Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Am J Physiol Heart Circ Physiol ; 307(8): H1216-25, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128174

ABSTRACT

In the first two-thirds of gestation, ovine fetal cardiomyocytes undergo mitosis to increase cardiac mass and accommodate fetal growth. Thereafter, some myocytes continue to proliferate while others mature and terminally differentiate into binucleated cells. At term (145 days gestational age; dGA) about 60% of cardiomyocytes become binucleated and exit the cell cycle under hormonal control. Rising thyroid hormone (T3) levels near term (135 dGA) inhibit proliferation and stimulate maturation. However, the degree to which intracellular signaling patterns change with age in response to T3 is unknown. We hypothesized that in vitro activation of ERK, Akt, and p70(S6K) by two regulators of cardiomyocyte cell cycle activity, T3 and insulin like growth factor-1 (IGF-1), would be similar in cardiomyocytes at gestational ages 100 and 135 dGA. IGF-1 and T3 each independently stimulated phosphorylation of ERK, Akt, and p70(S6K) in cells at both ages. In the younger mononucleated myocytes, the phosphorylation of ERK and Akt was reduced in the presence of IGF-1 and T3. However, the same hormone combination led to a dramatic twofold increase in the phosphorylation of these signaling proteins in the 135 dGA cardiomyocytes-even in cells that were not proliferating. In the older cells, both mono- and binucleated cells were affected. In conclusion, fetal ovine cardiomyocytes undergo profound maturation-related changes in signaling in response to T3 and IGF-1, but not to either factor alone. Differences in age-related response are likely to be related to milestones in fetal cardiac development as the myocardium prepares for ex utero life.


Subject(s)
Fetal Heart/metabolism , MAP Kinase Signaling System , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Heart/cytology , Fetal Heart/drug effects , Insulin-Like Growth Factor I/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sheep , Thyroid Hormones/pharmacology
2.
J Physiol ; 548(Pt 3): 881-91, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12626668

ABSTRACT

Rat and sheep cardiac myocytes become binucleate as they complete the 'terminal differentiation' process soon after birth and are not able to divide thereafter. Angiotensin II (Ang II) is known to stimulate hypertrophic changes in rodent cardiomyocytes under both in vivo and in vitro conditions via the AT1 receptor and intracellular extracellular regulated kinase (ERK) signalling cascade. We sought to develop culture methods for immature sheep cardiomyocytes in order to test the hypothesis that Ang II is a hypertrophic agent in the immature myocardium of the sheep. We isolated fetal sheep cardiomyocytes and cultured them for 96 h, added Ang II and phenylephrine (PE) for 48 h, and measured footprint area and proliferation (5-bromo-2'-deoxyuridine (BrdU) uptake) separately in mono- vs. binucleate myocytes. We found that neither Ang II nor PE changed the footprint area of mononucleated cells. PE stimulated an increase in footprint area of binucleate cells but Ang II did not. Ang II increased myocyte BrdU uptake compared to serum free conditions, but PE did not affect BrdU uptake. The MAP kinase kinase (MEK) inhibitor UO126 prevented BrdU uptake in Ang II-stimulated cells and prevented cell hypertrophy in PE-stimulated cells. This paper establishes culture methods for immature sheep cardiomyocytes and reports that: (1) Ang II is not a hypertrophic agent; (2) Ang II stimulates hyperplastic growth among mononucleate myocytes; (3) PE is a hypertrophic agent in binucleate myocytes; and (4) the ERK cascade is required for the proliferation effect of Ang II and the hypertrophic effect of PE.


Subject(s)
Angiotensin II/pharmacology , Heart/physiology , Myocardium/pathology , Ventricular Function , Animals , Bromodeoxyuridine/pharmacokinetics , Cell Culture Techniques/methods , Cells, Cultured , Female , Fetus , Gestational Age , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/pathology , Hyperplasia , Hypertrophy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pregnancy , Sheep
3.
Science ; 296(5573): 1648-9, 2002 May 31.
Article in English | MEDLINE | ID: mdl-12040181

ABSTRACT

A key issue in signal transduction is how signaling pathways common to many systems-so-called canonical signaling cassettes-integrate signals from molecules having a wide spectrum of activities, such as hormones and neurotrophins, to deliver distinct biological outcomes. The neuroendocrine cell line PC12, derived from rat pheochromocytoma, provides an example of how one canonical signaling cassette-the Raf --> mitogen-activated protein kinase kinase (MEK) --> extracellular signal-regulated kinase (ERK) pathway-can promote distinct outcomes, which in this case include neuritogenesis, gene induction, and proliferation. Two growth hormones, epidermal growth factor (EGF) and nerve growth factor (NGF), use the same pathway to cause PC12 proliferation and differentiation, respectively. In addition, pituitary adenylate cyclase-activating polypeptide (PACAP), a neurotransmitter that also causes differentiation, uses the same canonical cassette as NGF but in a different way. The Connections Map for PC12 Cell Differentiation brings into focus the complex array of specific cellular responses that rely on canonical signal transduction systems.


Subject(s)
Cell Differentiation , MAP Kinase Signaling System , PC12 Cells/physiology , Animals , Cell Division , Cyclic AMP/metabolism , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Neurites/physiology , Neuropeptides/metabolism , Neuropeptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Rats , Receptor, trkA/metabolism , Receptors, Cell Surface/metabolism , Response Elements , Transcription, Genetic
4.
J Biol Chem ; 276(33): 31238-46, 2001 Aug 17.
Article in English | MEDLINE | ID: mdl-11390399

ABSTRACT

Insulin-like growth factor-I (IGF-I) is essential for somatic growth and promotes bone cell replication and differentiation. IGF-I production by rat osteoblasts is stimulated by activation of cAMP-dependent protein kinase (PKA). In this report, we define two interacting PKA-regulated pathways that control IGF-I gene transcription in cultured human osteoblasts. Stimulation of cAMP led to a 12-fold increase in IGF-I mRNA and enhanced IGF-I promoter activity through a DNA response element termed HS3D and the transcription factor CCAAT/enhancer-binding protein delta (C/EBPdelta). Under basal conditions, C/EBPdelta was found in osteoblast nuclei but was transcriptionally silent. Treatment with the PKA inhibitor H-89 caused redistribution of C/EBPdelta to the cytoplasm. After hormone treatment, the catalytic subunit of PKA accumulated in osteoblast nuclei. Inhibition of active PKA with targeted nuclear expression of PKA inhibitor had no effect on the subcellular location of C/EBPdelta but prevented hormone-induced IGF-I gene activation, while cytoplasmic PKA inhibitor additionally caused the removal of C/EBPdelta from the nucleus. These results show that IGF-I gene expression is controlled in human osteoblasts by two PKA-dependent pathways. Cytoplasmic PKA mediates nuclear localization of C/EBPdelta under basal conditions, and nuclear PKA stimulates its transcriptional activity upon hormone treatment. Both mechanisms are indirect, since PKA failed to phosphorylate human C/EBPdelta in vitro.


Subject(s)
CCAAT-Enhancer-Binding Proteins/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , Insulin-Like Growth Factor I/genetics , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Transcription Factors , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Protein-delta , Colforsin/pharmacology , Dinoprostone/pharmacology , Humans , Rats
5.
Mol Cell Biol ; 21(11): 3671-83, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11340161

ABSTRACT

In many normal and transformed cell types, the intracellular second messenger cyclic AMP (cAMP) blocks the effects of growth factors and serum on mitogenesis, proliferation, and cell cycle progression. cAMP exerts these growth-inhibitory effects via inhibition of the mitogen-activated protein (MAP) kinase cascade. Here, using Hek293 and NIH 3T3 cells, we show that cAMP's inhibition of the MAP kinase cascade is mediated by the small G protein Rap1. Activation of Rap1 by cAMP induces the association of Rap1 with Raf-1 and limits Ras-dependent activation of ERK. In NIH 3T3 cells, Rap1 is required not only for cAMP's inhibition of ERK activation but for inhibition of cell proliferation and mitogenesis as well.


Subject(s)
Cyclic AMP/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinases/antagonists & inhibitors , rap1 GTP-Binding Proteins/metabolism , 3T3 Cells , Animals , Cell Division , Cell Line, Transformed , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism
6.
J Physiol Paris ; 94(3-4): 239-50, 2000.
Article in English | MEDLINE | ID: mdl-11088001

ABSTRACT

The mitogen activated protein (MAP) kinase cascade represents one of the major regulator of cell growth by hormones and growth factors. However, although the activation of this intracellular pathway has been often regarded as mediator of cell proliferation, in many cell types the increase in MAP kinase (also called extra-cellular signal regulated kinase: ERK) activity may result in cell growth arrest, depending on the length or the intensity of the stimulation. In this review we examine recent data concerning the effects of somatostatin on the MAP kinase cascade through one of its major receptor subtype, the somatostatin receptor 1 (SSTR1), stably expressed in CHO-K1 cells. Somatostatin inhibits the proliferative effects of basic FGF (bFGF) in CHO-SSTR1 cell line. However, in these cells, somatostatin robustly activates the MAP kinase and augments bFGF-induced stimulation of ERK. We show that the activation of ERK via SSTR1 is mediated by the betagamma subunit of a pertussis toxin-sensitive G-protein and requires both the small G protein Ras and the serine/threonine kinase Raf-1. Moreover the phosphatidyl inositol-3kinase and the cytosolic tyrosine kinase c-src participate in the signal transduction regulated by SSTRI to activate ERK, as well as it is involved the protein tyrosine phosphatase (PTP) SHP-2. Previous studies have suggested that somatostatin-stimulated PTP activity mediates the growth inhibitory actions of somatostatin, in CHO-SSTR1 cells. Thus, the activation of SHP-2 by SSTR1 may mediate the antiproliferative activity of somatostatin. SHP-2 may. in turn, regulate the activity of kinases upstream of ERK that require tyrosine dephosphorylation to be activated, such as c-src. Finally, the synergism between somatostatin and bFGF in the activation of ERK results in an increased expression of the cyclin-dependent kinase inhibitor p21cip/WAF1 as molecular effector of the antiproliferative activity of somatostatin.


Subject(s)
MAP Kinase Signaling System/physiology , Protein Tyrosine Phosphatases/metabolism , Receptors, Somatostatin/metabolism , Animals , CHO Cells , Cell Division/physiology , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Intracellular Signaling Peptides and Proteins , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6
7.
Mol Cell Biol ; 20(21): 8069-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11027277

ABSTRACT

Neurotrophins promote multiple actions on neuronal cells including cell survival and differentiation. The best-studied neurotrophin, nerve growth factor (NGF), is a major survival factor in sympathetic and sensory neurons and promotes differentiation in a well-studied model system, PC12 cells. To mediate these actions, NGF binds to the TrkA receptor to trigger intracellular signaling cascades. Two kinases whose activities mediate these processes include the mitogen-activated protein (MAP) kinase (or extracellular signal-regulated kinase [ERK]) and phosphoinositide 3-kinase (PI3-K). To examine potential interactions between the ERK and PI3-K pathways, we studied the requirement of PI3-K for NGF activation of the ERK signaling cascade in dorsal root ganglion cells and PC12 cells. We show that PI3-K is required for TrkA internalization and participates in NGF signaling to ERKs via distinct actions on the small G proteins Ras and Rap1. In PC12 cells, NGF activates Ras and Rap1 to elicit the rapid and sustained activation of ERKs respectively. We show here that Rap1 activation requires both TrkA internalization and PI3-K, whereas Ras activation requires neither TrkA internalization nor PI3-K. Both inhibitors of PI3-K and inhibitors of endocytosis prevent GTP loading of Rap1 and block sustained ERK activation by NGF. PI3-K and endocytosis may also regulate ERK signaling at a second site downstream of Ras, since both rapid ERK activation and the Ras-dependent activation of the MAP kinase kinase kinase B-Raf are blocked by inhibition of either PI3-K or endocytosis. The results of this study suggest that PI3-K may be required for the signals initiated by TrkA internalization and demonstrate that specific endocytic events may distinguish ERK signaling via Rap1 and Ras.


Subject(s)
Endocytosis , Nerve Growth Factor/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Blotting, Western , Cell Differentiation , Cell Survival , Cells, Cultured , Chromatography, Affinity , Chromones/pharmacology , Colforsin/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Ganglia, Spinal/metabolism , Immunohistochemistry , Luciferases/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Morpholines/pharmacology , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Receptor, trkA/metabolism , Time Factors , Transfection
8.
Mol Cell Biol ; 20(22): 8409-19, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11046138

ABSTRACT

Proliferation of T cells via activation of the T-cell receptor (TCR) requires concurrent engagement of accessory costimulatory molecules to achieve full activation. The best-studied costimulatory molecule, CD28, achieves these effects, in part, by augmenting signals from the TCR to the mitogen-activated protein (MAP) kinase cascade. We show here that TCR-mediated stimulation of MAP kinase extracellular-signal-regulated kinases (ERKs) is limited by activation of the Ras antagonist Rap1. CD28 increases ERK signaling by blocking Rap1 action. CD28 inhibits Rap1 activation because it selectively stimulates an extrinsic Rap1 GTPase activity. The ability of CD28 to stimulate Rap1 GTPase activity was dependent on the tyrosine kinase Lck. Our results suggest that CD28-mediated Rap1 GTPase-activating protein activation can help explain the augmentation of ERKs during CD28 costimulation.


Subject(s)
CD28 Antigens/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , T-Lymphocytes/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , CD3 Complex/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/metabolism , ras Proteins/genetics , ras Proteins/metabolism , src Homology Domains
9.
J Biol Chem ; 275(44): 34433-41, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10950954

ABSTRACT

Two major intracellular signals that regulate neuronal function are calcium and cAMP. In many cases, the actions of these two second messengers involve long term changes in gene expression. One well studied target of both calcium and cAMP signaling is the transcription factor cAMP-responsive element-binding protein (CREB). Multiple signaling pathways have been shown to contribute to the regulation of CREB-dependent transcription, including both protein kinase A (PKA)- and mitogen-activated protein (MAP) kinase/extracellular signal-regulated kinase (ERK)-dependent kinase cascades. We have previously described a mechanism by which cAMP and calcium influx may stimulate ERKs in neuronal cells. This pathway involves the PKA-dependent activation of the Ras-related small G-protein, Rap1, and subsequent stimulation of the neuronal Raf isoform, B-Raf. In this study, we examined the contribution of the Rap1-ERK pathway to the control of gene transcription by calcium influx and cAMP. Using the PC12 cell model system, we found that both calcium influx and cAMP stimulated CREB-dependent transcription via a Rap1-ERK pathway, but this regulation occurred through distinct mechanisms. Calcium-mediated phosphorylation of CREB through the PKA-Rap1-ERK pathway. In contrast, cAMP phosphorylated CREB via PKA directly but required a Rap1-ERK pathway to activate a component downstream of CREB phosphorylation and CREB-binding protein recruitment. These data suggest that the Rap1/B-Raf signaling pathway may have an important role in the regulation of CREB-dependent gene expression.


Subject(s)
Calcium/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , 3T3 Cells , Animals , Cell Line , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Ion Transport , Mice , PC12 Cells , Phosphorylation , Rats , Transcription, Genetic/physiology
10.
J Biol Chem ; 275(33): 25342-50, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10840035

ABSTRACT

G protein-coupled receptors can induce cellular proliferation by stimulating the mitogen-activated protein (MAP) kinase cascade. Heterotrimeric G proteins are composed of both alpha and betagamma subunits that can signal independently to diverse intracellular signaling pathways including those that activate MAP kinases. In this study, we examined the ability of isoproterenol, an agonist of the beta(2)-adrenergic receptor (beta(2)AR), to stimulate extracellular signal-regulated kinases (ERKs). Using HEK293 cells, which express endogenous beta(2)AR, we show that isoproterenol stimulates ERKs via beta(2)AR. This action of isoproterenol requires cAMP-dependent protein kinase and is insensitive to pertussis toxin, suggesting that Galpha(s) activation of cAMP-dependent protein kinase is required. Interestingly, beta(2)AR activates both the small G proteins Rap1 and Ras, but only Rap1 is capable of coupling to Raf isoforms. beta(2)AR inhibits the Ras-dependent activation of both Raf isoforms Raf-1 and B-Raf, whereas Rap1 activation by isoproterenol recruits and activates B-Raf. beta(2)AR activation of ERKs is not blocked by expression of RasN17, an interfering mutant of Ras, but is blocked by expression of either RapN17 or Rap1GAP1, both of which interfere with Rap1 signaling. We propose that isoproterenol can activate ERKs via Rap1 and B-Raf in these cells.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Adrenergic, beta-2/metabolism , rap1 GTP-Binding Proteins/metabolism , Adrenergic beta-Agonists/pharmacology , Blotting, Western , Cell Line , Chromatography, Affinity , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Escherichia coli/metabolism , Glutathione Transferase/metabolism , Humans , Isoproterenol/pharmacology , Nickel/metabolism , Pertussis Toxin , Phosphorylation , Plasmids/metabolism , Protein Isoforms , Recombinant Fusion Proteins/metabolism , Time Factors , Transfection , Virulence Factors, Bordetella/pharmacology , ras Proteins/metabolism
11.
J Biol Chem ; 275(5): 3722-8, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10652372

ABSTRACT

Activity-dependent regulation of neuronal events such as cell survival and synaptic plasticity is controlled by increases in neuronal calcium levels. These actions often involve stimulation of intracellular kinase signaling pathways. For example, the mitogen-activated protein kinase, or extracellular signal-regulated kinase (ERK), signaling cascade has increasingly been shown to be important for the induction of gene expression and long term potentiation. However, the mechanisms leading to ERK activation by neuronal calcium are still unclear. In the present study, we describe a protein kinase A (PKA)-dependent signaling pathway that may link neuronal calcium influx to ERKs via the small G-protein, Rap1, and the neuronal Raf isoform, B-Raf. Thus, in PC12 cells, depolarization-mediated calcium influx led to the activation of B-Raf, but not Raf-1, via PKA. Furthermore, depolarization also induced the PKA-dependent stimulation of Rap1 and led to the formation of a Rap1/B-Raf signaling complex. In contrast, depolarization did not lead to the association of Ras with B-Raf. The major action of PKA-dependent Rap1/B-Raf signaling in neuronal cells is the activation of ERKs. Thus, we further show that, in both PC12 cells and hippocampal neurons, depolarization-induced calcium influx stimulates ERK activity in a PKA-dependent manner. Given the fact that both Rap1 and B-Raf are highly expressed in the central nervous system, we suggest that this signaling pathway may regulate a number of activity-dependent neuronal functions.


Subject(s)
Calcium/metabolism , Neurons/physiology , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , rap1 GTP-Binding Proteins/metabolism , Animals , MAP Kinase Signaling System , Membrane Potentials , PC12 Cells , Rats
12.
Curr Opin Neurobiol ; 9(5): 544-53, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508738

ABSTRACT

Extracellular-signal-regulated kinases (ERKs) are emerging as important regulators of neuronal function. Recent advances have increased our understanding of ERK signalling at the molecular level. In particular, it has become evident that multiple second messengers, such as cyclic adenosine monophosphate, protein kinase A, calcium, and diacylglycerol, can control ERK signalling via the small G proteins Ras and Rap1. These findings may explain the role of ERKs in the regulation of activity-dependent neuronal events, such as synaptic plasticity, long-term potentiation and cell survival. Moreover, they allow us to begin to develop a model to understand both the control of ERKs at the subcellular level and the generation of ERK signal specificity.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Neurons/physiology , Signal Transduction/physiology , Animals , GTP-Binding Proteins/physiology , Learning/physiology , Memory/physiology , Neuronal Plasticity/physiology , Second Messenger Systems/physiology , Synapses/physiology
13.
J Biol Chem ; 274(35): 24858-64, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10455158

ABSTRACT

Atrial natriuretic factor (ANF) inhibits proliferation in non-myocardial cells and is thought to be anti-hypertrophic in cardiomyocytes. We investigated the possibility that the anti-hypertrophic actions of ANF involved the mitogen-activated protein kinase signal transduction cascade. Cultured neonatal rat ventricular myocytes treated for 48 h with the alpha(1)-adrenergic agonist phenylephrine (PE) had an 80% increase in cross-sectional area (CSA). ANF alone had no effect but inhibited PE-induced increases in CSA by approximately 50%. The mitogen-activated protein kinase/ERK kinase (MEK) inhibitor PD098059 minimally inhibited PE-induced increases in CSA, but it completely abolished ANF-induced inhibition of PE-induced increases. ANF-induced extracellular signal-regulated protein kinase (ERK) nuclear translocation was also eliminated by PD098059. ANF treatment caused MEK phosphorylation and activation but failed to activate any of the Raf isoforms. ANF induced a rapid increase in ERK phosphorylation and in vitro kinase activity. PE also increased ERK activity, and the combined effect of ANF and PE appeared to be additive. ANF-induced ERK phosphorylation was eliminated by PD098059. ANF induced minimal phosphorylation of JNK or p38, indicating that its effect on ERK was specific. ANF-induced activation of ERK was mimicked by cGMP analogs, suggesting that ANF-induced ERK activation involves the guanylyl cyclase activity of the ANF receptor. These data suggest that there is an important linkage between cGMP signaling and the mitogen-activated protein kinase cascade and that selective ANF activation of ERK is required for the anti-hypertrophic action of ANF. Thus, ANF expression might function as the natural defense of the heart against maladaptive hypertrophy through its ability to activate ERK.


Subject(s)
Atrial Natriuretic Factor/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cells, Cultured , Cyclic GMP/metabolism , Enzyme Activation/drug effects , Fluorescent Antibody Technique , Mitogen-Activated Protein Kinase Kinases , Myocardium , Phenylephrine/antagonists & inhibitors , Phenylephrine/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 274(31): 21507-10, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419452

ABSTRACT

We used the yeast two-hybrid system to identify proteins that interact directly with Galpha(o). Mutant-activated Galpha(o) was used as the bait to screen a cDNA library from chick dorsal root ganglion neurons. We found that Galpha(o) interacted with several proteins including Gz-GTPase-activating protein (Gz-GAP), a new RGS protein (RGS-17), a novel protein of unknown function (IP6), and Rap1GAP. This study focuses on Rap1GAP, which selectively interacts with Galpha(o) and Galpha(i) but not with Galpha(s) or Galpha(q). Rap1GAP interacts more avidly with the unactivated Galpha(o) as compared with the mutant (Q205L)-activated Galpha(o). When expressed in HEK-293 cells, unactivated Galpha(o) co-immunoprecipitates with the Rap1GAP. Expression of chick Rap1GAP in PC-12 cells inhibited activation of Rap1 by forskolin. When unactivated Galpha(o) was expressed, the amount of activated Rap1 was greatly increased. This effect was not observed with the Q205L-Galpha(o). Expression of unactivated Galpha(o) stimulated MAP-kinase (MAPK1/2) activity in a Rap1GAP-dependent manner. These results identify a novel function of Galpha(o), which in its resting state can sequester Rap1GAP thereby regulating Rap1 activity and consequently gating signal flow from Rap1 to MAPK1/2. Thus, activation of G(o) could modulate the Rap1 effects on a variety of cellular functions.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , GTP-Binding Proteins/metabolism , Ganglia, Spinal/metabolism , Neurons/metabolism , Proteins/metabolism , Amino Acid Substitution , Animals , Cell Line , Chickens , GTPase-Activating Proteins , Gene Library , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , PC12 Cells , Rats , Recombinant Proteins/metabolism , Transfection
15.
J Biol Chem ; 274(18): 12890-7, 1999 Apr 30.
Article in English | MEDLINE | ID: mdl-10212278

ABSTRACT

Pathophysiological hypoxia is an important modulator of gene expression in solid tumors and other pathologic conditions. We observed that transcriptional activation of the c-jun proto-oncogene in hypoxic tumor cells correlates with phosphorylation of the ATF2 transcription factor. This finding suggested that hypoxic signals transmitted to c-jun involve protein kinases that target AP-1 complexes (c-Jun and ATF2) that bind to its promoter region. Stress-inducible protein kinases capable of activating c-jun expression include stress-activated protein kinase/c-Jun N-terminal protein kinase (SAPK/JNK) and p38 members of the mitogen-activated protein kinase (MAPK) superfamily of signaling molecules. To investigate the potential role of MAPKs in the regulation of c-jun by tumor hypoxia, we focused on the activation SAPK/JNKs in SiHa human squamous carcinoma cells. Here, we describe the transient activation of SAPK/JNKs by tumor-like hypoxia, and the concurrent transcriptional activation of MKP-1, a stress-inducible member of the MAPK phosphatase (MKP) family of dual specificity protein-tyrosine phosphatases. MKP-1 antagonizes SAPK/JNK activation in response to diverse environmental stresses. Together, these findings identify MKP-1 as a hypoxia-responsive gene and suggest a critical role in the regulation of SAPK/JNK activity in the tumor microenvironment.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Cycle Proteins , Cell Hypoxia , Immediate-Early Proteins/metabolism , Mitogen-Activated Protein Kinases , Oxygen/metabolism , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Activating Transcription Factor 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dual Specificity Phosphatase 1 , Enzyme Activation , Humans , Immediate-Early Proteins/genetics , JNK Mitogen-Activated Protein Kinases , Phosphorylation , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Tumor Cells, Cultured
16.
Mol Endocrinol ; 13(1): 24-37, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9892010

ABSTRACT

Hormones and growth factors regulate cell growth via the mitogen-activated protein (MAP) kinase cascade. Here we examine the actions of the hormone somatostatin on the MAP kinase cascade through one of its two major receptor subtypes, the somatostatin receptor 1 (SSTR1) stably expressed in CHO-K1 cells. Somatostatin antagonizes the proliferative effects of fibroblast growth factor in CHO-SSTR1 cells via the SSTR1 receptor. However, in these cells, somatostatin robustly activates MAP kinase (also called extracellular signal regulated kinase; ERK) and augments fibroblast growth factor-stimulated ERK activity. We show that the activation of ERK via SSTR1 is pertussis toxin sensitive and requires the small G protein Ras, phosphatidylinositol 3-kinase, the serine/threonine kinase Raf-1, and the protein tyrosine phosphatase SHP-2. The activation of ERK by SSTR1 increased the expression of the cyclin-dependent protein kinase inhibitor p21(cip1/WAF1). Previous studies have suggested that somatostatin-stimulated protein tyrosine phosphatase activity mediates the growth effects of somatostatin. Our data suggest that SHP-2 stimulation by SSTR1 may mediate some of these effects through the activation of the MAP kinase cascade and the expression of p21(cip1/WAF1).


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mitogen-Activated Protein Kinases , Receptors, Somatostatin/metabolism , Somatostatin/metabolism , Animals , CHO Cells/drug effects , CHO Cells/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/drug effects , Cell Division , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/metabolism , Enzyme Activation/drug effects , Fibroblast Growth Factor 2/pharmacology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinase 3 , Pertussis Toxin , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Somatostatin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Somatostatin/pharmacology , Virulence Factors, Bordetella/pharmacology , ras Proteins/genetics , ras Proteins/metabolism , src Homology Domains
17.
Cancer Res ; 59(1): 213-8, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9892209

ABSTRACT

Prostate cells are simultaneously exposed to a variety of peptide growth factors and neuropeptides that elevate cAMP. Both the growth factors and cAMP have large effects on the growth, differentiation, and movement of many cell types. Because mitogen-activated protein kinase (MAPK) is central to these effects, we analyzed the ways in which these agonists interact in regulating MAPK in prostate cancer cells. We show that, in LNCaP prostate cancer cells, elevation of intracellular cAMP can potentiate the ability of epidermal growth factor (EGF), interleukin 6, and serum to activate MAPK and that this potentiation depends on protein kinase A and Rap1. The response to cAMP is different in the androgen-independent prostate cancer cell line PC-3, where elevation of cAMP slightly inhibits MAPK activation by EGF. We also show that treatment of LNCaP with the calcium ionophore A23187 or the phorbol ester phorbol 12-myristate 13-acetate activates MAPK, but the activation of MAPK by these agonists is inhibited rather than potentiated by increasing cAMP. Finally, we show that phorbol 12-myristate 13-acetate and interleukin 6 can potentiate the signaling activity of EGF. We conclude that neuroendocrine factors that elevate cAMP sensitize LNCaP prostate cancer cells to signaling by peptide growth factors and that low levels of mixtures of growth factors can activate intracellular signaling to a greater degree than would be predicted from the activity of the individual agonists.


Subject(s)
Blood Proteins/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Epidermal Growth Factor/pharmacology , Interleukin-6/pharmacology , Prostatic Neoplasms/metabolism , Enzyme Activation/drug effects , Humans , Male , Signal Transduction/drug effects , Tumor Cells, Cultured
18.
J Biol Chem ; 273(14): 8240-7, 1998 Apr 03.
Article in English | MEDLINE | ID: mdl-9525930

ABSTRACT

Induction of neuronal differentiation of the rat pheochromocytoma cell line, PC12 cells, by nerve growth factor (NGF) requires activation of the mitogen-activated protein (MAP) kinase or extracellular signal-regulated kinase (ERK). cAMP-dependent protein kinase (protein kinase A (PKA)) also can induce differentiation of these cells. Like NGF, the ability of PKA to differentiate PC12 cells is associated with a sustained activation of ERKs. Here we show that maximal sustained activation of ERK1 by NGF requires PKA. Inhibitors of PKA partially blocked activation of ERK1 by NGF but had no effect on activation of ERK1 by EGF. Inhibition of PKA also reduced the ability of NGF and cAMP, but not EGF, to activate the transcription factor Elk-1, reduced the induction of both immediate early and late genes after NGF treatment, and blocked the nuclear translocation of ERK1 induced by NGF. We propose that PKA is an important contributor to the activation of ERK1 by NGF and is required for maximal induction of gene expression by NGF.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation/drug effects , Nerve Growth Factors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Animals , Biological Transport/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases , PC12 Cells , Protein Serine-Threonine Kinases/genetics , Rats
19.
Nature ; 392(6676): 622-6, 1998 Apr 09.
Article in English | MEDLINE | ID: mdl-9560161

ABSTRACT

Activation of mitogen-activated protein (MAP) kinase (also known as extracellular-signal-regulated kinase, or ERK) by growth factors can trigger either cell growth or differentiation. The intracellular signals that couple growth factors to MAP kinase may determine the different effects of growth factors: for example, transient activation of MAP kinase by epidermal growth factor stimulates proliferation of PC12 cells, whereas they differentiate in response to nerve growth factor, which acts partly by inducing a sustained activation of MAP kinase. Here we show that activation of MAP kinase by nerve growth factor involves two distinct pathways: the initial activation of MAP kinase requires the small G protein Ras, but its activation is sustained by the small G protein Rap1. Rap1 is activated by CRK adaptor proteins and the guanine-nucleotide-exchange factor C3G, and forms a stable complex with B-Raf, an activator of MAP kinase. Rap1 is required for at least two indices of neuronal differentiation by nerve growth factor: electrical excitability and the induction of neuron-specific genes. We propose that the activation of Rap1 by C3G represents a common mechanism to induce sustained activation of the MAP kinase cascade in cells that express B-Raf.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , DNA-Binding Proteins , GTP-Binding Proteins/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Nerve Growth Factors/metabolism , Transcription Factors , 3T3 Cells , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Differentiation , Enzyme Activation , Epidermal Growth Factor/metabolism , GTP-Binding Proteins/genetics , Genes, Reporter , Guanine Nucleotide Exchange Factors , Guanosine Triphosphate/metabolism , MAP Kinase Kinase 4 , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Mitogen-Activated Protein Kinase 1 , Mutation , Neurites/metabolism , PC12 Cells , Protein Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-crk , Rats , Transfection , ets-Domain Protein Elk-1 , rap GTP-Binding Proteins , ras Guanine Nucleotide Exchange Factors , ras Proteins/metabolism
20.
Cell ; 89(1): 73-82, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9094716

ABSTRACT

Cyclic adenosine monophosphate (cAMP) has tissue-specific effects on growth, differentiation, and gene expression. We show here that cAMP can activate the transcription factor Elk-1 and induce neuronal differentiation of PC12 cells via its activation of the MAP kinase cascade. These cell type-specific actions of cAMP require the expression of the serine/threonine kinase B-Raf and activation of the small G protein Rap1. Rap1, activated by mutation or by the cAMP-dependent protein kinase PKA, is a selective activator of B-Raf and an inhibitor of Raf-1. Therefore, in B-Raf-expressing cells, the activation of Rap1 provides a mechanism for tissue-specific regulation of cell growth and differentiation via MAP kinase.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , DNA-Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Protozoan Proteins/metabolism , Animals , Cattle , Cell Differentiation/physiology , Cell Membrane/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Guanosine Triphosphate/metabolism , Neurons/cytology , Neurons/enzymology , PC12 Cells/cytology , PC12 Cells/drug effects , PC12 Cells/enzymology , Proto-Oncogene Proteins c-raf , Rats , Transcription Factors/metabolism , ets-Domain Protein Elk-1
SELECTION OF CITATIONS
SEARCH DETAIL
...