic (Val 12)-p21 or into oocytes subsequently incubated with insulin. We find that the first three peptides inhibit both oncogenic and wild-type p21-induced oocyte maturation, while the last peptide much more strongly inhibits oncogenic p21 protein-induced oocyte maturation. These results suggest that each identified SOS region is involved in ras-stimulated signal transduction and that the 994-1004 domain is involved uniquely with oncogenic ras-p21 signaling.


Subject(s)
Oncogene Protein p21(ras)/metabolism , Oocytes/physiology , Peptide Fragments/metabolism , Son of Sevenless Proteins/metabolism , Amino Acid Sequence , Animals , Dose-Response Relationship, Drug , Female , Insulin/metabolism , Insulin/pharmacology , Molecular Sequence Data , Oncogene Protein p21(ras)/antagonists & inhibitors , Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Peptide Fragments/pharmacology , Son of Sevenless Proteins/pharmacology , Time Factors
9.
Eur J Cancer ; 34(8): 1260-7, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9849489

ABSTRACT

8-Chloroadenosine 3',5'-monophosphate (8-Cl-cAMP), a site-selective cyclic adenosine 3',5'-monophosphate (cAMP) analogue, exhibits growth inhibition in a broad spectrum of cancer cell lines. We investigated the effect of 8-Cl-cAMP on c-ras-transformed mouse fibroblasts (MP3/3T3) which were established by transfection of Balb3T3 cells (Balb3T3) with the point-mutated c-ras gene [G12-->V12]. 8-Cl-cAMP (2-5 microM) exerted over 80% growth inhibition by day 4 on MP3/3T3, while inhibiting parental Balb3T3 cell growth less than 40%. In order to distinguish the effect of 8-Cl-cAMP from that of 8-chloroadenosine (8-Cl-adenosine), we examined the effect of 8-Cl-cAMP in serum-free medium. 8-Cl-cAMP demonstrated a potent growth inhibition of MP3/3T3 cells cultured in serum-free medium, suggesting that the growth inhibitory effect of 8-Cl-cAMP was not due to its hydrolysed product, 8-Cl-adenosine. In addition, both Balb3T3 and MP3/3T3 contained cAMP phosphodiesterases mainly composed of isozyme IV which has previously been reported to be insensitive towards the hydrolysis of 8-Cl-cAMP. Non-transformed Balb3T3 cells contained only type II cAMP-dependent protein kinase (PKA), whereas transformed MP3/3T3 exhibited a marked increase in type I PKA. The growth inhibition of MP3/3T3 by 8-Cl-cAMP accompanied almost complete elimination of type I PKA without affecting type II PKA. Moreover, 8-Cl-cAMP induced an arrest in the G0/G1-phase of the cell cycle in MP3/3T3. 8-Cl-adenosine had little or no effect on the cell cycle kinetics of MP3/3T3 cells. These results show that 8-Cl-cAMP is a novel cAMP analogue which selectively eliminates type I PKA to induce growth inhibition in transformed fibroblasts.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/analogs & derivatives , Antineoplastic Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/drug effects , Oncogene Protein p21(ras)/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cell Cycle/drug effects , Cell Division/drug effects , Cell Transformation, Neoplastic , Cyclic AMP-Dependent Protein Kinase Type II , Fibroblasts/metabolism , Fibroblasts/pathology , Genes, ras , Mice
10.
Cancer Lett ; 115(2): 235-41, 1997 May 19.
Article in English | MEDLINE | ID: mdl-9149130

ABSTRACT

Hypusine formation on the eukaryotic initiation factor 5A (eIF-5A) precursor is ubiquitously present in eukaryotic cells and archebacteria. In this reaction, deoxyhypusine synthase catalyzes the conversion of one unique lysine residue on eIF-5A to deoxyhypusine using spermidine as the substrate. Hydroxylation of the deoxyhypusine residue completes hypusine formation on eIF-5A. Hypusine formation activity can be measured by an in vitro labeling technique in polyamine-depleted cells. In addition, an in vitro cross-labeling assay can be employed to measure simultaneously the relative deoxyhypusine synthase activity and protein substrate amount. Using these approaches, together with Western blot analysis, we showed that hypusine formation activity is serum-responsive and significantly elevated in Ras oncogene transfected NIH3T3 cells as compared to NIH3T3 cells. The large difference, >30-fold, in hypusine formation activity between these two cells is mainly due to difference in the amount of newly synthesized eIF-5A precursor rather than deoxyhypusine synthase. The deoxyhypusine synthase activity is about three-fold higher in Ras-3T3 cells than in 3T3 cells, and remains constant throughout serum stimulation in both cells. Despite the significant difference in eIF-5A protein amounts, the eIF-5A mRNA levels in 3T3 cells and in Ras-3T3 cells are almost identical. Furthermore, unlike serum-dependent increase in eIF-5A precursor protein, the eIF-5A mRNA in both cells is constitutively expressed after serum stimulation, suggesting that eIF-5A gene is regulated at posttranscriptional/translational level during serum stimulation and cell transformation.


Subject(s)
3T3 Cells/metabolism , Cell Transformation, Neoplastic , Lysine/analogs & derivatives , Oncogene Protein p21(ras)/pharmacology , Peptide Initiation Factors/biosynthesis , RNA-Binding Proteins , 3T3 Cells/drug effects , Animals , Eflornithine/pharmacology , Gene Expression , Lysine/biosynthesis , Mice , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Spermidine/metabolism , Eukaryotic Translation Initiation Factor 5A
11.
Oncogene ; 13(9): 1839-46, 1996 Nov 07.
Article in English | MEDLINE | ID: mdl-8934529

ABSTRACT

Purified amino-terminal Src homology 2 (SH2) domains of GAP, PLCgamma1 and the p85alpha subunit of PI 3-kinase, as well as the carboxy-terminal SH2 domain of the latter protein and the unique SH2 domain of Grb2, were injected into full grown, stage VI Xenopus laevis oocytes. None of the injected domains showed any effect when injected alone, nor did they affect the rate of GVBD induced by progesterone, an adenylate cyclase-dependent process. On the other hand, the unique Grb2 SH2 domain and all N-terminal SH2 domains injected inhibited to various degrees the rate of insulin-induced GVBD, a tyrosine kinase dependent pathway. Interestingly, and in contrast to the behavior shown by the N-terminal domain of the same molecule, the C-terminal SH2 domain of p85 did not inhibit, but slightly accelerated the rate of GVBD induced by insulin. Furthermore, whereas the Grb SH2 domain and all N-terminal SH2 domains tested failed to co-operate with normal Ras protein to induce GVBD, the C-terminal SH2 domain of p85alpha exhibited significant synergy when coinjected with normal Ras protein, indicating that the C- and N-terminal SH2 domains of p85alpha exert opposite (positive and negative, respectively) regulatory roles in the control of oocyte insulin/Ras signaling pathways. Our results demonstrate that the purified, isolated SH2 domains retain structural and functional specificity and that Xenopus oocytes constitute an useful biological system to analyse their functional role in tyrosine kinase signaling pathways.


Subject(s)
Adaptor Proteins, Signal Transducing , Insulin/metabolism , Oncogene Protein p21(ras)/metabolism , Oocytes/physiology , Signal Transduction , src Homology Domains/physiology , Animals , Female , GRB2 Adaptor Protein , GTPase-Activating Proteins , Insulin/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Meiosis/drug effects , Microinjections , Oncogene Protein p21(ras)/isolation & purification , Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Phosphatidylinositol 3-Kinases , Phospholipase C gamma , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Progesterone/metabolism , Progesterone/pharmacology , Protein-Tyrosine Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Xenopus laevis/metabolism , ras GTPase-Activating Proteins
12.
Toxicol Appl Pharmacol ; 141(1): 93-101, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8917680

ABSTRACT

H-ras oncogenes have been identified in greater than 50% of the most common forms of human neoplasia. Ras-related proteins have been postulated to mediated signal transduction pathways involving mitogen-activated protein (MAP) kinases and nuclear responses that may be involved in the induction of apoptosis. We examined whether expression of H-ras oncogene conferred resistance or susceptibility to the morphologic effects of the protein phosphatase inhibitor, okadaic acid, using a tumorigenic H-ras-transformed normal rat kidney epithelial cell line, NRK-H/6.1. We also examined whether okadaic acid induced apoptosis correlated with a differential effect on kinase activity in H-Ras-transformed cells as compared to the nontransformed NRK-52E cells. Treatment with various concentrations of okadaic acid produced rapid and extensive morphologic changes characteristic of apoptosis in both cell types. Equimolar okadaic acid concentrations for 2 or 4 hr resulted in cell detachment and loss of membrane integrity (as measured by propidium iodide uptake) in 74% (0.5 microM) and 78% (1.0 microM) of the H-Ras-transformed cells as compared to 8 and 25%, respectively, in the non-transformed cells. Furthermore, a higher basal level of kinase activity was observed in the H-Ras-transformed cells as compared to the nontransformed cells. Okadaic acid-induced apoptosis correlated with activation of members of the MAP kinase family, raf-1 and protein kinase C (PKC). These studies show that H-ras oncogene expression imparts selective susceptibility to cell death induced by phosphatase inhibition. The observed increase in susceptibility to okadaic acid-induced apoptosis appears to involve the modulation of raf-1, PKC, and MAP kinase activities. These findings may be significant in the elucidation of mechanisms for selective induction of cell death in tumor cells expressing H-ras oncogene.


Subject(s)
Apoptosis/drug effects , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/pharmacology , Kidney/drug effects , Kidney/metabolism , Okadaic Acid/toxicity , Oncogene Protein p21(ras)/pharmacology , Phosphotransferases/metabolism , Animals , Cell Death , DNA Fragmentation/drug effects , Epithelium/drug effects , Epithelium/metabolism , Kidney/ultrastructure , Rats
13.
Neuroscience ; 70(4): 1067-79, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8848168

ABSTRACT

We have examined whether p21Ras proteins can rescue nerve growth factor-deprived rat sympathetic neurons from death, to test further our hypothesis that p21Ras is a central mediator in the nerve growth factor-to-survival signalling pathway. After crosslinking [125I]nerve growth factor to live neurons, two forms of Trk (molecular weight approximately 140,000 and 115,000) were immunoprecipitated with anti-Trk antibodies. Nerve growth factor induced tyrosine phosphorylation of both Trk forms and at least two additional proteins. When these phosphorylations were prevented by staurosporine (in a protein kinase C-independent manner) the neurons died. However, neurons were rescued from death due to staurosporine treatment by intracellular loading of oncogenic Ha-Ras(val12) protein. Both Ha-Ras(val12) and cellular Ha-Ras proteins maintained survival for several days in the absence of nerve growth factor and mimicked other actions of nerve growth factor, inducing rapid c-Fos protein expression and robust neurite outgrowth. Conversely, Fab fragments of neutralizing antibodies to p21Ras which blocked the capacity of nerve growth factor to promote neuron survival were also found to inhibit the early expression of c-Fos protein in these neurons. The close correspondence observed between the timing of onset of c-Fos responsiveness and acquisition of nerve growth factor-dependence in embryonic day 17 sympathetic neurons, and the coordinate increase found in both parameters until embryonic day 19 indicates that c-Fos protein expression is a good biochemical indicator of the presence of a functional nerve growth factor-to-survival signal transduction pathway. Nevertheless, expression of c-Fos is not sufficient for survival since phorbol esters induce c-Fos with no effect on survival. These data strengthen our proposal that p21Ras proteins are crucial anti-apoptotic mediators of survival in rat sympathetic neurons by demonstrating that p21Ras is both necessary and sufficient to rescue neurons which are disabled from signalling through Trk receptors.


Subject(s)
Ganglia, Sympathetic/physiology , Nerve Growth Factors/pharmacology , Oncogene Protein p21(ras)/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Phosphorylation , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar
14.
Eur J Biochem ; 229(3): 615-20, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7758454

ABSTRACT

A mutant gelsolin, [His321]gelsolin, was isolated from R1, a flat revertant of human activated c-Ha-ras oncogene-transformed NIH/3T3 cells (EJ-NIH/3T3) produced by ethylmethanesulfonate treatment. [His321]Gelsolin has a histidine instead of a proline residue at position 321 and suppresses the tumorigenicity of EJ-NIH/3T3 cells when it is constitutively expressed [Müllauer, L., Fujita, H., Ishizaki, A. & Kuzumaki, N. (1993) Oncogene 8, 2531-2536]. To investigate the biochemical consequences of the amino acid substitution of His321, we expressed the [His321]gelsolin and wild-type gelsolin in Escherichia coli, purified them, and analyzed their effects on actin, polyphosphoinositol lipids and phospholipase C. [His321]Gelsolin has decreased actin-filament-severing activity and increased nucleating activity compared with wild-type gelsolin in vitro. Furthermore, compared to wild-type gelsolin both nucleation and severing by [His321]gelsolin are inhibited more strongly by the phosphoinositol lipids phosphatidylinositol 4-phosphate (PtdInsP) and phosphatidylinositol 4,5-bisphosphate (PtdInsP2). In addition, [His321]gelsolin inhibits PtdInsP2 hydrolysis by phospholipase C gamma 1 more strongly than wild-type gelsolin in vitro because of its higher binding capacity for phosphoinositol lipid. Gelsolin has six homologous amino acid repeats called S1-S6. Our results suggest that the segment S3 which contains the mutation is functionally relevant for regulation of gelsolin's activities even though the relevant actin-binding domains are in segments 1, 2, and 4-6, and that the region around the residue 321 may contain a phosphoinositol-lipid-binding site. Altered functions of [His321]gelsolin might be important for the loss of tumorigenicity of the ras-transformed cells.


Subject(s)
Gelsolin/physiology , Histidine , Oncogene Protein p21(ras)/pharmacology , 3T3 Cells , Actins/metabolism , Animals , Base Sequence , Cell Line, Transformed/drug effects , Cell Line, Transformed/metabolism , DNA Primers/chemistry , Escherichia coli/genetics , Gelsolin/genetics , Gelsolin/isolation & purification , Gene Expression , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphatidylinositols/metabolism , Structure-Activity Relationship , Type C Phospholipases/metabolism
15.
J Biomol Struct Dyn ; 11(3): 443-58, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8129867

ABSTRACT

We previously reported a complete computer-based three-dimensional structure for residues 1-171 of the Gly 12-containing ras-gene-encoded p21 protein complexed with GDP. This structure was subsequently shown to closely agree with a high-resolution x-ray crystallographic structure of p21. In this communication, we report a molecular dynamics stimulation of the modelled structure in an explicit shell of water molecules to identify domains within the protein that are unusually flexible. These domains represent regions which are most likely to undergo important conformational changes when the protein is activated by binding to GTP or by oncogenic amino acid substitutions such as Val for Gly 12. The starting structure was surrounded with water molecules, temperature-equilibrated and then followed over a 100 ps trajectory during which time the energy converged after about 50 ps. Regions of the protein that were found to have the largest coordinate fluctuations involved residues 12-16, 30-35, 40-52, 60-73, 85-89, 101-109, 119-123, and 127-131. Many of these sequences with high flexibility have been implicated in the functioning of this protein. Since the overall largest fluctuations were observed for residues 101-106 and 119-123, p21 peptides containing these residues (96-110 and 115-126) were synthesized and were found to inhibit strongly the effects of oncogenic p21 protein in an oocyte maturation assay. These results indicate that the flexible p21 sequences may constitute critical functional domains of the activated protein and that this general approach may be useful for identification of important functional domains in proteins.


Subject(s)
Guanosine Triphosphate/metabolism , Oncogene Protein p21(ras)/chemistry , 3T3 Cells , Animals , Chromatography, High Pressure Liquid , Computer Simulation , Crystallography, X-Ray , Glycine/chemistry , Mice , Models, Molecular , Oncogene Protein p21(ras)/metabolism , Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Oocytes/physiology , Peptide Fragments/pharmacology , Protein Conformation , Valine/chemistry , Xenopus laevis
16.
Res Commun Chem Pathol Pharmacol ; 79(2): 131-40, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8451539

ABSTRACT

A nickel-binding serine proteinase inhibitor, pNiXa (43 kDa), was isolated from Xenopus ovary and assayed for effects on oocyte maturation. Microinjection of pNiXa (0.12 pmol/50 nl) induced maturation in 60% of Xenopus oocytes, beginning at 4 hours and reaching completion by 9 hours. Microinjection of oncogenic ras-p21 protein (0.12 pmol/50 nl) induced maturation in 79% of oocytes, beginning at 6 hours and reaching completion by 12 hours. Microinjection of pNiXa in combination with ras-p21 protein had a synergistic effect on maturation, which occurred in 92% of oocytes, beginning at 4 hours and reaching completion by 9 hours. Oocyte maturation did not occur in control oocytes, which received a microinjection of bovine serum albumin. In oocytes exposed to a combination of pNiXa (0.12 pmol/50 nl, by microinjection) and progesterone (10 micrograms/ml, in the medium), maturation was intermediate (68% at 9 hours) between that induced by pNiXa (60%) or progesterone (85%) alone. This study shows (a) that pNiXa is a potent inducer of oocyte maturation, (b) that pNiXa's effect is synergistic with that of oncogenic ras-p21 protein, and (c) that pNiXa partially antagonizes progesterone induction of oocyte maturation.


Subject(s)
Carrier Proteins/pharmacology , Nickel , Oncogene Protein p21(ras)/pharmacology , Oocytes/physiology , Serpins , Xenopus Proteins , Animals , Carrier Proteins/isolation & purification , Cell Cycle/drug effects , Drug Synergism , Female , Ovary/chemistry , Progesterone/pharmacology , Xenopus laevis
17.
Exp Cell Res ; 203(2): 329-35, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1459198

ABSTRACT

We have recently shown that a peptide (residues 35-47) from a functional region of the ras p21 protein, thought to be involved in the binding of p21 to GTPase activating protein, the antibiotic azatyrosine, known to induce the ras-recision gene, and the selective protein kinase C inhibitor, CGP 41,251, all inhibit oncogenic p21 protein-induced maturation of oocytes in a dose-dependent manner. We now show that these three agents only partially inhibit insulin-induced oocyte maturation, known to be dependent on activation of cellular p21 protein. On the other hand, the anti-p21 protein antibody Y13-259 completely inhibits both insulin- and oncogenic p21 protein-induced maturation as does a tetrapeptide, CVIM, known to block the enzyme farnesyl transferase which covalently attaches the farnesyl moiety to the p21 protein allowing it to attach to the cell membrane. Our results suggest that while the oncogenic and insulin-activated normal p21 proteins share certain elements of their signal transduction pathways in common, these pathways diverge and allow for selective inhibition of the oncogenic pathway.


Subject(s)
Insulin/pharmacology , Oncogene Protein p21(ras)/pharmacology , Oocytes/drug effects , Staurosporine/analogs & derivatives , Alanine/analogs & derivatives , Alanine/pharmacology , Alkaloids/pharmacology , Amino Acid Sequence , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Female , Molecular Sequence Data , Oligopeptides/pharmacology , Oocytes/cytology , Protein Kinase C/antagonists & inhibitors , Protein Prenylation , Signal Transduction , Xenopus laevis
18.
J Biol Chem ; 267(28): 20346-51, 1992 Oct 05.
Article in English | MEDLINE | ID: mdl-1328197

ABSTRACT

Mitogen-activated protein (MAP) kinase is a serine/threonine kinase whose function is thought to be essential for the transduction of mitogenic signals. MAP kinase is activated by phosphorylation induced by a variety of extracellular stimuli, and its direct upstream activator has been identified. Using amphibian and mammalian systems, we show here that ras can activate MAP kinase and its activator. Injection of v-Ha-ras p21 into Xenopus immature oocytes activated both MAP kinase and maturation-promoting factor (MPF) activities. The activation of MAP kinase preceded that of MPF, demonstrating that ras activates MAP kinase in an MPF-independent pathway. Moreover, we found that the MAP kinase activator is also activated in ras-injected oocytes. Activation of MAP kinase and its activator occurred also when the v-Ki-ras gene was conditionally induced in rat fibroblastic 3Y1 cells. Furthermore, we observed that ras activated MAP kinase and its activator in a cell-free system prepared from Xenopus oocytes. Using an antibody against the Xenopus 45-kDa MAP kinase activator, we demonstrated that the 45-kDa activator molecule was activated by ras. These findings suggest that the MAP kinase activator/MAP kinase system may be the downstream components of ras signal transduction pathways.


Subject(s)
Genes, ras , Oncogene Protein p21(ras)/pharmacology , Protein Kinases/metabolism , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases , Cations, Divalent , Cell-Free System , Cells, Cultured , Enzyme Activation , Magnesium/metabolism , Manganese/metabolism , Maturation-Promoting Factor/metabolism , Microinjections , Phosphorylation , Plasmids , Precipitin Tests , Rats , Xenopus
19.
Genes Dev ; 6(9): 1621-30, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1325391

ABSTRACT

Ras proteins are membrane-associated transducers of eternal stimuli to unknown intracellular targets. The constitutively activated v-ras oncogene induces dedifferentiation in thyroid cells. v-Ras appears to act by stimulating protein kinase C (PKC), which inhibits the nuclear migration of the catalytic subunit of the cAMP-dependent protein kinase A (PKA). Nuclear tissue-specific and housekeeping trans-acting factors that are dependent on phosphorylation by PKA are thus inactivated. Exclusion of the PKA subunit from the nucleus could represent a general mechanism for the pleiotropic effects of Ras and PKC on cellular growth and differentiation.


Subject(s)
Cell Differentiation/genetics , Oncogene Protein p21(ras)/genetics , Protein Kinase C/metabolism , Protein Kinases/genetics , Thyroid Gland/cytology , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/enzymology , Cyclic AMP/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Fluorescent Antibody Technique , Molecular Sequence Data , Oncogene Protein p21(ras)/pharmacology , Protein Kinases/metabolism , Rats , Sphingosine/pharmacology , Temperature , Thyroid Gland/enzymology
20.
FEBS Lett ; 304(2-3): 261-4, 1992 Jun 15.
Article in English | MEDLINE | ID: mdl-1535600

ABSTRACT

Activation of T-cells infected by HIV-1 results in activation of long terminal repeat (LTR)-dependent viral transcription and ultimately the production of infectious virus. Although full T-cell activation requires a complex series of intracellular signals, including protein kinase C activation, calcium mobilisation, and less-well defined lymphokine-induced signals, the HIV-1 LTR can be activated by subsets of these signals. We have studied the interaction of these signals in the human lymphoma line, Jurkat, in activation of the HIV-1 LTR. The HIV promoter was induced by IL-1 and phorbol ester activation of PKC but not by a calcium ionophore. The constitutively active form of Ha-ras could replace phorbol ester stimulation of the HIV promoter and of a synthetic promoter containing NF kappa B binding sites.


Subject(s)
HIV-1/growth & development , Oncogene Protein p21(ras)/pharmacology , Signal Transduction/physiology , T-Lymphocytes/microbiology , Virus Activation/drug effects , Enzyme Activation , HIV Long Terminal Repeat/drug effects , Humans , Interleukin-1/pharmacology , Phorbol Esters/pharmacology , Protein Kinase C/metabolism , Receptors, Immunologic/metabolism , Receptors, Interleukin-1 , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL