Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Med ; 10(3): 248-54, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14966518

RESUMO

The protein kinase C (PKC) family of serine/threonine kinases functions downstream of nearly all membrane-associated signal transduction pathways. Here we identify PKC-alpha as a fundamental regulator of cardiac contractility and Ca(2+) handling in myocytes. Hearts of Prkca-deficient mice are hypercontractile, whereas those of transgenic mice overexpressing Prkca are hypocontractile. Adenoviral gene transfer of dominant-negative or wild-type PKC-alpha into cardiac myocytes enhances or reduces contractility, respectively. Mechanistically, modulation of PKC-alpha activity affects dephosphorylation of the sarcoplasmic reticulum Ca(2+) ATPase-2 (SERCA-2) pump inhibitory protein phospholamban (PLB), and alters sarcoplasmic reticulum Ca(2+) loading and the Ca(2+) transient. PKC-alpha directly phosphorylates protein phosphatase inhibitor-1 (I-1), altering the activity of protein phosphatase-1 (PP-1), which may account for the effects of PKC-alpha on PLB phosphorylation. Hypercontractility caused by Prkca deletion protects against heart failure induced by pressure overload, and against dilated cardiomyopathy induced by deleting the gene encoding muscle LIM protein (Csrp3). Deletion of Prkca also rescues cardiomyopathy associated with overexpression of PP-1. Thus, PKC-alpha functions as a nodal integrator of cardiac contractility by sensing intracellular Ca(2+) and signal transduction events, which can profoundly affect propensity toward heart failure.


Assuntos
Baixo Débito Cardíaco/fisiopatologia , Isoenzimas/metabolismo , Contração Miocárdica/fisiologia , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Calsequestrina/metabolismo , Cardiomiopatias/metabolismo , Isoenzimas/genética , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/metabolismo , Proteína Quinase C/genética , Proteína Quinase C-alfa , Proteína Fosfatase 1 , Ratos , Fatores de Risco , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático
2.
J Cell Biol ; 156(5): 905-19, 2002 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-11864993

RESUMO

Members of the protein kinase C (PKC) isozyme family are important signal transducers in virtually every mammalian cell type. Within the heart, PKC isozymes are thought to participate in a signaling network that programs developmental and pathological cardiomyocyte hypertrophic growth. To investigate the function of PKC signaling in regulating cardiomyocyte growth, adenoviral-mediated gene transfer of wild-type and dominant negative mutants of PKC alpha, beta II, delta, and epsilon (only wild-type zeta) was performed in cultured neonatal rat cardiomyocytes. Overexpression of wild-type PKC alpha, beta II, delta, and epsilon revealed distinct subcellular localizations upon activation suggesting unique functions of each isozyme in cardiomyocytes. Indeed, overexpression of wild-type PKC alpha, but not betaI I, delta, epsilon, or zeta induced hypertrophic growth of cardiomyocytes characterized by increased cell surface area, increased [(3)H]-leucine incorporation, and increased expression of the hypertrophic marker gene atrial natriuretic factor. In contrast, expression of dominant negative PKC alpha, beta II, delta, and epsilon revealed a necessary role for PKC alpha as a mediator of agonist-induced cardiomyocyte hypertrophy, whereas dominant negative PKC epsilon reduced cellular viability. A mechanism whereby PKC alpha might regulate hypertrophy was suggested by the observations that wild-type PKC alpha induced extracellular signal-regulated kinase1/2 (ERK1/2), that dominant negative PKC alpha inhibited PMA-induced ERK1/2 activation, and that dominant negative MEK1 (up-stream of ERK1/2) inhibited wild-type PKC alpha-induced hypertrophic growth. These results implicate PKC alpha as a necessary mediator of cardiomyocyte hypertrophic growth, in part, through a ERK1/2-dependent signaling pathway.


Assuntos
Cardiomiopatia Hipertrófica/enzimologia , Diferenciação Celular/fisiologia , Coração/crescimento & desenvolvimento , Isoenzimas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Recém-Nascidos , Fator Natriurético Atrial/efeitos dos fármacos , Fator Natriurético Atrial/metabolismo , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/fisiopatologia , Compartimento Celular/efeitos dos fármacos , Compartimento Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Coração/efeitos dos fármacos , Coração/fisiologia , Imuno-Histoquímica , Isoenzimas/efeitos dos fármacos , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Miocárdio/citologia , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C-alfa , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tubulina (Proteína)/metabolismo
3.
Circulation ; 113(7): 995-1004, 2006 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-16476846

RESUMO

BACKGROUND: Phospholamban (PLN) is an inhibitor of the Ca2+ affinity of sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA2). The amino acid sequence of PLN is highly conserved, and although all species contain asparagine (Asn), human PLN is unique in containing lysine (Lys) at amino acid 27. METHODS AND RESULTS: Human PLN was introduced in the null background. Expression of human PLN, at similar levels to mouse wild-type PLN, resulted in significant decreases in the affinity of SERCA2 for Ca2+, attributed to unique spatial conformation of this PLN form and increases in its monomeric active unit compared with mouse PLN. The increased inhibition by human PLN was associated with attenuated cardiac contractility in the intact-animal, organ, and cardiomyocyte levels and with depressed calcium kinetics. These inhibitory effects could not be fully reversed even on maximal isoproterenol stimulation. There were no alterations in the expression levels of SERCA2, calsequestrin, ryanodine receptor, and FKBP12, although the sodium/calcium exchanger and the L-type Ca2+ channel expression levels were upregulated. The depressed function resulted in increased heart/body weight ratios and phosphorylation levels of Akt, p38, and Erk1/2. CONCLUSIONS: Human PLN may play a more inhibitory role than that of other species in Ca2+ cycling. Expression of human PLN in the mouse is compensated by alterations in Ca2+-handling proteins and cardiac remodeling in an effort to normalize cardiac contractility. Thus, the unique amino acid sequence of human PLN may be critical in maintaining a high cardiac reserve, which is of paramount importance in the regulation of human cardiac function.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/fisiologia , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Cardiomegalia/etiologia , Sequência de Aminoácidos , Animais , Arginina , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Humanos , Cinética , Lisina , Camundongos , Camundongos Knockout , Camundongos Transgênicos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Especificidade da Espécie
4.
J Clin Invest ; 111(10): 1475-86, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12750397

RESUMO

The MAPKs are important transducers of growth and stress stimuli in virtually all eukaryotic cell types. In the mammalian heart, MAPK signaling pathways have been hypothesized to regulate myocyte growth in response to developmental signals or physiologic and pathologic stimuli. Here we generated cardiac-specific transgenic mice expressing dominant-negative mutants of p38alpha, MKK3, or MKK6. Remarkably, attenuation of cardiac p38 activity produced a progressive growth response and myopathy in the heart that correlated with the degree of enzymatic inhibition. Moreover, dominant-negative p38alpha, MKK3, and MKK6 transgenic mice each showed enhanced cardiac hypertrophy following aortic banding, Ang II infusion, isoproterenol infusion, or phenylephrine infusion for 14 days. A mechanism underlying this enhanced-growth profile was suggested by the observation that dominant-negative p38alpha directly augmented nuclear factor of activated T cells (NFAT) transcriptional activity and its nuclear translocation. In vivo, NFAT-dependent luciferase reporter transgenic mice showed enhanced activation in the presence of the dominant-negative p38alpha transgene before and after the onset of cardiac hypertrophy. More significantly, genetic disruption of the calcineurin Abeta gene rescued hypertrophic cardiomyopathy and depressed functional capacity observed in p38-inhibited mice. Collectively, these observations indicate that reduced p38 signaling in the heart promotes myocyte growth through a mechanism involving enhanced calcineurin-NFAT signaling.


Assuntos
Calcineurina/metabolismo , Cardiomiopatia Hipertrófica/fisiopatologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Nucleares , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Animais , Calcineurina/deficiência , Calcineurina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/patologia , Células Cultivadas , Progressão da Doença , Marcação de Genes , Genes Dominantes , Genes Reporter , Técnicas In Vitro , MAP Quinase Quinase 3 , MAP Quinase Quinase 6 , Camundongos , Camundongos Transgênicos , Proteína Quinase 14 Ativada por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Ratos , Transgenes , Regulação para Cima
5.
J Biomed Opt ; 12(5): 054008, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17994896

RESUMO

Structural proteins such as elastin and collagen can be readily imaged by using two-photon excitation and second-harmonic generation microscopic techniques, respectively, without physical or biochemical processing of the tissues. This time- and effort-saving advantage makes these imaging techniques convenient for determining the structural characteristics of blood vessels in vivo. Fibrillar collagen is a well-known element involved in the formation of atherosclerotic lesions. It is also an important component of the fibrous cap responsible for structural stability of atherosclerotic plaques. High resolution in vivo microscopic imaging and characterization of atherosclerotic lesions in animal models can be particularly useful for drug discovery. However, it is hindered by the limitations of regular microscope objectives to gain access of the tissues of interest and motional artifacts. We report a technique that facilitates in vivo microscopic imaging of carotid arteries of rodents using conventional microscope objectives, and at the same time avoids motional artifacts. As a result, collagen, elastin, leukocytes, cell nuclei, and neutral lipids can be visualized in three dimensions in live animals. We present and discuss in vivo imaging results using a flow cessation mouse model of accelerated atherosclerosis.


Assuntos
Apolipoproteínas E/metabolismo , Doenças das Artérias Carótidas/patologia , Aumento da Imagem/métodos , Arteriosclerose Intracraniana/patologia , Microscopia de Fluorescência/métodos , Animais , Apolipoproteínas E/genética , Doenças das Artérias Carótidas/metabolismo , Arteriosclerose Intracraniana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dinâmica não Linear
6.
Mol Cell Biol ; 22(21): 7603-13, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12370307

RESUMO

A calcineurin-nuclear factor of activated T cells (NFAT) regulatory pathway has been implicated in the control of cardiac hypertrophy, suggesting one mechanism whereby alterations in intracellular calcium handling are linked to the expression of hypertrophy-associated genes. Although recent studies have demonstrated a necessary role for calcineurin as a mediator of cardiac hypertrophy, the potential involvement of NFAT transcription factors as downstream effectors of calcineurin signaling has not been evaluated. Accordingly, mice with targeted disruptions in NFATc3 and NFATc4 genes were characterized. Whereas the loss of NFATc4 did not compromise the ability of the myocardium to undergo hypertrophic growth, NFATc3-null mice demonstrated a significant reduction in calcineurin transgene-induced cardiac hypertrophy at 19 days, 26 days, 6 weeks, 8 weeks, and 10 weeks of age. NFATc3-null mice also demonstrated attenuated pressure overload- and angiotensin II-induced cardiac hypertrophy. These results provide genetic evidence that calcineurin-regulated responses require NFAT effectors in vivo.


Assuntos
Calcineurina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Mutagênese , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Angiotensina II/metabolismo , Animais , Western Blotting , Divisão Celular , Núcleo Celular/metabolismo , Feminino , Marcação de Genes , Hipertrofia , Masculino , Camundongos , Modelos Genéticos , Mutagênese Sítio-Dirigida , Miocárdio/metabolismo , Fatores de Transcrição NFATC , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transgenes
7.
Eur J Heart Fail ; 11(8): 739-48, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19633101

RESUMO

AIMS: Activation of phosphoinositide-3 kinase (PI3K) is essential for cell growth, relating to adaptive and maladaptive cardiac hypertrophy. This longitudinal canine study was designed to investigate the role of PI3Kalpha and PI3Kgamma in cardiac remodelling during congestive heart failure (CHF) and cardiac recovery (CR). METHODS AND RESULTS: All dogs were surgically instrumented. Congestive heart failure was induced by cardiac pacing for 3-4 weeks and CR was allowed by terminating pacing for 5-6 weeks after induction of HF. Control dogs had sham surgery, but did not undergo pacing. Left ventricular (LV) contractile function was depressed in CHF and restored to 80-90% of the normal level in CR, with a 25% increase in LV weight. The expression of PI3Kgamma was increased four-fold in CHF, but returned to control levels in CR. In contrast, the expression of PI3Kalpha in CHF was not different from that in controls, but increased three-fold in CR and was accompanied by increases in phosphorylation of Akt (five-fold), GSK-3beta (five-fold), beta-catenin (three-fold), mTOR (two-fold), and P70S6K (two-fold). CONCLUSION: Our results indicate that PI3K isoforms are regulated differently during the course of CHF/CR and that the selective activation of PI3Kalpha, through Akt, GSK-3beta, and mTOR signalling pathways, may be involved in the development of cardiac compensatory hypertrophy and functional restoration.


Assuntos
Quinase 3 da Glicogênio Sintase , Insuficiência Cardíaca/fisiopatologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Análise de Variância , Animais , Dobutamina , Cães , Glicogênio Sintase Quinase 3 beta , Hemodinâmica , Estudos Longitudinais , Masculino , Modelos Animais , RNA Mensageiro , Transdução de Sinais
8.
Am J Physiol Heart Circ Physiol ; 292(6): H2782-90, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17322419

RESUMO

In congestive heart failure (CHF), coronary vascular relaxation is associated with endothelial dysfunction and nitric oxide (NO) deficiency. This study explored the reversibility of this process in hearts recovering from CHF and its related mechanisms. Dogs were chronically instrumented to measure cardiac function and coronary blood flow (CBF). Heart failure was induced by right ventricular pacing at 240 beats/min for 3-4 wk, and cardiac recovery (CR) was allowed by the termination of cardiac pacing for 3-4 wk after the development of CHF, in which left ventricular contractile function was restored by 80-90%. The endothelium-dependent CBF response to bradykinin and acetylcholine was depressed in CHF and fully restored in CR. Myocardial NOx (nitrate/nitrite), endothelial NO synthase (eNOS) mRNA expression, total protein, and phosphorylated eNOS decreased significantly in failing hearts. However, myocardial NOx recovered to 78% of control and phosphorylated eNOS was fully restored in CR, despite the fact that eNOS mRNA expression and protein levels remained lower than control. Furthermore, the endothelium-independent CBF response to nitroglycerin did not change in CHF; however, it increased by 75% in CR, in conjunction with a near threefold increase in the phosphorylation of vasodilation-stimulated phosphoprotein (VASP) at Ser(239) in recovering hearts. Thus the complete restoration of endothelium-dependent coronary vascular relaxation during cardiac recovery from CHF was mediated by 1) a restoration of phosphorylated eNOS for partial recovery of the NO production and 2) an increase in cGMP/cGMP-dependent protein kinase-I pathway signaling activity for the enhancement of coronary vascular smooth muscle relaxation in response to NO.


Assuntos
Vasos Coronários/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Endotélio Vascular/metabolismo , Insuficiência Cardíaca/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Vasodilatação , Acetilcolina/farmacologia , Animais , Bradicinina/farmacologia , Circulação Coronária , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/enzimologia , Vasos Coronários/fisiopatologia , Modelos Animais de Doenças , Cães , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Masculino , Contração Miocárdica , Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Nitroglicerina/farmacologia , Fosfoproteínas/metabolismo , Fosforilação , RNA Mensageiro/metabolismo , Recuperação de Função Fisiológica , Transdução de Sinais , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Função Ventricular Esquerda
9.
Am J Physiol Heart Circ Physiol ; 291(6): H3154-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16877566

RESUMO

Left ventricular (LV) diastolic dysfunction is a fundamental impairment in congestive heart failure (CHF). This study examined LV diastolic function in the canine model of CHF induced by chronic coronary embolization (CCE). Dogs were implanted with coronary catheters (both left anterior descending and circumflex arteries) for CCE and instrumented for measurement of LV pressure and dimension. Heart failure was elicited by daily intracoronary injections of microspheres (1.2 million, 90- to 120-microm diameter) for 24 +/- 4 days, resulting in significant depression of cardiac systolic function. After CCE, LV maximum negative change of pressure with time (dP/dt(min)) decreased by 25 +/- 2% (P < 0.05) and LV isovolumic relaxation constant and duration increased by 19 +/- 5% and 25 +/- 6%, respectively (both P < 0.05), indicating an impairment of LV active relaxation, which was cardiac preload independent. LV passive viscoelastic properties were evaluated from the LV end-diastolic pressure (EDP)-volume (EDV) relationship (EDP = be(alpha*EDV)) during brief inferior vena caval occlusion and acute volume loading, while the chamber stiffness coefficient (alpha) increased by 62 +/- 10% (P < 0.05) and the stiffness constant (k) increased by 66 +/- 13% after CCE. The regional myocardial diastolic stiffness in LV anterior and posterior walls was increased by 70 +/- 25% and 63 +/- 24% (both P < 0.05), respectively, after CCE, associated with marked fibrosis, increase in collagen I and III, and enhancement of plasminogen activator inhibitor-1 (PAI-1) protein expression. Thus along with depressed LV systolic function there is significant impairment of LV diastolic relaxation and increase in chamber stiffness, with development of myocardial fibrosis and activation of PAI-1, in the canine model of CHF induced by CCE.


Assuntos
Embolia/complicações , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Disfunção Ventricular Esquerda/fisiopatologia , Animais , Pressão Sanguínea/fisiologia , Doença Crônica , Colágeno/metabolismo , Modelos Animais de Doenças , Cães , Fibrose/metabolismo , Fibrose/patologia , Insuficiência Cardíaca/metabolismo , Masculino , Microcirculação/fisiopatologia , Microesferas , Contração Miocárdica/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Disfunção Ventricular Esquerda/metabolismo
10.
J Biol Chem ; 278(16): 14555-64, 2003 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-12566450

RESUMO

Recent studies identify conventional protein kinase C (PKC) isoform phosphorylations at conserved residues in the activation loop and C terminus as maturational events that influence enzyme activity and targeting but are not dynamically regulated by second messengers. In contrast, this study identifies phorbol 12-myristoyl 13-acetate (PMA)- and norepinephrine-induced phosphorylations of PKC epsilon (at the C-terminal hydrophobic motif) and PKC delta (at the activation loop) as events that accompany endogenous novel PKC (nPKC) isoform activation in neonatal rat cardiomyocytes. Agonist-induced nPKC phosphorylations are prevented (and the kinetics of PMA-dependent PKC down-regulation are slowed) by pharmacologic inhibitors of nPKC kinase activity. PKC delta is recovered from PMA-treated cultures with increased in vitro lipid-independent kinase activity (and altered substrate specificity); the PMA-dependent increase in PKC delta kinase activity is attenuated when PKC delta activation loop phosphorylation is prevented. To distinguish roles of individual nPKC isoforms in nPKC phosphorylations, wild-type (WT) and dominant negative (DN) PKC delta and PKC epsilon mutants were introduced into cardiomyocyte cultures using adenovirus-mediated gene transfer. WT-PKC delta and WT-PKC epsilon are highly phosphorylated at activation loop and hydrophobic motif sites, even in the absence of allosteric activators. DN-PKC delta is phosphorylated at the activation loop but not the hydrophobic motif; DN-PKC epsilon is phosphorylated at the hydrophobic motif but not the activation loop. Collectively, these results identify a role for PKC epsilon in nPKC activation loop phosphorylations and PKC delta in nPKC hydrophobic motif phosphorylations. Agonist-induced nPKC isoform phosphorylations that accompany activation/translocation of the enzyme contribute to the regulation of PKC delta kinase activity, may influence nPKC isoform trafficking/down-regulation, and introduce functionally important cross-talk for nPKC signaling pathways in cardiomyocytes.


Assuntos
Miocárdio/citologia , Proteína Quinase C/química , Proteína Quinase C/fisiologia , Adenoviridae/genética , Sítio Alostérico , Motivos de Aminoácidos , Animais , Células Cultivadas , Sequência Conservada , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica , Técnicas de Transferência de Genes , Genes Dominantes , Immunoblotting , Cinética , Modelos Biológicos , Fosforilação , Isoformas de Proteínas , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Transporte Proteico , Ratos , Ratos Wistar , Transdução de Sinais , Especificidade por Substrato , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA