Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Oncogene ; 35(43): 5692-5698, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27086924

ABSTRACT

The Ras-related (R-Ras) isoforms TC21, R-Ras and M-Ras are members of the Ras superfamily of small GTPases. R-Ras family proteins are frequently overexpressed in human cancers, and expression of activated mutants of these GTPases is sufficient to induce cell transformation. Unlike Ras, few activating mutations of R-Ras proteins have been reported in human cancer, and very little is known about the regulation of their activity. In this study, we report that TC21 and R-Ras are phosphorylated on a conserved serine, Ser186 and Ser201, respectively, in intact cells. This residue is located in the C-terminal hypervariable region of the proteins and is not conserved in M-Ras. We show that the MAP kinases ERK1/2 phosphorylate TC21 and R-Ras on this C-terminal serine residue both in vitro and in vivo. Phosphorylation of R-Ras proteins does not affect their subcellular localization or stability but rather stimulates their activation. Phosphorylation-defective mutants of R-Ras and TC21 are compromised in their ability to promote cancer cell adhesion and migration/invasion, respectively. Importantly, we show that phosphorylation of TC21 and R-Ras potentiates their tumorigenic activity in immunodeficient mice. Our results identify a novel regulatory mechanism of the small GTPases TC21 and R-Ras that controls their oncogenic potential.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monomeric GTP-Binding Proteins/metabolism , Amino Acid Sequence , Humans , Intracellular Space , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Monomeric GTP-Binding Proteins/chemistry , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/pharmacology , Protein Transport
2.
Oncogene ; 26(22): 3227-39, 2007 May 14.
Article in English | MEDLINE | ID: mdl-17496918

ABSTRACT

The Ras-dependent extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein (MAP) kinase pathway plays a central role in cell proliferation control. In normal cells, sustained activation of ERK1/ERK2 is necessary for G1- to S-phase progression and is associated with induction of positive regulators of the cell cycle and inactivation of antiproliferative genes. In cells expressing activated Ras or Raf mutants, hyperactivation of the ERK1/2 pathway elicits cell cycle arrest by inducing the accumulation of cyclin-dependent kinase inhibitors. In this review, we discuss the mechanisms by which activated ERK1/ERK2 regulate growth and cell cycle progression of mammalian somatic cells. We also highlight the findings obtained from gene disruption studies.


Subject(s)
G1 Phase/physiology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , S Phase/physiology , Animals , Cell Proliferation , Enzyme Activation/genetics , Enzyme Activation/physiology , G1 Phase/genetics , Humans , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/deficiency , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , S Phase/genetics
3.
EMBO J ; 20(23): 6672-82, 2001 Dec 03.
Article in English | MEDLINE | ID: mdl-11726503

ABSTRACT

The activity of the cyclin-dependent kinase inhibitor p27 is controlled by its concentration and subcellular localization. However, the mechanisms that regulate its intracellular transport are poorly understood. Here we show that p27 is phosphorylated on Ser10 in vivo and that mutation of Ser10 to Ala inhibits p27 cytoplasmic relocalization in response to mitogenic stimulation. In contrast, a fraction of wild-type p27 and a p27(S10D)-phospho-mimetic mutant translocates to the cytoplasm in the presence of mitogens. G1 nuclear export of p27 and its Ser10 phosphorylation precede cyclin-dependent kinase 2 (Cdk2) activation and degradation of the bulk of p27. Interestingly, leptomycin B-mediated nuclear accumulation accelerates the turnover of endogenous p27; the p27(S10A) mutant, which is trapped in the nucleus, has a shorter half-life than wild-type p27 and the p27(S10D) mutant. In summary, p27 is efficiently degraded in the nucleus and phosphorylation of Ser10 is necessary for the nuclear to cytoplasmic redistribution of a fraction of p27 in response to mitogenic stimulation. This cytoplasmic localization may serve to decrease the abundance of p27 in the nucleus below a certain threshold required for activation of cyclin-Cdk2 complexes.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cytoplasm/metabolism , Serine/metabolism , Tumor Suppressor Proteins/biosynthesis , Alanine/genetics , Animals , Antifungal Agents/pharmacology , Cell Cycle , Cell Line , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Activation , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Fatty Acids, Unsaturated/pharmacology , Fibroblasts/metabolism , Immunoblotting , Microscopy, Fluorescence , Mutation , Phosphorylation , Protein Binding , Protein Transport , Rats , Serine/genetics , Tamoxifen/pharmacology , Time Factors , Transfection , Ubiquitin/metabolism
4.
Genes Dev ; 15(20): 2702-19, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11641276

ABSTRACT

Rho-like GTPases play a pivotal role in the orchestration of changes in the actin cytoskeleton in response to receptor stimulation, and have been implicated in transcriptional activation, cell growth regulation, and oncogenic transformation. Recently, a role for RhoA in the regulation of cardiac contractility and hypertrophic cardiomyocyte growth has been suggested but the mechanisms underlying RhoA function in the heart remain undefined. We now report that transcription factor GATA-4, a key regulator of cardiac genes, is a nuclear mediator of RhoA signaling and is involved in the control of sarcomere assembly in cardiomyocytes. Both RhoA and GATA-4 are essential for sarcomeric reorganization in response to hypertrophic growth stimuli and overexpression of either protein is sufficient to induce sarcomeric reorganization. Consistent with convergence of RhoA and GATA signaling, RhoA potentiates the transcriptional activity of GATA-4 via a p38 MAPK-dependent pathway that phosphorylates GATA-4 activation domains and GATA binding sites mediate RhoA activation of target cardiac promoters. Moreover, a dominant-negative GATA-4 protein abolishes RhoA-induced sarcomere reorganization. The identification of transcription factor GATA-4 as a RhoA mediator in sarcomere reorganization and cardiac gene regulation provides a link between RhoA effects on transcription and cell remodeling.


Subject(s)
DNA-Binding Proteins/metabolism , Myocardium/metabolism , Transcription Factors/metabolism , rhoA GTP-Binding Protein/genetics , Adenoviridae/genetics , Blotting, Northern , Blotting, Western , Cells, Cultured , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Endothelin-1/pharmacology , Enhancer Elements, Genetic , Fluorescent Antibody Technique , GATA4 Transcription Factor , Gene Expression Regulation , Humans , In Situ Hybridization , Mitogen-Activated Protein Kinases/metabolism , Phosphoprotein Phosphatases/pharmacology , Promoter Regions, Genetic , RNA/metabolism , Sarcomeres/physiology , Transcription Factors/genetics , p38 Mitogen-Activated Protein Kinases , rhoA GTP-Binding Protein/metabolism
5.
Circulation ; 104(8): 939-44, 2001 Aug 21.
Article in English | MEDLINE | ID: mdl-11514383

ABSTRACT

BACKGROUND: The role of kinins in the cardioprotective effects of ACE inhibitors remains controversial. METHODS AND RESULTS: Right ventricular pressure overload in rabbits was produced by pulmonary artery banding for 21 days. Rabbits were untreated, or they received the ACE inhibitor ramipril with or without bradykinin B(1) and B(2) receptor blockers or the angiotensin (Ang) II type I (AT(1)) receptor blocker losartan. Pulmonary artery banding caused right ventricular hypertrophy, depressed papillary muscle contractility, and loss of Ang II contractile effects because of a signaling defect downstream of AT(1) receptors. Paradoxically, AT(1) receptor density and G protein alpha subunits alphaq and alphai1/2 increased. Inotropic responsiveness to the alpha-receptor agonist phenylephrine was normal. Ramipril preserved cardiac contractility, but this effect was attenuated by simultaneous use of kinin receptor blockers. Ramipril also maintained responsiveness to Ang II and prevented AT(1) receptor and G protein upregulation. The simultaneous use of a kinin receptor blocker attenuated but did not prevent upregulation in the AT(1) receptor and G protein. Losartan had no effect on baseline contractility, but it maintained cardiac inotropic responsiveness to Ang II, prevented upregulation of AT(1) receptors, but did not modify G protein upregulation. CONCLUSIONS: Pressure overload of the right ventricle decreases contractility, uncouples AT(1) receptors to downstream signaling pathways, and changes the expression of components of the AT(1) receptor signaling pathway. Ramipril attenuates these effects via kinins. Interventions that prevent local increases in Ang II or block AT(1) receptors also prevent decreased responsiveness of the AT(1) receptor in this model.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kinins/metabolism , Receptors, Angiotensin/metabolism , Signal Transduction/drug effects , Ventricular Dysfunction, Right/drug therapy , Angiotensin II/metabolism , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Bradykinin Receptor Antagonists , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , GTP-Binding Proteins/metabolism , Hemodynamics/drug effects , In Vitro Techniques , Losartan/pharmacology , Male , Myocardial Contraction/drug effects , Myocardium/metabolism , Myocardium/pathology , Organ Size/drug effects , Papillary Muscles/drug effects , Papillary Muscles/metabolism , Protein Subunits , Pulmonary Artery/physiopathology , Rabbits , Ramipril/pharmacology , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Ventricular Dysfunction, Right/physiopathology
6.
Mol Cell Biochem ; 212(1-2): 99-109, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11108141

ABSTRACT

Angiotensin II (Ang II), the primary effector of the renin-angiotensin system, is a multifunctional hormone that plays an important role in vascular function. In addition to its classical vasoconstrictor action, more recent studies demonstrated that Ang II stimulates the growth of a number of cell types, including vascular smooth muscle cells (SMC) (reviewed in [1-3]). In vivo studies have shown that chronic infusion of Ang II leads to the development of vascular hypertrophy in rats, whereas administration of angiotensin-converting enzyme (ACE) inhibitors or Ang II receptor antagonists prevents or regresses vascular hypertrophy in models of genetic and experimental hypertension [4]. Consistent with in vivo data, several laboratories have shown that Ang II stimulates protein synthesis and induces cellular hypertrophy, but not cell proliferation, in cultured aortic SMC [5-9]. Ang II also induces directed migration (chemotaxis) of vascular SMC [10, 11], although its effect is less prominent than that of platelet-derived growth factor (PDGF). The cellular mechanisms underlying these diverse actions of Ang II are not clearly understood but are likely to involve the activation of distinct signaling pathways.


Subject(s)
Cyclic AMP/physiology , DNA-Binding Proteins/metabolism , Muscle, Smooth, Vascular/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Angiotensin/physiology , Signal Transduction/physiology , Angiotensin II/physiology , Animals , Humans , Muscle, Smooth, Vascular/cytology , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2
7.
Am J Physiol Heart Circ Physiol ; 279(2): H741-51, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10924074

ABSTRACT

Angiotensin II (ANG II) is a multifunctional hormone that exerts potent vasoconstrictor and hypertrophic effects on vascular smooth muscle. Here, we demonstrate that the p38 mitogen-activated protein (MAP) kinase pathway is involved in ANG II-induced vascular contraction. Addition of ANG II to rat aortic smooth muscle cells (SMC) caused a rapid and transient increase of p38 activity through activation of the AT(1) receptor subtype. This response to ANG II was strongly attenuated by pretreating cells with antioxidants and diphenylene iodonium and was mimicked by exposure of cells to H(2)O(2). Stimulation of p38 by ANG II resulted in the enzymatic activation of MAP kinase-activated protein (MAPKAP) kinase-2 and the phosphorylation of heat shock protein 27 (HSP27) in aortic SMC. Pretreatment of cells with the specific p38 MAP kinase inhibitor SB-203580 completely blocked the ANG II-dependent activation of MAPKAP kinase-2 and phosphorylation of HSP27. ANG II also caused a robust activation of MAPKAP kinase-2 in the intact rat aorta. Incubation with SB-203580 significantly decreased the potency of ANG II to induce contraction of rat aortic rings and depressed the maximal hormone response. These results suggest that the p38 MAP kinase pathway selectively modulates the vasoconstrictor action of ANG II in vascular smooth muscle.


Subject(s)
Angiotensin II/pharmacology , Aorta/physiology , Heat-Shock Proteins , Mitogen-Activated Protein Kinases/metabolism , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Signal Transduction/physiology , Animals , Aorta/drug effects , Aorta, Thoracic/physiology , Calcium/physiology , Cells, Cultured , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , HSP27 Heat-Shock Proteins , Imidazoles/pharmacology , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Neoplasm Proteins/metabolism , Phenylephrine/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Rats , Reactive Oxygen Species/physiology , Time Factors , Vasoconstriction/drug effects , p38 Mitogen-Activated Protein Kinases
8.
Oncogene ; 19(31): 3460-9, 2000 Jul 20.
Article in English | MEDLINE | ID: mdl-10918604

ABSTRACT

The interaction between the erbB tyrosine kinase receptors and their ligands plays an important role in tumor growth via the regulation of autocrine and paracrine loops. We report the effect of heregulin beta1, the ligand for erbB-3 and erbB-4 receptors, on the regulation of vascular endothelial growth factor (VEGF) expression, using a panel of breast and lung cancer cell lines with constitutive erbB-2 overexpression or engineered to stably overexpress the erbB-2 receptor. We demonstrate that heregulin beta1 induces VEGF secretion in most cancer cell lines, while no significant effect was observed in normal human mammary and bronchial primary cells. Overexpression of erbB-2 receptor results in induction of the basal level of VEGF and exposure to heregulin further enhances VEGF secretion. This is associated with increased VEGF mRNA expression. In contrast, VEGF induction is significantly decreased in a T47D cell line where erbB-2 is functionally inactivated. Conditioned media from heregulin-treated cancer cells, but not from normal cells, stimulates endothelial cell proliferation; this paracrine stimulation is inhibited by co-exposure to a specific VEGF neutralizing antibody. Furthermore, heregulin-mediated angiogenesis is observed in the in vivo CAM assay. This study reports the first evidence of VEGF regulation by heregulin in cancer cells. Oncogene (2000) 19, 3460 - 3469


Subject(s)
Endothelial Growth Factors/metabolism , ErbB Receptors/physiology , Lymphokines/metabolism , Neoplasm Proteins/physiology , Neovascularization, Pathologic , Neuregulin-1/physiology , Receptor, ErbB-2/physiology , Receptor, ErbB-3/physiology , Adenocarcinoma/pathology , Animals , Breast/cytology , Breast Neoplasms/pathology , Bronchi/cytology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division , Cells, Cultured/drug effects , Chick Embryo , Culture Media, Conditioned/pharmacology , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Genes, erbB-2 , Humans , Lung Neoplasms/pathology , Lymphokines/antagonists & inhibitors , Lymphokines/biosynthesis , Lymphokines/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Neovascularization, Physiologic , Phosphorylation , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptor, ErbB-2/biosynthesis , Receptor, ErbB-3/biosynthesis , Receptor, ErbB-3/genetics , Receptor, ErbB-4 , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Cytogenet Cell Genet ; 88(3-4): 249-52, 2000.
Article in English | MEDLINE | ID: mdl-10828601

ABSTRACT

Activation of the ERK mitogen-activated protein (MAP) kinase pathway has been implicated in the regulation of cell growth, differentiation and senescence. In this pathway, the MAP kinases ERK1/ERK2 are phosphorylated and activated by the dual-specificity kinases MEK1 and MEK2, which in turn are activated by serine phosphorylation by a number of MAP kinase kinase kinases. We report here the chromosomal localization of the human genes encoding the MAP kinase kinase isoforms MEK1 and MEK2. Using a combination of fluorescence in situ hybridization, somatic cell hybrid analysis, DNA sequencing and yeast artificial chromosome (YAC) clone analysis, we have mapped the MEK1 gene (MAP2K1) to chromosome 15q21. We also present evidence for the presence of a MEK1 pseudogene on chromosome 8p21. The MEK2 gene (MAP2K2) was mapped to chromosome 7q32 by fluorescence in situ hybridization and YAC clone analysis.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 7/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Blotting, Southern , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Isoenzymes/genetics , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Molecular Sequence Data , Physical Chromosome Mapping , Pseudogenes/genetics , Sequence Analysis, DNA
10.
Biochem J ; 346 Pt 1: 169-75, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657254

ABSTRACT

MAP (mitogen-activated protein) kinases are a family of serine/threonine kinases that have a pivotal role in signal transduction. Here we report the cloning and characterization of a mouse homologue of extracellular-signal-regulated protein kinase (ERK)3. The mouse Erk3 cDNA encodes a predicted protein of 720 residues, which displays 94% identity with human ERK3. Transcription and translation of this cDNA in vitro generates a 100 kDa protein similar to the human gene product ERK3. Immunoblot analysis with an antibody raised against a unique sequence of ERK3 also recognizes a 100 kDa protein in mouse tissues. A single transcript of Erk3 was detected in every adult mouse tissue examined, with the highest expression being found in the brain. Interestingly, expression of Erk3 mRNA is acutely regulated during mouse development, with a peak of expression observed at embryonic day 11. The mouse Erk3 gene was mapped to a single locus on central mouse chromosome 9, adjacent to the dilute mutation locus and in a region syntenic to human chromosome 15q21. Finally, we provide several lines of evidence to support the existence of a unique Erk3 gene product of 100 kDa in mammalian cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/chemistry , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Mitogen-Activated Protein Kinases , Aging , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Brain/metabolism , Chromosomes/genetics , Cloning, Molecular , Embryo, Mammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genetic Markers/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Mice , Mitogen-Activated Protein Kinase 6 , Molecular Sequence Data , Molecular Weight , Physical Chromosome Mapping , RNA, Messenger/analysis , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid
11.
J Cell Biol ; 148(3): 543-56, 2000 Feb 07.
Article in English | MEDLINE | ID: mdl-10662779

ABSTRACT

Platelet-derived growth factor-BB (PDGF-BB) acts as a full mitogen for cultured aortic smooth muscle cells (SMC), promoting DNA synthesis and cell proliferation. In contrast, angiotensin II (Ang II) induces cellular hypertrophy as a result of increased protein synthesis, but is unable to drive cells into S phase. In an effort to understand the molecular basis for this differential growth response, we have examined the downstream effects of PDGF-BB and Ang II on regulators of the cell cycle machinery in rat aortic SMC. Both PDGF-BB and Ang II were found to stimulate the accumulation of G(1) cyclins with similar kinetics. In addition, little difference was observed in the expression level of their catalytic partners, Cdk4 and Cdk2. However, while both factors increased the enzymatic activity of Cdk4, only PDGF-BB stimulated Cdk2 activity in late G(1) phase. The lack of activation of Cdk2 in Ang II-treated cells was causally related to the failure of Ang II to stimulate phosphorylation of the enzyme on threonine and to downregulate p27(Kip1) expression. By contrast, exposure to PDGF-BB resulted in a progressive and dramatic reduction in the level of p27(Kip1) protein. The time course of p27(Kip1) decline was correlated with a reduced rate of synthesis and an increased rate of degradation of the protein. Importantly, the repression of p27(Kip1) synthesis by PDGF-BB was associated with a marked attenuation of Kip1 gene transcription and a corresponding decrease in Kip1 mRNA accumulation. We also show that the failure of Ang II to promote S phase entry is not related to the autocrine production of transforming growth factor-beta1 by aortic SMC. These results identify p27(Kip1) as an important regulator of the phenotypic response of vascular SMC to mitogenic and hypertrophic stimuli.


Subject(s)
Angiotensin II/pharmacology , CDC2-CDC28 Kinases , Cell Cycle Proteins , Gene Expression Regulation/drug effects , Microtubule-Associated Proteins/genetics , Mitogens/pharmacology , Platelet-Derived Growth Factor/pharmacology , Protein Processing, Post-Translational/drug effects , Tumor Suppressor Proteins , Animals , Aorta/cytology , Becaplermin , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , DNA/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Muscle, Smooth, Vascular/cytology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-sis , RNA, Messenger , Rats , S Phase , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism
12.
J Cardiovasc Pharmacol ; 34(3): 402-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10470999

ABSTRACT

We monitored cardiac angiotensin II concentration and AT1-receptor density after long-term blockade of the renin-angiotensin system in inbred control hamsters treated with placebo or losartan (100 mg/kg/day) and cardiomyopathic hamsters treated with placebo, low-(30 mg/kg/day), or high-dose (100 mg/kg/day) losartan or quinapril (100 mg/kg/day). All treatments were started at age 50 days. Angiotensin II-receptor density and affinity were measured by radioligand-binding assays, and ventricular angiotensin II concentration was determined by radioimmunoassay. After 125 and 275 days of treatment, both doses of losartan significantly reduced AT1-receptor density, whereas quinapril had no effect. The administration of both drugs resulted in significant reductions in ventricular angiotensin II concentration. The prolonged administration of losartan was associated with an increase in cardiac hypertrophy, suggesting that angiotensin II signaling is not directly involved or at least does not play a major role in the remodeling process observed in cardiomyopathic hamsters.


Subject(s)
Angiotensin II/metabolism , Cardiomegaly/metabolism , Receptors, Angiotensin/biosynthesis , Renin-Angiotensin System/physiology , Angiotensin Receptor Antagonists , Animals , Binding, Competitive , Cricetinae , Down-Regulation , Heart Ventricles/metabolism , Losartan/pharmacology , Male , Mesocricetus , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/metabolism
13.
Biochem J ; 341 ( Pt 1): 217-23, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10377265

ABSTRACT

Nck is a small adaptor protein consisting exclusively of three SH3 domains and one SH2 domain. Nck is thought to have an important role in cell signalling by coupling receptor tyrosine kinases, via its SH2 domain, to downstream SH3-binding effectors. We report here that angiotensin II, working through the AT1 receptor subtype, stimulates the phosphorylation of Nck in rat aortic smooth muscle cells. Phosphopeptide mapping analysis revealed that Nck is phosphorylated on four peptides containing exclusively phosphoserine in quiescent cells. Treatment with angiotensin II resulted in increased phosphorylation of these four peptides, without the appearance of new phosphopeptides. We show that Nck, via its SH3 domains, specifically binds three major phosphoproteins of 95, 82 and 66 kDa both in vitro and in intact cells. Notably, the phosphorylation of these Nck-binding proteins was found to increase in parallel with that of Nck on stimulation by angiotensin II. One candidate for the 66 kDa phosphoprotein is the serine/threonine kinase p21-activated kinase 1 (Pak1), which was found to form a stable complex with Nck in aortic smooth muscle cells. We have also identified the gamma2 isoform of casein kinase I as another protein kinase that associates with Nck in these cells. These findings indicate that Nck is a target of G-protein-coupled receptors and suggest a role for Pak1 and casein kinase I-gamma2 in downstream signalling or regulation of the AT1 receptor.


Subject(s)
Angiotensin II/pharmacology , Oncogene Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Angiotensin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Casein Kinases , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Peptide Mapping , Phosphopeptides/isolation & purification , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Signal Transduction , p21-Activated Kinases , src Homology Domains
14.
Infect Immun ; 66(6): 2713-21, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9596738

ABSTRACT

Extracellular signal-regulated protein kinase (ERK, or mitogen-activated protein kinase [MAPK]) regulatory cascades in fungi turn on transcription factors that control developmental processes, stress responses, and cell wall integrity. CEK1 encodes a Candida albicans MAPK homolog (Cek1p), isolated by its ability to interfere with the Saccharomyces cerevisiae MAPK mating pathway. C. albicans cells with a deletion of the CEK1 gene are defective in shifting from a unicellular budding colonial growth mode to an agar-invasive hyphal growth mode when nutrients become limiting on solid medium with mannitol as a carbon source or on glucose when nitrogen is severely limited. The same phenotype is seen in C. albicans mutants in which the homologs (CST20, HST7, and CPH1) of the S. cerevisiae STE20, STE7, and STE12 genes are disrupted. In S. cerevisiae, the products of these genes function as part of a MAPK cascade required for mating and invasiveness of haploid cells and for pseudohyphal development of diploid cells. Epistasis studies revealed that the C. albicans CST20, HST7, CEK1, and CPH1 gene products lie in an equivalent, canonical, MAPK cascade. While Cek1p acts as part of the MAPK cascade involved in starvation-specific hyphal development, it may also play independent roles in C. albicans. In contrast to disruptions of the HST7 and CPH1 genes, disruption of the CEK1 gene adversely affects the growth of serum-induced mycelial colonies and attenuates virulence in a mouse model for systemic candidiasis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Candida albicans/growth & development , Fungal Proteins/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Candida albicans/cytology , Candida albicans/pathogenicity , Candidiasis/mortality , Cell Differentiation , Fungal Proteins/genetics , Gene Dosage , Mice , Models, Biological , Mutation , Protein Kinases/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Suppression, Genetic , Transcription Factors/metabolism , Virulence/genetics
15.
J Cell Physiol ; 174(1): 35-47, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9397154

ABSTRACT

The observation that mitogen-activated protein (MAP) kinases ERK1 and ERK2 are constitutively activated in a number of oncogene-transformed cell lines has led to the hypothesis that prolonged activation of these enzymes is required for the transformation process. To investigate this question, we have examined the regulation of the ERK pathway in Rat1 fibroblasts transformed with activated c-Raf-1 (Raf22W), v-Ha-Ras, and v-Src. Expression of these oncoproteins had no effect on the enzymatic activity of ERK1 and ERK2 in either serum-starved or exponentially growing cells. Moreover, the stimulatory effect of serum on ERK1/ERK2 activity was substantially reduced or abrogated in these cells; this impairment was associated with a strong attenuation of c-fos gene induction. In contrast, expression of Raf22w, v-Ha-Ras, or v-Src resulted in the constitutive activation of the upstream kinases MEK1 and MEK2. Treatment of the cells with vanadate completely restored the activation of ERK1/ERK2 in oncogene-transformed cells, suggesting the involvement of a vanadate-sensitive tyrosine phosphatase. Northern blot analysis of VH1-like dual-specificity MAP kinase phosphatases did not reveal any significant difference in the mRNA expression pattern of these genes between parental and transformed Rat1 cells. Phosphoamino acid analysis indicated that ERK1 is phosphorylated on threonine, but not on tyrosine, in oncogene-transformed cells and that vanadate treatment restores tyrosine phosphorylation. We conclude from these results that ERK1/ERK2 activity is repressed by a single-specificity tyrosine phosphatase in oncogene-transformed rat fibroblasts.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Transformation, Neoplastic , Mitogen-Activated Protein Kinases , Oncogene Proteins/biosynthesis , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Animals , Cell Line, Transformed , Fibroblasts/metabolism , Fibroblasts/pathology , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Oncogene Proteins/genetics , Rats
16.
Mol Biol Cell ; 8(12): 2539-51, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9398674

ABSTRACT

Mitogen-activated protein (MAP) kinases are pivotal components of eukaryotic signaling cascades. Phosphorylation of tyrosine and threonine residues activates MAP kinases, but either dual-specificity or monospecificity phosphatases can inactivate them. The Candida albicans CPP1 gene, a structural member of the VH1 family of dual- specificity phosphatases, was previously cloned by its ability to block the pheromone response MAP kinase cascade in Saccharomyces cerevisiae. Cpp1p inactivated mammalian MAP kinases in vitro and acted as a tyrosine-specific enzyme. In C. albicans a MAP kinase cascade can trigger the transition from the budding yeast form to a more invasive filamentous form. Disruption of the CPP1 gene in C. albicans derepressed the yeast to hyphal transition at ambient temperatures, on solid surfaces. A hyphal growth rate defect under physiological conditions in vitro was also observed and could explain a reduction in virulence associated with reduced fungal burden in the kidneys seen in a systemic mouse model. A hyper-hyphal pathway may thus have some detrimental effects on C. albicans cells. Disruption of the MAP kinase homologue CEK1 suppressed the morphological effects of the CPP1 disruption in C. albicans. The results presented here demonstrate the biological importance of a tyrosine phosphatase in cell-fate decisions and virulence in C. albicans.


Subject(s)
Candida albicans/enzymology , Candida albicans/pathogenicity , Mitogen-Activated Protein Kinase 3 , Mutation/genetics , Protein Tyrosine Phosphatases/metabolism , Spores, Fungal/enzymology , Spores, Fungal/growth & development , Amino Acid Sequence , Animals , Binding Sites , Candida albicans/genetics , Candida albicans/metabolism , Candidiasis/microbiology , Cell Division , Cell Size , Dual-Specificity Phosphatases , Female , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Kidney/microbiology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phenotype , Phosphorylation , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sequence Alignment , Spores, Fungal/genetics , Spores, Fungal/pathogenicity , Temperature , Virulence/genetics
17.
J Biol Chem ; 272(43): 26879-86, 1997 Oct 24.
Article in English | MEDLINE | ID: mdl-9341120

ABSTRACT

In the present study, we have examined the effect of increased cyclic AMP (cAMP) levels on the stimulatory action of angiotensin II (Ang II) on protein synthesis. Treatment with cAMP-elevating agents potently inhibited Ang II-induced protein synthesis in rat aortic smooth muscle cells and in rat fibroblasts expressing the human AT1 receptor. The inhibition was dose-dependent and was observed at all concentrations of the peptide. To explore the mechanism of cAMP action, we have analyzed the effects of forskolin and 3-isobutyl-1-methylxanthine on various receptor-mediated responses. Elevation of cAMP did not alter the binding properties of the AT1 receptor and did not interfere with the activation of phospholipase C or the induction of early growth response genes by Ang II. Likewise, Ang II-dependent activation of the mitogen-activated protein kinases ERK1/ERK2 and p70 S6 kinase was unaffected by cAMP. In contrast, we found that increased concentration of cAMP strongly inhibited the stimulatory effect of Ang II on protein tyrosine phosphorylation. Specifically, cAMP abolished Ang II-induced tyrosine phosphorylation of the focal adhesion-associated protein paxillin and of the tyrosine kinase Tyk2. These results identify a novel mechanism by which the cAMP signaling system may exert growth-inhibitory effects in specific cell types.


Subject(s)
Angiotensin II/pharmacology , Cyclic AMP/metabolism , Mitogen-Activated Protein Kinases , Muscle, Smooth, Vascular/metabolism , Phosphotyrosine , Receptors, Angiotensin/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Aorta , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Cholera Toxin/pharmacology , Colforsin/pharmacology , Fibroblasts , Gene Expression Regulation/drug effects , Humans , Isoproterenol/pharmacology , Kinetics , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Rats , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/biosynthesis , Recombinant Proteins/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , Transfection , Type C Phospholipases/metabolism
18.
Oncogene ; 15(6): 717-25, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9264412

ABSTRACT

Mitogen-activated protein (MAP) kinase phosphatase-1 (MKP-1) is a dual-specificity protein phosphatase encoded by an immediate-early gene responsive to growth factors and stress. The MKP-1 protein selectively inactivates MAP kinases in vitro by dephosphorylation of the regulatory Thr and Tyr residues. Little is known on the mechanisms that regulate MKP-1 gene expression. Here, we demonstrate that Ca2+ is both necessary and sufficient for the induction of MKP-1 gene expression. Treatment of Rat1 fibroblasts with the Ca2+ chelating agent BAPTA completely suppressed serum-induced MKP-1 expression in a dose- and time-dependent manner. The inhibitory effect of BAPTA was observed at the level of the protein and the mRNA. Importantly, Ca2+ chelation blocked the induction of MKP-1 expression in response to all stimuli tested and in different cell types. Increasing the intracellular concentration of Ca2+ with the ionophore A23187 was sufficient to induce MKP-1 mRNA and protein expression in rat fibroblasts. We also provide evidence that activation of MAP kinases is not an absolute requirement for induction of the MKP-1 gene. Exposure of rat fibroblasts to A23187 induced MKP-1 expression without activating the JNK and p38 MAP kinase pathways. Also, inhibition of the ERK pathway with the selective MEK inhibitor PD98059 did not interfere with serum-stimulated MKP-1 mRNA expression. These results will help define the regulatory mechanisms that govern MKP-1 gene transcription in target cells.


Subject(s)
Calcium/physiology , Cell Cycle Proteins , Gene Expression Regulation , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Calcimycin/pharmacology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Dual Specificity Phosphatase 1 , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Flavonoids/pharmacology , Ionophores/pharmacology , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Protein Kinases/metabolism , Protein Phosphatase 1 , Proteins/metabolism , RNA, Messenger/metabolism , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases
19.
Am J Physiol ; 272(6 Pt 1): G1425-32, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9227478

ABSTRACT

Isolated rat hepatocyte couplets were used to study the direct effect of insulin on intracellular Ca2+ homeostasis. Insulin induced a dose-dependent increase in hepatocellular Ca2+ that was gradual, generally monophasic, and reversible. Chelation of extracellular Ca2+ abolished the insulin-induced Ca2+ response, and this suppression was not related to an effect on insulin binding, as indicated by displacement studies. We thus tested the effect of several Ca2+ channel inhibitors on insulin-induced Ca2+ influx. Verapamil at 20 or 200 microM was without effect, whereas 500 microM nickel and 50 microM gadolinium strongly inhibited insulin-induced Ca2+ entry. Finally, we tested whether insulin-induced Ca2+ movements were implicated in the stimulation of mitogen-activated protein kinase (MAPK) activity, which we measured with the use of an immune-complex assay. Verapamil was without effect on the insulin-dependent stimulation of p44mapk activity, whereas addition of ethylene glycol-bis(beta-aminoethyl ether)-N,N,N',N'-tetraacetic acid, nickel, or gadolinium strongly inhibited the effect of the peptide hormone. Our results indicate that insulin triggers Ca2+ influx into hepatocytes, possibly through the opening of channels on the plasma membrane, and that this effect is important for insulin activation of MAPK.


Subject(s)
Calcium/metabolism , Insulin/pharmacology , Liver/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Fluorescent Dyes , Fura-2/analogs & derivatives , Gadolinium/pharmacology , Homeostasis , Kinetics , Liver/drug effects , Male , Nickel/pharmacology , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Spectrometry, Fluorescence , Verapamil/pharmacology
20.
J Biol Chem ; 272(7): 4006-12, 1997 Feb 14.
Article in English | MEDLINE | ID: mdl-9020107

ABSTRACT

To investigate the molecular basis of the hypertrophic action of angiotensin II (AII) in vascular smooth muscle cells (SMC), we have examined the ability of the hormone to regulate the function of the translational repressor 4E-binding protein 1 (4E-BP1). Addition of AII to quiescent aortic SMC potently increased the phosphorylation of 4E-BP1 as revealed by a decreased electrophoretic mobility and an increased phosphate content of the protein. The stimulation of 4E-BP1 phosphorylation was maximal at 15 min and persisted up to 120 min. Results from affinity chromatography on m7GTP-agarose demonstrated that AII-induced phosphorylation of 4E-BP1 promotes its dissociation from eIF4E in target cells. Further characterization of 4E-BP1 phosphorylation by phosphoamino acid analysis and phosphopeptide mapping revealed that 4E-BP1 is phosphorylated on eight distinct peptides containing serine and threonine residues in AII-treated cells. The combination of results obtained from kinetics experiments, phosphopeptide analysis of in vitro and in vivo phosphorylated 4E-BP1, and pharmacological studies with the MAP kinase kinase inhibitor PD 98059 provided strong evidence that the MAP kinases ERK1/ERK2 are not involved in the regulation of 4E-BP1 phosphorylation in aortic SMC. Together, our results demonstrate that AII treatment of vascular SMC leads to hyperphosphorylation of the translational regulator 4E-BP1 and to its dissociation from eIF4E by a MAP kinase-independent mechanism.


Subject(s)
Angiotensin II/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins , Mitogen-Activated Protein Kinases , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Angiotensin II/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Phosphorylation , Protein Binding , Rats , Serine/metabolism , Threonine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...