Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 153 Suppl 1: S137-53, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18204489

RESUMEN

Glycogen synthase kinase 3 (GSK3, of which there are two isoforms, GSK3alpha and GSK3beta) was originally characterized in the context of regulation of glycogen metabolism, though it is now known to regulate many other cellular processes. Phosphorylation of GSK3alpha(Ser21) and GSK3beta(Ser9) inhibits their activity. In the heart, emphasis has been placed particularly on GSK3beta, rather than GSK3alpha. Importantly, catalytically-active GSK3 generally restrains gene expression and, in the heart, catalytically-active GSK3 has been implicated in anti-hypertrophic signalling. Inhibition of GSK3 results in changes in the activities of transcription and translation factors in the heart and promotes hypertrophic responses, and it is generally assumed that signal transduction from hypertrophic stimuli to GSK3 passes primarily through protein kinase B/Akt (PKB/Akt). However, recent data suggest that the situation is far more complex. We review evidence pertaining to the role of GSK3 in the myocardium and discuss effects of genetic manipulation of GSK3 activity in vivo. We also discuss the signalling pathways potentially regulating GSK3 activity and propose that, depending on the stimulus, phosphorylation of GSK3 is independent of PKB/Akt. Potential GSK3 substrates studied in relation to myocardial hypertrophy include nuclear factors of activated T cells, beta-catenin, GATA4, myocardin, CREB, and eukaryotic initiation factor 2Bvarepsilon. These and other transcription factor substrates putatively important in the heart are considered. We discuss whether cardiac pathologies could be treated by therapeutic intervention at the GSK3 level but conclude that any intervention would be premature without greater understanding of the precise role of GSK3 in cardiac processes.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Cardiomegalia/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Miocardio/enzimología , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/fisiología , Cardiomegalia/patología , Inhibidores Enzimáticos/uso terapéutico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Histona Desacetilasas/metabolismo , Humanos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología
5.
Mol Cell Biol ; 21(4): 1173-84, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11158304

RESUMEN

Small guanine nucleotide-binding proteins of the Ras and Rho (Rac, Cdc42, and Rho) families have been implicated in cardiac myocyte hypertrophy, and this may involve the extracellular signal-related kinase (ERK), c-Jun N-terminal kinase (JNK), and/or p38 mitogen-activated protein kinase (MAPK) cascades. In other systems, Rac and Cdc42 have been particularly implicated in the activation of JNKs and p38-MAPKs. We examined the activation of Rho family small G proteins and the regulation of MAPKs through Rac1 in cardiac myocytes. Endothelin 1 and phenylephrine (both hypertrophic agonists) induced rapid activation of endogenous Rac1, and endothelin 1 also promoted significant activation of RhoA. Toxin B (which inactivates Rho family proteins) attenuated the activation of JNKs by hyperosmotic shock or endothelin 1 but had no effect on p38-MAPK activation. Toxin B also inhibited the activation of the ERK cascade by these stimuli. In transfection experiments, dominant-negative N17Rac1 inhibited activation of ERK by endothelin 1, whereas activated V12Rac1 cooperated with c-Raf to activate ERK. Rac1 may stimulate the ERK cascade either by promoting the phosphorylation of c-Raf or by increasing MEK1 and/or -2 association with c-Raf to facilitate MEK1 and/or -2 activation. In cardiac myocytes, toxin B attenuated c-Raf(Ser-338) phosphorylation (50 to 70% inhibition), but this had no effect on c-Raf activity. However, toxin B decreased both the association of MEK1 and/or -2 with c-Raf and c-Raf-associated ERK-activating activity. V12Rac1 cooperated with c-Raf to increase expression of atrial natriuretic factor (ANF), whereas N17Rac1 inhibited endothelin 1-stimulated ANF expression, indicating that the synergy between Rac1 and c-Raf is potentially physiologically important. We conclude that activation of Rac1 by hypertrophic stimuli contributes to the hypertrophic response by modulating the ERK and/or possibly the JNK (but not the p38-MAPK) cascades.


Asunto(s)
Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Factor Natriurético Atrial/genética , Cardiomegalia/etiología , Células Cultivadas , Endotelina-1/farmacología , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica , Guanosina Trifosfato/metabolismo , Humanos , Miocardio/citología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Ratas , Transfección , Proteína de Unión al GTP rhoA/metabolismo
7.
Ann Med ; 33(9): 611-22, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11817656

RESUMEN

In response to a requirement for increased contractile power in vivo, mammalian cardiac myocytes adapt through a hypertrophic response (cell enlargement in the absence of cell division). This response can be simulated by exposing isolated myocytes in primary culture to alpha-adrenergic agonists or the vasoactive peptide, endothelin-1. The signalling pathways responsible for hypertrophic growth have been actively studied, and it is likely that reversible protein phosphorylation and dephosphorylation are involved. Three signalling pathways show particular potential as regulators of the response, ie protein kinase C (PKC), mitogen-activated protein kinase (MAPK) cascades, and calcineurin. These species are thought to regulate the rate and specificity of gene transcription ultimately through modifying the transactivating activity of nuclear transcription factors. There are three pertinent MAPK cascades, the extracellular signal-regulated kinase (ERK) cascade, the c-Jun N-terminal kinase (JNK or SAPK1) cascade, and the p38-MAPK (SAPK2-5) cascade. PKC participates in the activation of the ERK cascade but does not contribute significantly to the activation of the two remaining cascades. Calcineurin (or protein phosphatase 2B) is activated by increases in [Ca2+i] through the [Ca2+]-sensing protein, calmodulin. In this review, I discuss the evidence for and against the involvement of these signalling proteins in the induction of myocyte hypertrophy and emphasize that the ERK cascade should perhaps feature more widely in the collective consciousness.


Asunto(s)
Miocardio/citología , Transducción de Señal , Animales , Calcineurina/fisiología , Regulación de la Expresión Génica , Humanos , Hipertrofia , Proteínas Quinasas Activadas por Mitógenos/fisiología , Miocardio/patología , Estrés Oxidativo , Fosforilación , Proteína Quinasa C/fisiología , Tacrolimus/farmacología
8.
Circ Res ; 86(12): 1252-8, 2000 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-10864916

RESUMEN

Stimulation of phosphatidylinositol 3'-kinase (PI3K) and protein kinase B (PKB) is implicated in the regulation of protein synthesis in various cells. One mechanism involves PI3K/PKB-dependent phosphorylation of 4E-BP1, which dissociates from eIF4E, allowing initiation of translation from the 7-methylGTP cap of mRNAs. We examined the effects of insulin and H(2)O(2) on this pathway in neonatal cardiac myocytes. Cardiac myocyte protein synthesis was increased by insulin, but was inhibited by H(2)O(2). PI3K inhibitors attenuated basal levels of protein synthesis and inhibited the insulin-induced increase in protein synthesis. Insulin or H(2)O(2) increased the phosphorylation (activation) of PKB through PI3K, but, whereas insulin induced a sustained response, the response to H(2)O(2) was transient. 4E-BP1 was phosphorylated in unstimulated cells, and 4E-BP1 phosphorylation was increased by insulin. H(2)O(2) stimulated dephosphorylation of 4E-BP1 by increasing protein phosphatase (PP1/PP2A) activity. This increased the association of 4E-BP1 with eIF4E, consistent with H(2)O(2) inhibition of protein synthesis. The effects of H(2)O(2) were sufficient to override the stimulation of protein synthesis and 4E-BP1 phosphorylation induced by insulin. These results indicate that PI3K and PKB are important regulators of protein synthesis in cardiac myocytes, but other factors, including phosphatase activity, modulate the overall response.


Asunto(s)
Proteínas Portadoras , Miocardio/metabolismo , Estrés Oxidativo/fisiología , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Animales , Células Cultivadas , Factor 4E Eucariótico de Iniciación , Péptidos y Proteínas de Señalización Intracelular , Proteínas Musculares/biosíntesis , Miocardio/citología , Factores de Iniciación de Péptidos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt , Análogos de Caperuza de ARN/metabolismo , Ratas , Ratas Sprague-Dawley , Sefarosa/metabolismo
9.
Biochem Biophys Res Commun ; 273(2): 736-44, 2000 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-10873673

RESUMEN

This study aims to elucidate the signaling pathway for insulin-like growth factor-1 (IGF-1) in cultured neonatal rat cardiomyocytes and particularly the role of IGF-1 in cardiac apoptosis. IGF-1 stimulated polyphosphoinositide turnover, translocation of protein kinase C (PKC) isoforms (alpha, epsilon, and delta) from the soluble to the particulate fraction, activation of phospholipid-dependent and Ca(2+)-, phospholipid-dependent PKC, and activation of the extracellular-regulated kinase (ERK). IGF-1 attenuated sorbitol-induced cardiomyocyte viability and nuclear DNA fragmentation. These antiapoptotic effects of IGF-1 were blocked by PD-098059 (an MEK inhibitor) but not by bisindolylmaleimide I (BIM, a specific PKC inhibitor). The ERK pathway may therefore be an important component in the mechanism whereby IGF-1 exerts its antiapoptotic effect on the cardiomyocyte.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Hidrólisis , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Miocardio/citología , Miocardio/metabolismo , Presión Osmótica , Fosfatidilinositoles/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Ratas , Transducción de Señal/efectos de los fármacos
10.
Circ Res ; 86(10): 1019-23, 2000 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-10827130

RESUMEN

The small (21 kDa) guanine nucleotide-binding protein (small G protein) superfamily comprises 5 subfamilies (Ras, Rho, ADP ribosylation factors [ARFs], Rab, and Ran) that act as molecular switches to regulate numerous cellular responses. Cardiac myocyte hypertrophy is associated with cell growth and changes in the cytoskeleton and myofibrillar apparatus. In other cells, the Ras subfamily regulates cell growth whereas the Rho subfamily (RhoA, Rac1, and Cdc42) regulates cell morphology. Thus, the involvement of small G proteins in hypertrophy has become an area of significant interest. Hearts from transgenic mice expressing activated Ras develop features consistent with hypertrophy, whereas mice overexpressing RhoA develop lethal heart failure. In isolated neonatal rat cardiac myocytes, transfection or infection with activated Ras, RhoA, or Rac1 induces many of the features of hypertrophy. We discuss the mechanisms of activation of the small G proteins and the downstream signaling pathways involved. The latter may include protein kinases, particularly the mitogen-activated or Rho-activated protein kinases. We conclude that although there is significant evidence implicating Ras, RhoA, and Rac1 in hypertrophy, the mechanisms are not fully understood.


Asunto(s)
Cardiomegalia/etiología , Proteínas de Unión al GTP Monoméricas/fisiología , Animales , Humanos , Transducción de Señal , Proteínas ras/fisiología , Proteínas de Unión al GTP rho/fisiología
11.
Trends Cardiovasc Med ; 10(1): 1-8, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11150721

RESUMEN

The small (21-kDa) guanine nucleotide-binding protein Ras plays a central role in the regulation of cell growth and division. In the cardiac myocyte, it has been implicated in the hypertrophic adaptation. We have recently examined the ability of hypertrophic agonists such as endothelin-1, phenylephrine and phorbol esters to increase the "activity" (GTP loading) of Ras. We have also studied the signaling events that lead to activation of Ras and the processes that respond to Ras activation. In this brief review, we describe these studies and set them within the context of the hypertrophic response.


Asunto(s)
Cardiomegalia/metabolismo , Corazón/efectos de los fármacos , Proteínas de Unión al GTP Monoméricas/metabolismo , Miocardio/metabolismo , Agonistas alfa-Adrenérgicos/farmacología , Animales , Cardiomegalia/inducido químicamente , Endotelina-1/farmacología , Humanos , Proteínas de Unión al GTP Monoméricas/efectos de los fármacos , Fenilefrina/farmacología , Unión Proteica/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Vasoconstrictores/farmacología , Proteínas ras/efectos de los fármacos , Proteínas ras/metabolismo
12.
Circ Res ; 85(10): 940-9, 1999 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-10559141

RESUMEN

Cardiac myocyte apoptosis is potentially important in many cardiac disorders. In other cells, Bcl-2 family proteins and mitochondrial dysfunction are probably key regulators of the apoptotic response. In the present study, we characterized the regulation of antiapoptotic (Bcl-2, Bcl-xL) and proapoptotic (Bad, Bax) Bcl-2 family proteins in the rat heart during development and in oxidative stress-induced apoptosis. Bcl-2 and Bcl-xL were expressed at high levels in the neonate, and their expression was sustained during development. In contrast, although Bad and Bax were present at high levels in neonatal hearts, they were barely detectable in adult hearts. We confirmed that H(2)O(2) induced cardiac myocyte cell death, stimulating poly(ADP-ribose) polymerase proteolysis (from 2 hours), caspase-3 proteolysis (from 2 hours), and DNA fragmentation (from 8 hours). In unstimulated neonatal cardiac myocytes, Bcl-2 and Bcl-xL were associated with the mitochondria, but Bad and Bax were predominantly present in a crude cytosolic fraction. Exposure of myocytes to H(2)O(2) stimulated rapid translocation of Bad (<5 minutes) to the mitochondria. This was followed by the subsequent degradation of Bad and Bcl-2 (from approximately 30 minutes). The levels of the mitochondrial membrane marker cytochrome oxidase remained unchanged. H(2)O(2) also induced translocation of cytochrome c from the mitochondria to the cytosol within 15 to 30 minutes, which was indicative of mitochondrial dysfunction. Myocytes exposed to H(2)O(2) showed an early loss of mitochondrial membrane potential (assessed by fluorescence-activated cell sorter analysis) from 15 to 30 minutes, which was partially restored by approximately 1 hour. However, a subsequent irreversible loss of mitochondrial membrane potential occurred that correlated with cell death. These data suggest that the regulation of Bcl-2 and mitochondrial function are important factors in oxidative stress-induced cardiac myocyte apoptosis.


Asunto(s)
Envejecimiento/metabolismo , Miocardio/metabolismo , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Grupo Citocromo c/metabolismo , Peróxido de Hidrógeno/farmacología , Potenciales de la Membrana/fisiología , Mitocondrias Cardíacas/fisiología , Miocardio/citología , Oxidantes/farmacología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/metabolismo , Distribución Tisular/fisiología , Proteína Letal Asociada a bcl
13.
J Mol Cell Cardiol ; 31(8): 1429-34, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10423341

RESUMEN

Three well-characterized mitogen-activated protein kinase (MAPK) subfamilies are expressed in rodent and rabbit hearts, and are activated by pathophysiological stimuli. We have determined and compared the expression and activation of these MAPKs in donor and failing human hearts. The amount and activation of MAPKs was assessed in samples from the left ventricles of 4 unused donor hearts and 12 explanted hearts from patients with heart failure secondary to ischaemic heart disease. Total MAPKs or dually phosphorylated (activated) MAPKs were detected by Western blotting and MAPK activities were measured by in gel kinase assays. As in rat heart, c-Jun N-terminal kinases (JNKs) were detected in human hearts as bands corresponding to 46 and 54 kDa; p38-MAPK(s) was detected as a band corresponding to approximately 40 kDa, and extracellularly regulated kinases, ERK1 and ERK2, were detected as 44- and 42-kDa bands respectively. The total amounts of 54 kDa JNK, p38-MAPK and ERK2 were similar in all samples, although 46-kDa JNK was reduced in the failing hearts. However, the mean activities of JNKs and p38-MAPK(s) were significantly higher in failing heart samples than in those from donor hearts (P<0.05). There was no significant difference in phosphorylated (activated) ERKs between the two groups. In conclusion, JNKs, p38-MAPK(s) and ERKs are expressed in the human heart and the activities of JNKs and p38-MAPK(s) were increased in heart failure secondary to ischaemic heart disease. These data indicate that JNKs and p38-MAPKs may be important in human cardiac pathology.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Isquemia Miocárdica/enzimología , Animales , Activación Enzimática , Corazón/fisiopatología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Isquemia Miocárdica/fisiopatología , Miocardio/enzimología , Conejos , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos
14.
J Biol Chem ; 274(28): 19762-70, 1999 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-10391918

RESUMEN

The small G protein Ras has been implicated in hypertrophy of cardiac myocytes. We therefore examined the activation (GTP loading) of Ras by the following hypertrophic agonists: phorbol 12-myristate 13-acetate (PMA), endothelin-1 (ET-1), and phenylephrine (PE). All three increased Ras.GTP loading by 10-15-fold (maximal in 1-2 min), as did bradykinin. Other G protein-coupled receptor agonists (e.g. angiotensin II, carbachol, isoproterenol) were less effective. Activation of Ras by PMA, ET-1, or PE was reduced by inhibition of protein kinase C (PKC), and that induced by ET-1 or PE was partly sensitive to pertussis toxin. 8-(4-Chlorophenylthio)-cAMP (CPT-cAMP) did not inhibit Ras.GTP loading by PMA, ET-1, or PE. The association of Ras with c-Raf protein was increased by PMA, ET-1, or PE, and this was inhibited by CPT-cAMP. However, only PMA and ET-1 increased Ras-associated mitogen-activated protein kinase kinase 1-activating activity, and this was decreased by PKC inhibition, pertussis toxin, and CPT-cAMP. PMA caused the rapid appearance of phosphorylated (activated) extracellular signal-regulated kinase in the nucleus, which was inhibited by a microinjected neutralizing anti-Ras antibody. We conclude that PKC- and Gi-dependent mechanisms mediate the activation of Ras in myocytes and that Ras activation is required for stimulation of extracellular signal-regulated kinase by PMA.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Guanosina Trifosfato/metabolismo , Ventrículos Cardíacos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Proteínas ras/metabolismo , Animales , Células Cultivadas , AMP Cíclico/farmacología , Regulación hacia Abajo/efectos de los fármacos , Endotelina-1/farmacología , Activación Enzimática , MAP Quinasa Quinasa 1 , Toxina del Pertussis , Fenilefrina/farmacología , Fosforilación , Unión Proteica/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Virulencia de Bordetella/farmacología
16.
J Mol Cell Cardiol ; 31(12): 2087-99, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10640438

RESUMEN

Pro-inflammatory cytokines may be important in the pathophysiological responses of the heart. We investigated the activation of the three mitogen-activated protein kinase (MAPK) subfamilies ¿c-Jun N-terminal kinases (JNKs), p38-MAPKs and extracellularly-responsive kinases (ERKs) by interleukin-1 beta (IL-1 beta) or tumour necrosis factor alpha (TNF alpha) in primary cultures of myocytes isolated from neonatal rat ventricles. Both cytokines stimulated a rapid (maximal within 10 min) increase in JNK activity. Although activation of JNKs by IL-1 beta was transient returning to control values within 1 h, the response to TNF alpha was sustained. IL-1 beta and TNF alpha also stimulated p38-MAPK phosphorylation, but the response to IL-1 beta was consistently greater than TNF alpha. Both cytokines activated ERKs, but to a lesser degree than that induced by phorbol esters. The transcription factors, c-Jun and ATF2, are phosphorylated by the MAPKs and are implicated in the upregulation of c-Jun. IL-1 beta and TNF alpha stimulated the phosphorylation of c-Jun and ATF2. However, IL-1 beta induced a greater increase in c-Jun protein. Inhibitors of protein kinase C (PKC) (Ro318220, GF109203X) and the ERK cascade (PD98059) attenuated the increase in c-Jun induced by IL-1 beta, but LY294002 (an inhibitor of phosphatidylinositol 3' kinase) and SB203580 (an inhibitor of p38-MAPK, which also inhibits certain JNK isoforms) had no effect. These data illustrate that some of the pathological effects of IL-1 beta and TNF alpha may be mediated through the MAPK cascades, and that the ERK cascade, rather than JNKs or p38-MAPKs, are implicated in the upregulation of c-Jun by IL-1 beta.


Asunto(s)
Interleucina-1/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocardio/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología , Factor de Transcripción Activador 2 , Animales , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Activación Enzimática , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
17.
Am J Cardiol ; 83(12A): 64H-69H, 1999 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-10750590

RESUMEN

Cardiac myocyte hypertrophy involves changes in cell structure and alterations in protein expression regulated at both the transcriptional and translational levels. Hypertrophic G protein-coupled receptor (GPCR) agonists such as endothelin-(ET-1) and phenylephrine stimulate a number of protein kinase cascades in the heart. Mitogen-activated protein kinase (MAPK) cascades stimulated include the extracellularly regulated kinase cascade, the stress-activated protein kinase/c-Jun N-terminal kinase cascade, and the p38 MAPK cascade. All 3 pathways have been implicated in hypertrophy, but recent ex vivo evidence also suggests that there may be additional effects on cell survival. ET-1 and phenylephrine also stimulate the protein kinase B pathway, and this may be involved in the regulation of protein synthesis by these agonists. Thus, protein kinase-mediated signaling may be important in the regulation of the development of myocyte hypertrophy.


Asunto(s)
Agonistas alfa-Adrenérgicos/farmacología , Cardiomegalia/metabolismo , Endotelina-1/farmacología , Proteínas de Unión al GTP/agonistas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/metabolismo , Fenilefrina/farmacología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Cardiomegalia/genética , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Biosíntesis de Proteínas , Proteínas Quinasas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Transcripción Genética
18.
J Mol Med (Berl) ; 76(11): 725-46, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9826118

RESUMEN

Hypertrophy of myocytes in the heart ventricles is an important adaptation that in vivo occurs in response to a requirement for increased contractile power. It involves changes at the level of gene transcription, stimulation of the rate of protein synthesis (translation), and increased assembly of myofibrils. There is mounting evidence of the involvement of reversible protein phosphorylation and dephosphorylation in most of these processes. Protein kinase C, mitogen-activated protein kinases, and transcription factors have been implicated in the modulation of the transcriptional changes. Activation of translation may also be mediated through protein phosphorylation/dephosphorylation, although this has not been clearly established in the heart. Here we provide a critical overview of the signalling pathways involved in the hypertrophic response and provide a scheme to account for many of its features.


Asunto(s)
Cardiomegalia/metabolismo , Transducción de Señal , Animales , Cardiomegalia/patología , Humanos
19.
Biochem J ; 335 ( Pt 2): 241-6, 1998 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9761720

RESUMEN

Treatment of cultured neonatal ventricular myocytes with oncogenic Ras increases their size and stimulates the re-expression of genes which are normally restricted to the fetal stage of ventricular development, including atrial natriuretic factor (ANF) and skeletal muscle (SkM)-alpha-actin. To determine which signalling pathways mediate these responses, myocytes were transfected with oncogenic (V12) Ras mutants which interact selectively with different effectors and their effects on luciferase (LUX) reporter plasmids were examined. V12 human Ras (V12HRas), itself, activated ANF-LUX 9. 6-fold, whereas mutants of V12HRas, which selectively stimulate Ral guanine nucleotide dissociation stimulator (Ral.GDS) (E37G), c-Raf (D38E) and phosphatidylinositol 3-kinase (PI-3-K; Y40C) enhanced ANF-LUX expression 3.0-, 3.7- and 1.7-fold respectively. The full response of ANF-LUX to V12HRas was restored by using a combination of the individual effector domain mutants. Likewise, SkM-alpha-actin-LUX expression was activated 12.0-, 3.5-, 4.5- and 3. 0-fold by V12HRas, E37G, D38E and Y40C respectively, and a similar pattern of activation was also observed using a c-fos serum-response element-LUX reporter gene. Cell size was also increased by each of the mutants, but simultaneous expression of all three mutant constructs was needed to reconstitute the full effect of V12HRas on cell size (50% increase). Transfection with a constitutively active mutant of PI-3-K (p110K227E) stimulated ANF-LUX, SkM-alpha-actin-LUX, c-fos-serum-response element-LUX and Rous sarcoma virus-LUX by 3.1-, 3.2-, 2.1- and 2.9-fold respectively, but the co-transfected cytomegalovirus-beta-galactosidase reporter gene was activated to a similar extent (1.9-fold). These results suggest that Raf, Ral.GDS and PI-3-K can all transduce transcriptional responses to V12HRas, but that the specific induction of genes associated with the hypertrophic response is not mediated through PI-3-K.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Genes ras , Ventrículos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Actinas/genética , Actinas/metabolismo , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Tamaño de la Célula , Células Cultivadas , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Genes fos , Ventrículos Cardíacos/citología , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mutación , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Respuesta , Factor de Intercambio de Guanina Nucleótido ral , Proteínas de Unión al GTP rap
20.
Adv Enzyme Regul ; 38: 87-98, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9762348

RESUMEN

Using primary cultures of neonatal rat ventricular myocytes and isolated adult rat hearts as models, we have characterized extensively the regulation of MAPKs in the heart. The ERKs are activated primarily by GPCR agonists acting through PKC. These agonists can also activate the JNKs although the mechanism is unclear. Cellular stresses stimulate strong activation of the JNKs, but also cause some stimulation of ERKs. Activation of p38-MAPK has so far only been demonstrated in intact adult hearts subjected to stresses and probably leads to activation of MAPKAPK2. Both cellular stresses and GPCR agonists induce phosphorylation of c-Jun, but only the latter causes upregulation of c-Jun protein.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Miocardio/enzimología , Animales , Células Cultivadas , Citocinas/farmacología , Activación Enzimática , Proteínas de Unión al GTP/metabolismo , Peróxido de Hidrógeno/farmacología , Fosforilación , Ratas , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/farmacología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...