Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 285(24): 18352-63, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20356841

RESUMEN

Protein kinase A (PKA) activation by cAMP phosphorylates multiple target proteins in numerous platelet inhibitory pathways that have a very important role in maintaining circulating platelets in a resting state. Here we show that in thrombin- and collagen-stimulated platelets, PKA is activated by cAMP-independent mechanisms involving dissociation of the catalytic subunit of PKA (PKAc) from an NFkappaB-IkappaBalpha-PKAc complex. We demonstrate mRNA and protein expression for most of the NFkappaB family members in platelets. From resting platelets, PKAc was co-immunoprecipitated with IkappaBalpha, and conversely, IkappaBalpha was also co-immunoprecipitated with PKAc. This interaction was significantly reduced in thrombin- and collagen-stimulated platelets. Stimulation of platelets with thrombin- or collagen-activated IKK, at least partly by PI3 kinase-dependent pathways, leading to phosphorylation of IkappaBalpha, disruption of an IkappaBalpha-PKAc complex, and release of free, active PKAc, which phosphorylated VASP and other PKA substrates. IKK inhibitor inhibited thrombin-stimulated IkBalpha phosphorylation, PKA-IkBalpha dissociation, and VASP phosphorylation, and potentiated integrin alphaIIbbeta3 activation and the early phase of platelet aggregation. We conclude that thrombin and collagen not only cause platelet activation but also appear to fine-tune this response by initiating downstream NFkappaB-dependent PKAc activation, as a novel feedback inhibitory signaling mechanism for preventing undesired platelet activation.


Asunto(s)
Plaquetas/metabolismo , Colágeno/química , Proteínas Quinasas Dependientes de AMP Cíclico/química , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , Trombina/química , Dominio Catalítico , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Células HL-60 , Humanos , Proteínas I-kappa B/metabolismo , Inhibidor NF-kappaB alfa , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Agregación Plaquetaria
2.
J Biol Chem ; 281(50): 38644-52, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17038326

RESUMEN

Cyclic GMP-dependent protein kinases protein kinase G (PKG) Ialpha and PKGIbeta are major mediators of cGMP signaling in the cardiovascular system. PKGIalpha is present in the heart, although its role in protection against ischemia/reperfusion injury is not known. We investigated the direct effect of PKGIalpha against necrosis and apoptosis following simulated ischemia (SI) and reoxygenation (RO) in cardiomyocytes. Adult rat cardiomyocytes were infected with adenoviral vectors containing hPKGIalpha or catalytically inactive mutant hPKGIalphaK390A. After 24 h, the cells were subjected to 90 min of SI and 2 h RO for necrosis (trypan blue exclusion and lactate dehydrogenase release) or 18 h RO for apoptosis studies. To evaluate the role of K(ATP) channels, subgroups of cells were treated with 5-hydroxydecanoate (100 microm), HMR1098 (30 microm), or glibenclamide (50 microm), the respective blockers of mitochondrial, sarcolemmal, or both types of K(ATP) channels prior to SI. The necrosis observed in 33.7 +/- 1.6% of total myocytes in the SI-RO control group was reduced to 18.6 +/- 0.8% by PKGIalpha (mean +/- S.E., n = 7, p < 0.001). The apoptosis observed in 17.9 +/- 1.3% of total myocytes in the SI-RO control group was reduced to 6.0 +/- 0.6% by PKGIalpha (mean +/- S.E., n = 7, p < 0.001). In addition, PKGIalpha inhibited the activation of caspase-3 after SI-RO in myocytes. Myocytes infected with the inactive PKGIalphaK390A mutant showed no protection. PKGIalpha enhanced phosphorylation of Akt, ERK1/2, and JNK, increased Bcl-2, inducible nitric-oxide synthase, endothelial nitric-oxide synthase, and decreased Bax expression. 5-Hydroxydecanoate and glibenclamide abolished PKGIalpha-mediated protection against necrosis and apoptosis. However, HMR1098, had no effect. A scavenger of reactive oxygen species, as well as inhibitors of phosphatidylinositol 3-kinase, ERK, JNK1, and NOS, also blocked PKGIalpha-mediated protection against necrosis and apoptosis. These results show that opening of mitochondrial K(ATP) channels and generation of reactive oxygen species, in association with phosphorylation of Akt, ERK, and JNK, and increased expression of NOS and Bcl-2, play an essential role in the protective effect of PKGIalpha.


Asunto(s)
Apoptosis , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Isquemia Miocárdica/enzimología , Oxígeno/metabolismo , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Depuradores de Radicales Libres/farmacología , Masculino , Isquemia Miocárdica/patología , Necrosis , Óxido Nítrico Sintasa/metabolismo , Canales de Potasio/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteína X Asociada a bcl-2/metabolismo
3.
J Biol Chem ; 281(43): 32831-40, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16943189

RESUMEN

Cardiac myocyte apoptosis during ischemia and reperfusion (I/R) is tightly controlled by a complex network of stress-responsive signaling pathways. One pro-apoptotic pathway involves the interaction of the scaffold protein TAB1 with p38 mitogen-activated protein kinase (p38 MAPK) leading to the autophosphorylation and activation of p38 MAPK. Conversely, NO and its second messenger cGMP protect cardiac myocytes from apoptosis during I/R. We provide evidence that the cGMP target cGMP-dependent protein kinase type I (PKG I) interferes with TAB1-p38 MAPK signaling to protect cardiac myocytes from I/R injury. In isolated neonatal cardiac myocytes, activation of PKG I inhibited the interaction of TAB1 with p38 MAPK, p38 MAPK phosphorylation, and apoptosis induced by simulated I/R. During I/R in vivo, mice with a cardiac myocyte-restricted deletion of PKG I displayed a more pronounced interaction of TAB1 with p38 MAPK and a stronger phosphorylation of p38 MAPK in the myocardial area at risk during reperfusion and more apoptotic cardiac myocytes in the infarct border zone as compared with wild-type littermates. Notably, adenoviral expression of a constitutively active PKG I mutant truncated at the N terminus(PKGI-DeltaN1-92) did not inhibit p38 MAPK phosphorylation and apoptosis induced by simulated I/R in vitro, indicating that the N terminus of PKG I is required. As shown by co-immunoprecipitation experiments in HEK293 cells, cGMP-activated PKG I, but not constitutively active PKG I-DeltaN1-92 or PKG I mutants carrying point mutations in the N-terminal leucine-isoleucine zipper, interacted with p38 MAPK, and prevented the binding of TAB1 to p38 MAPK. Together, our data identify a novel interaction between the cGMP target PKG I and the TAB1-p38 MAPK signaling pathway that serves as a defense mechanism against myocardial I/R injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adenoviridae/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Activación Enzimática , Ventrículos Cardíacos/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Biochemistry ; 45(4): 1295-303, 2006 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-16430226

RESUMEN

Natriuretic peptide receptor A (NPR-A) and natriuretic peptide receptor B (NPR-B) are transmembrane guanylyl cyclases that catalyze the synthesis of cGMP in response to natriuretic peptides. Phosphorylation and dephosphorylation regulate these receptors and have been traditionally studied by (32)PO(4) labeling of transfected cells. However, this approach cannot be used to determine the phosphorylation state of receptors isolated from unlabeled sources. Here, we use Pro-Q Diamond and SYPRO Ruby dyes to quantify the phosphorylation status and protein levels, respectively, of natriuretic peptide receptors from tissues and cells. Strong Pro-Q Diamond signals for NPR-A and NPR-B were obtained when receptors were isolated from lung tissue, liver tissue and overexpressing cells. The level of NPR-A Pro-Q staining was also high in kidney but was much lower in heart tissue. In contrast, the SYPRO Ruby protein signal was weaker and more variable. In a direct comparison, Pro-Q Diamond staining was as sensitive as but more specific than the (32)PO(4) labeling method. The two approaches were highly correlated (R(2) = 0.98). We exploited these techniques to measure the effect of cGMP-dependent protein kinase Ialpha on the phosphate content and guanylyl cyclase activity of NPR-A. Neither value was significantly affected in cells overexpressing cGK-Ialpha or in tissues from mice lacking cGK-I. We conclude that cGK-I does not regulate the cyclase activity or phosphorylation state of NPR-A. Furthermore, we find that Pro-Q Diamond staining is a sensitive method for measuring the phosphate levels of natriuretic peptide receptors, but protein levels are best detected by Western blot analysis, not SYPRO Ruby staining.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Guanilato Ciclasa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Coloración y Etiquetado/métodos , Animales , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Activación Enzimática , Péptidos y Proteínas de Señalización Intracelular/genética , Marcaje Isotópico/métodos , Ratones , Fosforilación , Ratas , Sensibilidad y Especificidad , Factores de Tiempo , Transfección
5.
Mol Endocrinol ; 20(2): 348-61, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16210344

RESUMEN

Cyclic GMP (cGMP)-dependent protein kinase II (Prkg2, cGK II) was identified as a potential target of the progesterone receptor (Nr3c3) in the mouse ovary based on microarray analyses. To document this further, the expression patterns of cGK II and other components of the cGMP signaling pathway were analyzed during follicular development and ovulation using the pregnant mare serum gonadotropin (PMSG)-human chorionic gonadotropin (hCG)-primed immature mice. Levels of cGK II mRNA were low in ovaries of immature mice, increased 4-fold in response to pregnant mare serum gonadotropin and 5-fold more within 12 h after hCG, the time of ovulation. In situ hybridization localized cGK II mRNA to granulosa cells and cumulus oocyte complexes of periovulatory follicles. In progesterone receptor (PR) null mice, cGK II mRNA was reduced significantly at 12 h after hCG in contrast to heterozygous littermates. In primary granulosa cell cultures, cGK II mRNA was induced by phorbol 12-myristate 13-acetate enhanced by adenoviral expression of PR-A and blocked by RU486 and trilostane. PR-A in the absence of phorbol 12-myristate 13-acetate was insufficient to induce cGK II. Expression of cGK I (Prkg1) was restricted to the residual tissue and not regulated by hormones. Guanylate cyclase-A (Npr1; GC-A) mRNA expression increased 6-fold by 4 h after hCG treatment in contrast to pregnant mare serum gonadotropin alone and was localized to granulosa cells of preovulatory follicles. Collectively, these data show for the first time that cGK II (not cGK I) and GC-A are selectively induced in granulosa cells of preovulatory follicles by LH- and PR-dependent mechanisms, thereby providing a pathway for cGMP function during ovulation.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Células de la Granulosa/enzimología , Hormona Luteinizante/farmacología , Folículo Ovárico/efectos de los fármacos , Receptores de Progesterona/metabolismo , Animales , Gonadotropina Coriónica/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo II , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Femenino , Gonadotropinas Equinas/farmacología , Células de la Granulosa/química , Guanilato Ciclasa/metabolismo , Ratones , Mifepristona/farmacología , Oocitos/química , Oocitos/enzimología , Folículo Ovárico/citología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Receptores de Progesterona/genética , Acetato de Tetradecanoilforbol/farmacología
6.
Front Biosci ; 10: 1313-28, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15769627

RESUMEN

The options available for distinguishing the effects of cGMP mediated by cGK versus those mediated by other cGMP targets are discussed and evaluated. These include the unnecessary but often sole reliance on synthetic, small-molecule activators and inhibitors of cGK which are increasingly recognized as deficient in specificity. Other important adjunct options include cGK overexpression using adenoviral vectors and transgenic animals, or use of cGK-deficient systems, i.e. cells which have spontaneously lost cGK during repetitive passaging in cell culture, cells treated with siRNA, or genetically-engineered cGK-deficient mice. Finally, cGK-dependent phosphorylation of substrates such as vasodilator stimulated phosphoprotein (VASP) and phosphodiesterase 5 (PDE 5) is described as a useful monitor of cGK presence and activity associated with physiological functions or dysfunctions of signaling pathways.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Animales , GMP Cíclico/fisiología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteína Quinasa Dependiente de GMP Cíclico Tipo II , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Humanos , Ratones , Fosforilación
7.
Neuron ; 45(3): 389-403, 2005 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-15694326

RESUMEN

Recent results suggest that long-lasting potentiation at hippocampal synapses involves the rapid formation of clusters or puncta of presynaptic as well as postsynaptic proteins, both of which are blocked by antagonists of NMDA receptors and an inhibitor of actin polymerization. We have investigated whether the increase in puncta involves retrograde signaling through the NO-cGMP-cGK pathway and also examined the possible roles of two classes of molecules that regulate the actin cytoskeleton: Ena/VASP proteins and Rho GTPases. Our results suggest that NO, cGMP, cGK, actin, and Rho GTPases including RhoA play important roles in the potentiation and act directly in both the presynaptic and postsynaptic neurons, where they contribute to the increase in puncta of synaptic proteins. cGK phosphorylates synaptic VASP during the potentiation, whereas Rho GTPases act both in parallel and upstream of cGMP, in part by maintaining the synaptic localization of soluble guanylyl cyclase.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Óxido Nítrico/metabolismo , Terminales Presinápticos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Animales Recién Nacidos , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , GMP Cíclico/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Guanilato Ciclasa , Hipocampo/citología , Proteínas de Microfilamentos , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Terminales Presinápticos/ultraestructura , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/fisiología , Guanilil Ciclasa Soluble , Membranas Sinápticas/metabolismo , Transmisión Sináptica/fisiología , Sinaptofisina/metabolismo
8.
J Biol Chem ; 280(17): 16642-50, 2005 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15722341

RESUMEN

Electroneutral NaCl absorption mediated by Na+/H+ exchanger 3 (NHE3) is important in intestinal and renal functions related to water/Na+ homeostasis. cGMP inhibits NHE3 in intact epithelia. However, unexpectedly it failed to inhibit NHE3 stably transfected in PS120 cells, even upon co-expression of cGMP-dependent protein kinase type II (cGKII). Additional co-expression of NHERF2, the tandem PDZ domain adapter protein involved in cAMP inhibition of NHE3, restored cGMP as well as cAMP inhibition, whereas NHERF1 solely restored cAMP inhibition. In vitro conditions were identified in which NHERF2 but not NHERF1 bound cGKII. The NHERF2 PDZ2 C terminus, which binds NHE3, also bound cGKII. A non-myristoylated mutant of cGKII did not support cGMP inhibition of NHE3. Although cGKI also bound NHERF2 in vitro, it did not evoke inhibition of NHE3 unless a myristoylation site was added. These results show that NHERF2, acting as a novel protein kinase G-anchoring protein, is required for cGMP inhibition of NHE3 and that cGKII must be bound both to the plasma membrane by its myristoyl anchor and to NHERF2 to inhibit NHE3.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/química , GMP Cíclico/metabolismo , Proteínas del Citoesqueleto/química , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/fisiología , Adenoviridae/genética , Animales , Sitios de Unión , Línea Celular , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo II , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Proteínas de Unión al GTP/química , Humanos , Inmunoprecipitación , Sustancias Macromoleculares/metabolismo , Mutación , Ácido Mirístico/química , Zarigüeyas , Fosfoproteínas , Unión Proteica , Proteínas Quinasas/química , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Intercambiador 3 de Sodio-Hidrógeno , Especificidad por Sustrato , Transfección
9.
Am J Physiol Cell Physiol ; 287(4): C1077-86, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15355857

RESUMEN

Diabetes mellitus is a major risk factor in the development of atherosclerosis and cardiovascular disease conditions, involving intimal injury and enhanced vascular smooth muscle cell (VSMC) migration. We report a mechanistic basis for divergences between insulin's inhibitory effects on migration of aortic VSMC from control Wistar Kyoto (WKY) rats versus Goto-Kakizaki (GK) diabetic rats. In normal WKY VSMC, insulin increased MAPK phosphatase-1 (MKP-1) expression as well as MKP-1 phosphorylation, which stabilizes it, and inhibited PDGF-mediated MAPK phosphorylation and cell migration. In contrast, basal migration was elevated in GK diabetic VSMCs, and all of insulin's effects on MKP-1 expression and phosphorylation, MAPK phosphorylation, and PDGF-stimulated migration were markedly inhibited. The critical importance of MKP-1 in insulin inhibition of VSMC migration was evident from several observations. MKP-1 small interfering RNA inhibited MKP-1 expression and abolished insulin inhibition of PDGF-induced VSMC migration. Conversely, adenoviral expression of MKP-1 decreased MAPK phosphorylation and basal migration rate and restored insulin's ability to inhibit PDGF-directed migration in GK diabetic VSMCs. Also, the proteasomal inhibitors lactacystin and MG132 partially restored MKP-1 protein levels in GK diabetic VSMCs and inhibited their migration. Furthermore, GK diabetic aortic VSMCs had reduced cGMP-dependent protein kinase Ialpha (cGK Ialpha) levels as well as insulin-dependent, but not sodium nitroprusside-dependent, stimulation of cGMP. Adenoviral expression of cGK Ialpha enhanced MKP-1 inhibition of MAPK phosphorylation and VSMC migration. We conclude that enhanced VSMC migration in GK diabetic rats is due at least in part to a failure of insulin-stimulated cGMP/cGK Ialpha signaling, MKP-1 expression, and stabilization and thus MAPK inactivation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Proteínas Inmediatas-Precoces/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Western Blotting , Proteínas Portadoras/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fosfatasa 1 de Especificidad Dual , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Proteínas Inmediatas-Precoces/efectos de los fármacos , Insulina/farmacología , Masculino , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Fosfoproteínas Fosfatasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/efectos de los fármacos , Ratas , Transducción de Señal
10.
Cardiovasc Res ; 63(3): 545-52, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15276480

RESUMEN

OBJECTIVE: Heme oxygenases (HO) are the rate-limiting enzymes in heme degradation, catalyzing the breakdown of heme to equimolar quantities of biliverdin (BV), carbon monoxide (CO), and ferrous iron. The inducible HO isoform, HO-1, confers protection against ischemia/reperfusion (I/R)-injury in the heart. We hypothesized that HO-1 and its catalytic by-products constitute an antihypertrophic signaling module in cardiac myocytes. METHODS AND RESULTS: The G protein-coupled receptor (GPCR) agonist endothelin-1 (ET-1) (30 nmol/l) stimulated a robust hypertrophic response in cardiac myocytes isolated from 1- to 3-day-old Sprague-Dawley rats, with increases in cell surface area (planimetry), sarcomere assembly (confocal laser scanning microscopy), and prepro-atrial natriuretic peptide (ANP) mRNA expression. Adenoviral overexpression of HO-1, but not beta-galactosidase, significantly inhibited ET-1 induced cardiac myocyte hypertrophy. The antihypertrophic effects of HO-1 were mimicked by BV (10 micromol/l) and the CO-releasing molecule [Ru(CO)3Cl2]2 (10 micromol/l), strongly suggesting a critical involvement of BV and CO in the antihypertrophic effects of HO-1. Both BV and CO suppressed extracellular signal-regulated kinases (ERK1/ERK2) and p38 mitogen-activated protein kinase (MAPK) activation by ET-1 stimulation. Moreover, BV and CO inhibited the prohypertrophic calcineurin/NFAT pathway. This inhibition occurred upstream from calcineurin because BV and CO inhibited NFAT activation in response to ET-1 stimulation but not in response to adenoviral expression of a constitutively active calcineurin mutant. Upstream-inhibition of the calcineurin/NFAT pathway by CO occurred independent from cGMP and cGMP-dependent protein kinase type I (PKG I). CONCLUSIONS: Heme oxygenase-1 and its catalytic by-products, BV and CO, constitute a novel antihypertrophic signaling pathway in cardiac myocytes. Biliverdin and CO inhibition of MAPKs and calcineurin/NFAT signaling provides a mechanistic framework how heme degradation products may promote their antihypertrophic effects.


Asunto(s)
Calcineurina/metabolismo , Proteínas de Unión al ADN/metabolismo , Hemo Oxigenasa (Desciclizante)/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteínas Nucleares , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Biliverdina/farmacología , Monóxido de Carbono/farmacología , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Endotelina-1/farmacología , Inhibidores Enzimáticos/farmacología , Hemo-Oxigenasa 1 , Miocitos Cardíacos/efectos de los fármacos , Factores de Transcripción NFATC , Ratas , Ratas Sprague-Dawley
11.
J Biol Chem ; 279(38): 40026-34, 2004 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-15218025

RESUMEN

cGMP-dependent protein kinase (cGK) forms encoded by the dg2 (for) gene are implicated in behavior and epithelial transport in Drosophila melanogaster. Here, we provide the first biochemical characterization and cellular localization of cGKs encoded by the major transcripts of dg2: dg2P1 and dg2P2. cGMP stimulates kinase activity of DG2P1 (EC(50): 0.13 +/- 0.039 microm) and DG2P2 (EC(50): 0.32 +/- 0.14 microm) in Malpighian tubule and S2 cell extracts. DG2P1 and DG2P2 are magnesium-requiring enzymes and were inhibited by 10 and 100 microm of a cGK inhibitor, 8-(4-chlorophenylthio)guanosine-3',5'-cyclic monophosphorothioate, Rp isomer; whereas DG1, the cGK encoded by the D. melanogaster dg1 gene, was unaffected. DG2P1 and DG2P2 were localized in the plasma membrane in S2 cells, whereas DG1 was localized in the cytosol. The D. melanogaster fluid-transporting Malpighian tubule was used as an organotypic model to analyze cGK localization and function in vivo. Targeted expression of DG2P2, DG2P1, and DG1 in tubule cells via the UAS/GAL4 system in transgenic flies revealed differential localization of all three cGKs in vivo: DG2P2 expression at the apical membrane; DG2P1 expression at both the apical and basolateral membranes; and DG1 expression at the basolateral membrane and in the cytosol. Transgenic tubules for all three cGKs displayed enhanced cGK activity compared with wild-type tubules. The physiological impact of targeted expression of individual cGKs in tubule principal cells was assessed by measuring basal and stimulated rates of fluid transport. DG1 expression greatly enhanced fluid transport by the tubule in response to exogenous cGMP, whereas DG2P2 expression significantly increased fluid transport in response to the nitridergic neuropeptide, capa-1. Thus, dg2-encoded proteins are bona fide cGKs, which have differential roles in epithelial fluid transport, as assessed by in vivo studies. Furthermore, a novel epithelial role is suggested for DG1, which is considerably more responsive to cGMP than to capa-1 stimulation.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila melanogaster , Células Epiteliales/enzimología , Riñón/metabolismo , Túbulos de Malpighi/enzimología , Técnicas de Cultivo de Órganos , Fenotipo
12.
Genomics ; 83(4): 577-87, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15028281

RESUMEN

Cardiac fibroblasts regulate tissue repair and remodeling in the heart. To quantify transcript levels in these cells we performed a comprehensive gene expression study using serial analysis of gene expression (SAGE). Among 110,169 sequenced tags we could identify 30,507 unique transcripts. A comparison of SAGE data from cardiac fibroblasts with data derived from total mouse heart revealed a number of fibroblast-specific genes. Cardiac fibroblasts expressed a specific collection of collagens, matrix proteins and metalloproteinases, growth factors, and components of signaling pathways. The NO/cGMP signaling pathway was represented by the mRNAs for alpha(1) and beta(1) subunits of guanylyl cyclase, cGMP-dependent protein kinase type I (cGK I), and, interestingly, the G-kinase-anchoring protein GKAP42. The expression of cGK I was verified by RT-PCR and Western blot. To establish a functional role for cGK I in cardiac fibroblasts we studied its effect on cell proliferation. Selective activation of cGK I with a cGMP analog inhibited the proliferation of serum-stimulated cardiac fibroblasts, which express cGK I, but not higher passage fibroblasts, which contain no detectable cGK I. Currently, our data suggest that cGK I mediates the inhibitory effects of the NO/cGMP pathway on cardiac fibroblast growth. Furthermore the SAGE library of transcripts expressed in cardiac fibroblasts provides a basis for future investigations into the pathological regulatory mechanisms underlying cardiac fibrosis.


Asunto(s)
GMP Cíclico/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Miocardio/metabolismo , Óxido Nítrico/metabolismo , Proteoma , Proteínas Adaptadoras Transductoras de Señales , Animales , Northern Blotting , Proteínas Portadoras/metabolismo , División Celular , Matriz Extracelular/metabolismo , Fibrosis/patología , Guanilato Ciclasa/metabolismo , Ratones , ARN Mensajero/metabolismo , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa , Receptores de Péptidos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
13.
Cardiovasc Res ; 60(2): 268-77, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14613856

RESUMEN

OBJECTIVE: Calcium entry via the L-type Ca(2+) channel (LTCC) is crucial for excitation-contraction (EC) coupling and activation of Ca(2+)-dependent signal transduction pathways in cardiac myocytes. Both nitric oxide (NO), signaling via cGMP, and acetylcholine, signaling via the muscarinic receptor, have been identified as negative regulators of beta-adrenoreceptor-stimulated LTCC activity in cardiac myocytes. METHODS: To examine the potential role of cGMP-dependent protein kinase type I (PKG I) in the inhibitory effects of NO/cGMP and the muscarinic receptor on LTCC activity, we generated transgenic (TG) mice overexpressing PKG I selectively in cardiac myocytes under the control of the alpha-myocin heavy chain promoter. Single LTCC-gating properties were assessed in isolated ventricular myocytes from adult wild-type (WT) and PKG I transgenic (TG) mice. RESULTS: Basal LTCC activity (peak average current, mean open probability, mean availability) was significantly decreased by the nitric oxide donor DEA-NO (0.1 micromol/l) and the cGMP-analog 8-Br-cGMP (1 mmol/l) in TG but not in WT cardiac myocytes. Conversely, muscarinic (carbachol, 1 micromol/l) stimulation had no significant effect on basal LTCC activity in either WT or TG cardiac myocytes. beta-Adrenergic stimulation with isoproterenol (1 micromol/l) increases single LTCC activity in WT and TG cardiac myocytes to the same extent. The inhibitory effects of DEA-NO and 8-Br-cGMP on isoproterenol activation of the LTCC current were significantly enhanced in TG as compared to WT cardiac myocytes. By contrast, carbachol inhibition of isoproterenol-stimulated single LTCC activity was not enhanced in TG cardiac myocytes. CONCLUSION: Transgenic overexpression of PKG I augments NO/cGMP inhibition but not muscarinic inhibition of single LTCC activity, indicating that PKG I is a downstream target for NO/cGMP, but not the muscarinic receptor in adult cardiac myocytes.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Miocitos Cardíacos/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Canales de Calcio Tipo L/efectos de los fármacos , Carbacol/farmacología , Cardiotónicos/farmacología , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Hidrazinas/farmacología , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxidos de Nitrógeno
14.
Circ Res ; 93(10): 907-16, 2003 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-14615494

RESUMEN

Signaling cascades initiated by nitric oxide (NO) and natriuretic peptides (NPs) play an important role in the maintenance of cardiovascular homeostasis. It is currently accepted that many effects of these endogenous signaling molecules are mediated via stimulation of guanylyl cyclases and intracellular production of the second messenger cGMP. Indeed, cGMP-elevating drugs like glyceryl trinitrate have been used for more than 100 years to treat cardiovascular diseases. However, the molecular mechanisms of NO/NP signaling downstream of cGMP are not completely understood. Recent in vitro and in vivo evidence identifies cGMP-dependent protein kinases (cGKs) as major mediators of cGMP signaling in the cardiovascular system. In particular, the analysis of conventional and conditional knockout mice indicates that cGKs are critically involved in regulating vascular remodeling and thrombosis. Thus, cGKs may represent novel drug targets for the treatment of human cardiovascular disorders.


Asunto(s)
Sistema Cardiovascular/enzimología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Ratones Transgénicos/fisiología , Animales , Plaquetas/fisiología , Presión Sanguínea/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Humanos , Riñón/fisiología , Ratones , Transducción de Señal/fisiología , Sistema Vasomotor/fisiología
16.
Br J Pharmacol ; 140(7): 1227-36, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14609817

RESUMEN

1. C-type natriuretic peptide (CNP) and its receptor guanylyl cyclase (GC-B) are expressed in the heart and modulate cardiac contractility in a cGMP-dependent manner. Since the distal cellular signalling pathways remain unclear, we evaluated the peptide effects on cardiac function and calcium regulation in wild-type (WT) and transgenic mice with cardiac overexpression of cGMP-dependent protein kinase I (PKG ITG). 2. In isolated, perfused working WT hearts, CNP (10 nm) provoked an immediate increase in the maximal rates of contraction and relaxation, a small increase in the left ventricular systolic pressure and a decrease in the time of relaxation. These changes in cardiac function were accompanied by a marked increase in the levels of Ser16-phosphorylated phospholamban (PLB). 3. In PKG ITG hearts, the effects of CNP on cardiac contractility and relaxation as well as on PLB phosphorylation were markedly enhanced. 4. CNP increased cell shortening and systolic Cai2+ levels, and accelerated Cai2+ decay in isolated, Indo-1/AM-loaded WT cardiomyocytes, and these effects were enhanced in PKG I-overexpressing cardiomyocytes. 5. 8-pCPT-cGMP, a membrane-permeable PKG activator, mimicked the contractile and molecular actions of CNP, the effects again being more pronounced in PKG ITG hearts. In contrast, the cardiac responses to beta-adrenergic stimulation were not different between genotypes. 6. Taken together, our data indicate that PKG I is a downstream target activated by the CNP/GC-B/cGMP-signalling pathway in cardiac myocytes. cGMP/PKG I-stimulated phosphorylation of PLB and subsequent activation of the sarcoplasmic reticulum Ca2+ pump appear to mediate the positive inotropic and lusitropic responses to CNP.


Asunto(s)
Calcio/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Contracción Miocárdica/efectos de los fármacos , Natriuréticos/farmacología , Péptido Natriurético Tipo-C/farmacología , Animales , Western Blotting , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Ventrículos Cardíacos/citología , Humanos , Cinética , Ratones , Ratones Transgénicos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología
17.
Circulation ; 107(10): 1424-32, 2003 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-12642365

RESUMEN

BACKGROUND: In chronic heart failure, myocardial expression of the inducible isoform of nitric oxide (NO) synthase (NOS2) is enhanced, leading to a sustained production of NO. We postulated that NO modulates expression of genes in cardiac myocytes that may be functionally important in the context of cardiac hypertrophy and failure. METHODS AND RESULTS: As revealed by cDNA expression array analyses, the NO donor SNAP, which has been shown previously to inhibit agonist-induced cardiac myocyte hypertrophy, downregulates expression of the cytoskeleton-associated muscle LIM protein (MLP) in endothelin-1 (ET-1)-stimulated neonatal rat cardiac myocytes. Northern blotting and immunoblotting experiments confirmed this finding and established that SNAP negatively controls MLP mRNA (-49%, P<0.01) and protein (-52%, P<0.01) abundance in ET-1-treated cardiomyocytes via cGMP-dependent protein kinase and superoxide/peroxynitrite-dependent signaling pathways. Treatment of cardiac myocytes with IL-1beta and IFN-gamma downregulated MLP expression levels via induction of NOS2. Moreover, expression levels of NOS2 and MLP were inversely correlated in the failing human heart, indicating that NOS2 may regulate MLP abundance in vitro and in vivo. Antisense oligonucleotides were used to explore the functional consequences of reduced MLP expression levels in cardiac myocytes. Like SNAP, antisense downregulation of MLP protein expression (-52%, P<0.01) blunted the increases in protein synthesis, cell size, and sarcomere organization in response to ET-1 stimulation. Conversely, overexpression of MLP augmented cell size and sarcomere organization in cardiac myocytes. CONCLUSIONS: NO negatively controls MLP expression in cardiac myocytes. Because MLP is necessary and sufficient for hypertrophy and sarcomere assembly, MLP downregulation may restrain hypertrophic growth in pathophysiological situations with increased cardiac NO production.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Óxido Nítrico/fisiología , Animales , Tamaño de la Célula , Células Cultivadas , GMP Cíclico/fisiología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Citocinas/farmacología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/fisiología , Regulación hacia Abajo , Endotelina-1/farmacología , Regulación de la Expresión Génica , Humanos , Proteínas con Dominio LIM , Proteínas Musculares/genética , Miocitos Cardíacos/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Oligonucleótidos Antisentido/genética , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Sarcómeros/ultraestructura , Superóxidos/metabolismo
18.
Proc Natl Acad Sci U S A ; 99(17): 11363-8, 2002 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-12177418

RESUMEN

Recent investigation has focused on identifying signaling pathways that inhibit cardiac hypertrophy, a major risk factor for cardiovascular morbidity and mortality. In this context, nitric oxide (NO), signaling via cGMP and cGMP-dependent protein kinase type I (PKG I), has been recognized as a negative regulator of cardiac myocyte (CM) hypertrophy. However, the underlying mechanisms are poorly understood. Here, we show that PKG I inhibits CM hypertrophy by targeting the calcineurin-NFAT signaling pathway. Calcineurin, a Ca2+-dependent phosphatase, promotes hypertrophy in part by activating NFAT transcription factors which induce expression of hypertrophic genes, including brain natriuretic peptide (BNP). Activation of PKG I by NO/cGMP in CM suppressed NFAT transcriptional activity, BNP induction, and cell enlargement in response to alpha(1)-adrenoreceptor stimulation but not in response to adenoviral expression of a Ca2+-independent, constitutively active calcineurin mutant, thus demonstrating NO-cGMP-PKG I inhibition of calcineurin-NFAT signaling upstream of calcineurin. PKG I suppressed single L-type Ca2+-channel open probability, [Ca2+]i transient amplitude, and, most importantly, L-type Ca2+-channel current-induced NFAT activation, indicating that PKG I targets Ca2+-dependent steps upstream of calcineurin. Adenoviral expression of PKG I enhanced NO/cGMP inhibitory effects upstream of calcineurin, confirming that PKG I mediates NO/cGMP inhibition of calcineurin-NFAT signaling. In CM overexpressing PKG I, NO/cGMP also suppressed BNP induction and cell enlargement but not NFAT activation elicited by constitutively active calcineurin, which is consistent with additional, NFAT-independent inhibitory effect(s) of PKG I downstream of calcineurin. Inhibition of calcineurin-NFAT signaling by PKG I provides a framework for understanding how NO inhibits cardiac myocyte hypertrophy.


Asunto(s)
Calcineurina/fisiología , Cardiomegalia/prevención & control , Cardiomegalia/fisiopatología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/análogos & derivados , Proteínas de Unión al ADN/fisiología , Miocardio/metabolismo , Proteínas Nucleares , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Animales , Animales Recién Nacidos , Inhibidores de la Calcineurina , Canales de Calcio Tipo L/fisiología , Señalización del Calcio/fisiología , Cardiomegalia/enzimología , Células Cultivadas , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Activación Enzimática , Corazón/fisiología , Ventrículos Cardíacos , Activación del Canal Iónico/fisiología , Luciferasas/genética , Luciferasas/metabolismo , Factores de Transcripción NFATC , Péptido Natriurético Encefálico/genética , Plásmidos , Probabilidad , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Tionucleótidos/farmacología , Factores de Transcripción/antagonistas & inhibidores , Transcripción Genética , Transfección
19.
Am J Physiol Cell Physiol ; 283(3): C704-13, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12176727

RESUMEN

In this study, we examined the role of insulin in the control of vascular smooth muscle cell (VSMC) migration in the normal vasculature. Platelet-derived growth factor (PDGF) increased VSMC migration, which was inhibited by pretreatment with insulin in a dose-dependent manner. Insulin also caused a 60% decrease in PDGF-stimulated mitogen-activated protein kinase (MAPK) phosphorylation and activation. Insulin inhibition of MAPK was accompanied by a rapid induction of MAPK phosphatase (MKP-1), which inactivates MAPKs by dephosphorylation. Pretreatment with inhibitors of the nitric oxide (NO)/cGMP pathway, blocked insulin-induced MKP-1 expression and restored PDGF-stimulated MAPK activation and migration. In contrast, adenoviral infection of VSMCs with MKP-1 or cGMP-dependent protein kinase Ialpha (cGK Ialpha), the downstream effector of cGMP signaling, blocked the activation of MAPK and prevented PDGF-directed VSMC migration. Expression of antisense MKP-1 RNA prevented insulin's inhibitory effect and restored PDGF-directed VSMC migration and MAPK phosphorylation. We conclude that insulin inhibition of VSMC migration may be mediated in part by NO/cGMP/cGK Ialpha induction of MKP-1 and consequent inactivation of MAPKs.


Asunto(s)
Proteínas de Ciclo Celular , GMP Cíclico/análogos & derivados , Proteínas Inmediatas-Precoces/metabolismo , Insulina/farmacología , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Fosfoproteínas Fosfatasas , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Tirosina Fosfatasas/metabolismo , Animales , Becaplermina , Movimiento Celular/efectos de los fármacos , Células Cultivadas , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfatasa 1 de Especificidad Dual , Inhibidores Enzimáticos/farmacología , Humanos , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/genética , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas c-sis , ARN sin Sentido/biosíntesis , ARN sin Sentido/farmacología , Ratas , Ratas Endogámicas WKY , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
20.
Biochem Biophys Res Commun ; 293(5): 1438-44, 2002 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-12054676

RESUMEN

cGMP- and cAMP-dependent protein kinases (cGK I, cGK II, and cAK) are important mediators of many signaling pathways that increase cyclic nucleotide concentrations and ultimately phosphorylation of substrates vital to cellular functions. Here we demonstrate a novel mRNA splice isoform of cGK II arising from alternative 5' splicing within exon 11. The novel splice variant encodes a protein (cGK II Delta(441-469)) lacking 29 amino acids of the cGK II Mg-ATP-binding/catalytic domain, including the conserved glycine-rich loop consensus motif Gly-x-Gly-x-x-Gly-x-Val which interacts with ATP in the protein kinase family of enzymes. cGK II Delta(441-469) has no intrinsic enzymatic activity itself, however, it antagonizes cGK II and cGK I, but not cAK. Thus, the activation and cellular functions of cGK II may be determined not only by intracellular cGMP levels but also by alternative splicing which may regulate the balance of expression of cGK II versus its own inhibitor, cGK II Delta(441-469).


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/química , GMP Cíclico/metabolismo , Células 3T3 , Regiones no Traducidas 5' , Adenosina Trifosfato/metabolismo , Empalme Alternativo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Western Blotting , Dominio Catalítico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteína Quinasa Dependiente de GMP Cíclico Tipo II , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Exones , Vectores Genéticos , Glicina/química , Humanos , Ratones , Microscopía Fluorescente , Modelos Genéticos , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína , ARN/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Distribución Tisular , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...