Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 320(6): H2188-H2200, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33861144

RESUMEN

The interaction of phospholamban (PLB) and the sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) is a key regulator of cardiac contractility and a therapeutic target in heart failure (HF). PLB-mediated increases in SERCA2a activity improve cardiac function and HF. Clinically, this mechanism can only be exploited by a general activation of the proteinkinase A (PKA), which is associated with side effects and adverse clinical outcomes. A selective interference of the PLB-SERCA2a interaction is desirable but will require novel tools that allow for an integrated assessment of this interaction under both physiological and pathophysiological conditions. A circularly permutated green fluorescent protein (cpGFP) was interposed between SERCA2a and PLB to result into a single SERCA2a-cpGFP-PLB recombinant protein (SGP). Expression, phosphorylation, fluorescence, and function of SGP were evaluated. Expression of SGP-cDNA results in a functional recombinant protein at the predicted molecular weight. The PLB domain of SGP retains its ability to polymerize and can be phosphorylated by PKA activation. This increases the fluorescent yield of SGP by between 10% and 165% depending on cell line and conditions. In conclusion, a single recombinant fusion protein that combines SERCA2a, a circularly permutated green fluorescent protein, and PLB can be expressed in cells and can be phosphorylated at the PLB domain that markedly increases the fluorescence yield. SGP is a novel cellular SERCA2a-PLB interaction monitor.NEW & NOTEWORTHY This study describes the design and characterization of a novel biosensor that can visualize the interaction of SERCA2a and phospholamban (PLB). The biosensor combines SERCA2a, a circularly permutated green fluorescent protein, and PLB into one recombinant protein (SGP). Proteinkinase A activation results in phosphorylation of the PLB domain and is associated with a marked increase in the fluorescence yield to allow for real-time monitoring of the SERCA2a and PLB interaction in cells.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , ADN Complementario , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Ratas , Proteínas Recombinantes de Fusión , Proteínas Recombinantes , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Transfección
2.
J Cell Biochem ; 120(8): 13783-13791, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30938854

RESUMEN

The chimeric DnaJ-PKAc enzymeresulting from an approximately 400-kb deletion of chromosome 19 is a primary contributor to the oncogenic transformation that occurs in fibrolamellar hepatocellular carcinoma, also called fibrolamellar carcinoma (FLC). This oncogenic deletion juxtaposes exon 1 of the DNAJB1 heat shock protein gene with exon 2 of the PRKACA gene encoding the protein kinase A catalytic subunit, resulting in DnaJ-PKAc fusion under the transcriptional control of the DNAJB1 promoter. The expression of DnaJ-PKAc is approximately 10 times that of wild-type (wt) PKAc catalytic subunits, causing elevated and dysregulated kinase activity that contributes to oncogenic transformation. In normal cells, PKAc activity is regulated by a group of endogenous proteins, termed protein kinase inhibitors (PKI) that competitively inhibit PKAc and assist with the nuclear export of the enzyme. Currently, it is scarcely known whether interactions with PKI are perturbed in DnaJ-PKAc. In this report, we survey existing data sets to assess the expression levels of the various PKI isoforms that exist in humans to identify those that are candidates to encounter DnaJ-PKAc in both normal liver and FLC tumors. We then compare inhibition profiles of wtPKAc and DnaJ-PKAc against PKI and demonstrate that extensive structural homology in the active site clefts of the two enzymes confers similar kinase activities and inhibition by full-length PKI and PKI-derived peptides.


Asunto(s)
Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico , Proteínas del Choque Térmico HSP40 , Proteínas de Fusión Oncogénica , Péptidos/química , Inhibidores de Proteínas Quinasas/química , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/antagonistas & inhibidores , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/química , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Proteínas del Choque Térmico HSP40/antagonistas & inhibidores , Proteínas del Choque Térmico HSP40/química , Proteínas del Choque Térmico HSP40/genética , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética
3.
J Biol Chem ; 293(43): 16791-16802, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30206122

RESUMEN

The type I cGMP-dependent protein kinase (PKG I) is an essential regulator of vascular tone. It has been demonstrated that the type Iα isoform can be constitutively activated by oxidizing conditions. However, the amino acid residues implicated in this phenomenon are not fully elucidated. To investigate the molecular basis for this mechanism, we studied the effects of oxidation using recombinant WT, truncated, and mutant constructs of PKG I. Using an in vitro assay, we observed that oxidation with hydrogen peroxide (H2O2) resulted in constitutive, cGMP-independent activation of PKG Iα. PKG Iα C42S and a truncation construct that does not contain Cys-42 (Δ53) were both constitutively activated by H2O2 In contrast, oxidation of PKG Iα C117S maintained its cGMP-dependent activation characteristics, although oxidized PKG Iα C195S did not. To corroborate these results, we also tested the effects of our constructs on the PKG Iα-specific substrate, the large conductance potassium channel (KCa 1.1). Application of WT PKG Iα activated by either cGMP or H2O2 increased the open probabilities of the channel. Neither cGMP nor H2O2 activation of PKG Iα C42S significantly increased channel open probabilities. Moreover, cGMP-stimulated PKG Iα C117S increased KCa 1.1 activity, but this effect was not observed under oxidizing conditions. Finally, we observed that PKG Iα C42S caused channel flickers, indicating dramatically altered KCa 1.1 channel characteristics compared with channels exposed to WT PKG Iα. Cumulatively, these results indicate that constitutive activation of PKG Iα proceeds through oxidation of Cys-117 and further suggest that the formation of a sulfur acid is necessary for this phenotype.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico/metabolismo , Cisteína/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/química , Cisteína/química , Modelos Moleculares , Óxido Nítrico/metabolismo , Oxidación-Reducción , Fosforilación , Conformación Proteica , Homología de Secuencia
4.
J Biol Chem ; 293(21): 7916-7929, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29602907

RESUMEN

The type I cGMP-dependent protein kinases (PKG I) serve essential physiological functions, including smooth muscle relaxation, cardiac remodeling, and platelet aggregation. These enzymes form homodimers through their N-terminal dimerization domains, a feature implicated in regulating their cooperative activation. Previous investigations into the activation mechanisms of PKG I isoforms have been largely influenced by structures of the cAMP-dependent protein kinase (PKA). Here, we examined PKG Iα activation by cGMP and cAMP by engineering a monomeric form that lacks N-terminal residues 1-53 (Δ53). We found that the construct exists as a monomer as assessed by whole-protein MS, size-exclusion chromatography, and small-angle X-ray scattering (SAXS). Reconstruction of the SAXS 3D envelope indicates that Δ53 has a similar shape to the heterodimeric RIα-C complex of PKA. Moreover, we found that the Δ53 construct is autoinhibited in its cGMP-free state and can bind to and be activated by cGMP in a manner similar to full-length PKG Iα as assessed by surface plasmon resonance (SPR) spectroscopy. However, we found that the Δ53 variant does not exhibit cooperative activation, and its cyclic nucleotide selectivity is diminished. These findings support a model in which, despite structural similarities, PKG Iα activation is distinct from that of PKA, and its cooperativity is driven by in trans interactions between protomers.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/antagonistas & inhibidores , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Animales , Bovinos , Cristalografía por Rayos X , Activación Enzimática , Humanos , Ratones , Modelos Moleculares , Fosforilación , Unión Proteica , Ratas , Dispersión del Ángulo Pequeño , Homología de Secuencia
5.
Chem Biol ; 22(12): 1653-61, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26687482

RESUMEN

PKG is a multifaceted signaling molecule and potential pharmaceutical target due to its role in smooth muscle function. A helix identified in the structure of the regulatory domain of PKG Iα suggests a novel architecture of the holoenzyme. In this study, a set of synthetic peptides (S-tides), derived from this helix, was found to bind to and activate PKG Iα in a cyclic guanosine monophosphate (cGMP)-independent manner. The most potent S-tide derivative (S1.5) increased the open probability of the potassium channel KCa1.1 to levels equivalent to saturating cGMP. Introduction of S1.5 to smooth muscle cells in isolated, endothelium-denuded cerebral arteries through a modified reversible permeabilization procedure inhibited myogenic constriction. In contrast, in endothelium-intact vessels S1.5 had no effect on myogenic tone. This suggests that PKG Iα activation by S1.5 in vascular smooth muscle would be sufficient to inhibit augmented arterial contractility that frequently occurs following endothelial damage associated with cardiovascular disease.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , GMP Cíclico , Diseño de Fármacos , Biblioteca de Péptidos , Péptidos/farmacología , Animales , Dicroismo Circular , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/aislamiento & purificación , Activación Enzimática/efectos de los fármacos , Activadores de Enzimas/síntesis química , Activadores de Enzimas/farmacología , Microscopía Confocal , Músculo Liso Vascular/efectos de los fármacos , Péptidos/síntesis química , Isoformas de Proteínas/aislamiento & purificación , Ratas
6.
Am J Pathol ; 185(10): 2790-804, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26261085

RESUMEN

Ulcerative colitis (UC) belongs to inflammatory bowel disorders, a group of gastrointestinal disorders that can produce serious recurring diarrhea in affected patients. The mechanism for UC- and inflammatory bowel disorder-associated diarrhea is not well understood. The cystic fibrosis transmembrane-conductance regulator (CFTR) chloride channel plays an important role in fluid and water transport across the intestinal mucosa. CFTR channel function is regulated in a compartmentalized manner through the formation of CFTR-containing macromolecular complexes at the plasma membrane. In this study, we demonstrate the involvement of a novel macromolecular signaling pathway that causes diarrhea in UC. We found that a nitric oxide-producing enzyme, inducible nitric oxide synthase (iNOS), is overexpressed under the plasma membrane and generates compartmentalized cGMP in gut epithelia in UC. The scaffolding protein Na(+)/H(+) exchanger regulatory factor 2 (NHERF2) bridges iNOS with CFTR, forming CFTR-NHERF2-iNOS macromolecular complexes that potentiate CFTR channel function via the nitric oxide-cGMP pathway under inflammatory conditions both in vitro and in vivo. Potential disruption of these complexes in Nherf2(-/-) mice may render them more resistant to CFTR-mediated secretory diarrhea than Nherf2(+/+) mice in murine colitis models. Our study provides insight into the mechanism of pathophysiologic occurrence of diarrhea in UC and suggests that targeting CFTR and CFTR-containing macromolecular complexes will ameliorate diarrheal symptoms and improve conditions associated with inflammatory bowel disorders.


Asunto(s)
Membrana Celular/metabolismo , Colitis Ulcerosa/metabolismo , GMP Cíclico/metabolismo , Diarrea/metabolismo , Animales , Células Cultivadas , Colitis Ulcerosa/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/metabolismo , Transducción de Señal/fisiología , Intercambiadores de Sodio-Hidrógeno/metabolismo
7.
Nature ; 519(7544): 472-6, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25799991

RESUMEN

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , Cardiomegalia/enzimología , Cardiomegalia/metabolismo , GMP Cíclico/metabolismo , Óxido Nítrico , 3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , 3',5'-AMP Cíclico Fosfodiesterasas/deficiencia , 3',5'-AMP Cíclico Fosfodiesterasas/genética , Animales , Estenosis de la Válvula Aórtica/complicaciones , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/etiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Musculares/enzimología , Miocardio/enzimología , Péptidos Natriuréticos/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/uso terapéutico , Presión , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico , Regulación hacia Arriba
8.
Methods Mol Biol ; 1020: 131-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23709030

RESUMEN

Real-time and noninvasive imaging of intracellular second messengers in mammalian cells, while -preserving their in vivo phenotype, requires biosensors of exquisite constitution. Here we provide the methodology for utilizing the single wavelength cGMP-biosensor δ-FlincG in aortic vascular smooth muscle cells.


Asunto(s)
GMP Cíclico/metabolismo , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Adenoviridae/genética , Animales , Separación Celular/métodos , GMP Cíclico/química , Vectores Genéticos/genética , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Miocitos del Músculo Liso/citología , Ratas , Transducción Genética
9.
Biochim Biophys Acta ; 1834(7): 1346-51, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23416533

RESUMEN

For over three decades the isozymes of cGMP-dependent protein kinase (PKG) have been studied using an array of biochemical and biophysical techniques. When compared to its closest cousin, cAMP-dependent protein kinase (PKA), these studies revealed a set of identical domain types, yet containing distinct, sequence-specific features. The recently solved structure of the PKG regulatory domain showed the presence of the switch helix (SW), a novel motif that promotes the formation of a domain-swapped dimer in the asymmetric unit. This dimer is mediated by the interaction of a knob motif on the C-terminal locus of the SW, with a hydrophobic nest on the opposing protomer. This nest sits adjacent to the cGMP binding pocket of the B-site. Priming of this site by cGMP may influence the geometry of the hydrophobic nest. Moreover, this unique interaction may have wide implications for the architecture of the inactive and active forms of PKG. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases (2012).


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/química , GMP Cíclico/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Secuencia de Aminoácidos , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Homología de Secuencia de Aminoácido
10.
Front Pharmacol ; 3: 130, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22807915

RESUMEN

Nitric oxide (NO) is a potent dilator of vascular smooth muscle (VSM) by modulating intracellular cGMP ([cGMP](i)) through the binding and activation of receptor guanylyl cylases (sGC). The kinetic relationship of NO and sGC, as well as the subsequent regulation of [cGMP](i) and its effects on blood vessel vasodilation, is largely unknown. In isolated VSM cells exposed to both pulsed and clamped NO we observed transient and sustained increases in [cGMP](i), with sub-nanomolar sensitivity to NO (EC(50) = 0.28 nM). Through the use of pharmacological inhibitors of sGC, PDE5, and PKG, a comprehensive VSM-specific modeling algorithm was constructed to elucidate the concerted activity profiles of sGC, PDE5, phosphorylated PDE5, and PDE1 in the maintenance of [cGMP](i). In small pressure-constricted arteries of the resistance vasculature we again observed both transient and sustained relaxations upon delivery of pulsed and clamped NO, while maintaining a similarly high sensitivity to NO (EC(50) = 0.42 nM). Our results propose an intricate dependency of the messengers and enzymes involved in cGMP homeostasis, and vasodilation in VSM. Particularly, the high sensitivity of sGC to NO in primary tissue indicates how small changes in the concentrations of NO, irrespective of the form of NO delivery, can have significant effects on the dynamic regulation of vascular tone.

11.
J Cell Biochem ; 113(1): 39-48, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21866565

RESUMEN

The cyclic-AMP-dependent protein kinase A (PKA) regulates processes such as cell proliferation and migration following activation of growth factor receptor tyrosine kinases (RTKs), yet the signaling mechanisms that link PKA with growth factor receptors remain largely undefined. Here we report that RTKs can directly modulate the function of the catalytic subunit of PKA (PKA-C) through post-translational modification. In vitro kinase assays revealed that both the epidermal growth factor and platelet derived growth factor receptors (EGFR and PDGFR, respectively) tyrosine phosphorylate PKA-C. Mass spectrometry identified tyrosine 330 (Y330) as a receptor-mediated phosphorylation site and mutation of Y330 to phenylalanine (Y330F) all but abolished the RTK-mediated phosphorylation of PKA-C in vitro. Y330 resides within a conserved region at the C-terminal tail of PKA-C that allosterically regulates enzymatic activity. Therefore, the effect of phosphorylation at Y330 on the activity of PKA-C was investigated. The K(m) for a peptide substrate was markedly decreased when PKA-C subunits were tyrosine phosphorylated by the receptors as compared to un-phosphorylated controls. Importantly, tyrosine-phosphorylated PKA-C subunits were detected in cells stimulated with EGF, PDGF, and Fibroblast growth factor 2 (FGF2) and in fibroblasts undergoing PDGF-mediated chemotaxis. These results demonstrate a direct, functional interaction between RTKs and PKA-C and identify tyrosine phosphorylation as a novel mechanism for regulating PKA activity.


Asunto(s)
Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Células COS , Línea Celular , Proliferación Celular , Chlorocebus aethiops , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Receptores ErbB/metabolismo , Ratones , Células 3T3 NIH , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Receptores de Factores de Crecimiento/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Tirosina/metabolismo
12.
Basic Res Cardiol ; 106(6): 1023-39, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22012077

RESUMEN

Cardiac fibroblasts become activated and differentiate to smooth muscle-like myofibroblasts in response to hypertension and myocardial infarction (MI), resulting in extracellular matrix (ECM) remodeling, scar formation and impaired cardiac function. cAMP and cGMP-dependent signaling have been implicated in cardiac fibroblast activation and ECM synthesis. Dysregulation of cyclic nucleotide phosphodiesterase (PDE) activity/expression is also associated with various diseases and several PDE inhibitors are currently available or in development for treating these pathological conditions. The objective of this study is to define and characterize the specific PDE isoform that is altered during cardiac fibroblast activation and functionally important for regulating myofibroblast activation and ECM synthesis. We have found that Ca(2+)/calmodulin-stimulated PDE1A isoform is specifically induced in activated cardiac myofibroblasts stimulated by Ang II and TGF-ß in vitro as well as in vivo within fibrotic regions of mouse, rat, and human diseased hearts. Inhibition of PDE1A function via PDE1-selective inhibitor or PDE1A shRNA significantly reduced Ang II or TGF-ß-induced myofibroblast activation, ECM synthesis, and pro-fibrotic gene expression in rat cardiac fibroblasts. Moreover, the PDE1 inhibitor attenuated isoproterenol-induced interstitial fibrosis in mice. Mechanistic studies revealed that PDE1A modulates unique pools of cAMP and cGMP, predominantly in perinuclear and nuclear regions of cardiac fibroblasts. Further, both cAMP-Epac-Rap1 and cGMP-PKG signaling was involved in PDE1A-mediated regulation of collagen synthesis. These results suggest that induction of PDE1A plays a critical role in cardiac fibroblast activation and cardiac fibrosis, and targeting PDE1A may lead to regression of the adverse cardiac remodeling associated with various cardiac diseases.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Miocardio/metabolismo , Remodelación Ventricular/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Western Blotting , Matriz Extracelular/patología , Fibrosis/patología , Fibrosis/prevención & control , Humanos , Inmunohistoquímica , Isoenzimas , Isoproterenol/farmacología , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transfección
13.
Structure ; 19(9): 1317-27, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21893290

RESUMEN

The cGMP-dependent protein kinase (PKG) serves as an integral component of second messenger signaling in a number of biological contexts including cell differentiation, memory, and vasodilation. PKG is homodimeric and large conformational changes accompany cGMP binding. However, the structure of PKG and the molecular mechanisms associated with protomer communication following cGMP-induced activation remain unknown. Here, we report the 2.5 Å crystal structure of a regulatory domain construct (aa 78-355) containing both cGMP binding sites of PKG Iα. A distinct and segregated architecture with an extended central helix separates the two cGMP binding domains. Additionally, a previously uncharacterized helical domain (switch helix) promotes the formation of a hydrophobic interface between protomers. Mutational disruption of this interaction in full-length PKG implicates the switch helix as a critical site of dimer communication in PKG biology. These results offer new structural insight into the mechanism of allosteric PKG activation.


Asunto(s)
Sitio Alostérico , Proteínas Quinasas Dependientes de GMP Cíclico/química , Regulación Alostérica , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Bovinos , Secuencia Conservada , Cristalografía por Rayos X , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Pruebas de Enzimas , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
14.
Proc Natl Acad Sci U S A ; 107(51): 22060-5, 2010 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-21135206

RESUMEN

Nitric oxide (NO) functions as a diffusible transmitter in most tissues of the body and exerts its effects by binding to receptors harboring a guanylyl cyclase transduction domain, resulting in cGMP accumulation in target cells. Despite its widespread importance, very little is known about how this signaling pathway operates at physiological NO concentrations and in real time. To address these deficiencies, we have exploited the properties of a novel cGMP biosensor, named δ-FlincG, expressed in cells containing varying mixtures of NO-activated guanylyl cyclase and cGMP-hydrolyzing phosphodiesterase activity. Responsiveness to NO, signifying a physiologically relevant rise in cGMP to 30 nM or more, was seen at concentrations as low as 1 pM, making cells by far the most sensitive NO detectors yet encountered. Even cells coexpressing phosphodiesterase-5, a cGMP-activated isoform found in many NO target cells, responded to NO in concentrations as low as 10 pM. The dynamics of NO capture and signal transduction was revealed by administering timed puffs of NO from a local pipette. A puff lasting only 100 ms, giving a calculated peak intracellular NO concentration of 23 pM, was detectable. The results could be encapsulated in a quantitative model of cellular NO-cGMP signaling, which recapitulates the NO responsiveness reported previously from crude cGMP measurements on native cells, and which explains how NO is able to exert physiological effects at extremely low concentrations, when only a tiny proportion of its receptors would be occupied.


Asunto(s)
GMP Cíclico/metabolismo , Óxido Nítrico/metabolismo , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Transducción de Señal/fisiología , Animales , Técnicas Biosensibles/métodos , Bovinos , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Óxido Nítrico/farmacología , Transducción de Señal/efectos de los fármacos
15.
Biochim Biophys Acta ; 1804(9): 1857-68, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20406699

RESUMEN

INTRODUCTION: Type I cGMP-dependent protein kinase (PKGIalpha) belongs to the family of cyclic nucleotide-dependent protein kinases and is one of the main effectors of cGMP. PKGIalpha is involved in regulation of cardiac contractility, vasorelaxation, and blood pressure; hence, the development of potent modulators of PKGIalpha would lead to advances in the treatment of a variety of cardiovascular diseases. AIM: Representatives of ARC-type compounds previously characterized as potent inhibitors and high-affinity fluorescent probes of PKA catalytic subunit (PKAc) were tested towards PKGIalpha to determine that ARCs could serve as activity regulators and sensors for the latter protein kinase both in vitro and in complex biological systems. RESULTS: Structure-activity profiling of ARCs with PKGIalpha in vitro demonstrated both similarities as well as differences to corresponding profiling with PKAc, whereas ARC-903 and ARC-668 revealed low nanomolar displacement constants and inhibition IC(50) values with both cyclic nucleotide-dependent kinases. The ability of ARC-based fluorescent probes to penetrate cell plasma membrane was demonstrated in the smooth muscle tissue of rat cerebellum isolated arteries, and the compound with the highest affinity in vitro (ARC-903) showed also potential for in vivo applications, fully abolishing the PKG1alpha-induced vasodilation.


Asunto(s)
Adenosina/química , Arginina/química , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes , Músculo Liso Vascular/efectos de los fármacos , Animales , Arterias Cerebrales/citología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/metabolismo , GMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Inhibidores Enzimáticos/química , Fluorescencia , Humanos , Ratas , Vasodilatación/efectos de los fármacos
16.
Biochim Biophys Acta ; 1804(3): 524-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20018259

RESUMEN

The cGMP-dependent protein kinase type I (PKG I) is an essential regulator of cellular function in blood vessels throughout the body. DT-2, a peptidic inhibitor of PKG, has played a central role in determining the molecular mechanisms of vascular control involving PKG and its signaling partners. Here, we report the development of (d)-amino acid DT-2 derivatives, namely the retro-inverso ri-(d)-DT-2 and the all (d)-amino acid analog, (d)-DT-2. Both peptide analogs were potent PKG Ialpha inhibitors with K(i) values of 5.5 nM (ri-(d)-DT-2) and 0.8 nM ((d)-DT-2) as determined using a hyperbolic mixed-type inhibition model. Also, both analogs were proteolytically stable in vivo, showed elevated selectivity, and displayed enhanced membrane translocation properties. Studies on isolated arteries from the resistance vasculature demonstrated that intraluminally perfused (d)-DT-2 significantly inhibited vasodilation induced by 8-Br-cGMP. Furthermore, in vivo application of (d)-DT-2 established a uniform translocation pattern in the resistance vasculature, with exception of the brain. Thus, (d)-DT-2 caused significant increases in mean arterial blood pressure in unrestrained, awake mice. Further, mesenteric arteries isolated from (d)-DT-2 treated animals showed a markedly reduced dilator response to 8-Br-cGMP in vitro. Our results clearly demonstrate that (d)-DT-2 is a superior inhibitor of PKG Ialpha and its application in vivo leads to sustained inhibition of PKG in vascular smooth muscle cells. The discovery of (d)-DT-2 may help our understanding of how blood vessels constrict and dilate and may also aid the development of new strategies and therapeutic agents targeted to the prevention and treatment of vascular disorders such as hypertension, stroke and coronary artery disease.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Fluoresceínas/farmacología , Fragmentos de Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Vasodilatación/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Línea Celular , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/enzimología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/química , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fluoresceínas/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertensión/enzimología , Masculino , Arterias Mesentéricas/enzimología , Ratones , Modelos Biológicos , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Fragmentos de Péptidos/uso terapéutico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Spodoptera , Vasoconstricción/efectos de los fármacos
17.
Mol Brain ; 2: 26, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19656393

RESUMEN

BACKGROUND: The Na(+)/Cl(-)-dependent serotonin (5-hydroxytryptamine, 5-HT) transporter (SERT) is a critical element in neuronal 5-HT signaling, being responsible for the efficient elimination of 5-HT after release. SERTs are not only targets for exogenous addictive and therapeutic agents but also can be modulated by endogenous, receptor-linked signaling pathways. We have shown that neuronal A3 adenosine receptor activation leads to enhanced presynaptic 5-HT transport in vitro and an increased rate of SERT-mediated 5-HT clearance in vivo. SERT stimulation by A3 adenosine receptors derives from an elevation of cGMP and subsequent activation of both cGMP-dependent protein kinase (PKG) and p38 mitogen-activated protein kinase. PKG activators such as 8-Br-cGMP are known to lead to transporter phosphorylation, though how this modification supports SERT regulation is unclear. RESULTS: In this report, we explore the kinase isoform specificity underlying the rapid stimulation of SERT activity by PKG activators. Using immortalized, rat serotonergic raphe neurons (RN46A) previously shown to support 8-Br-cGMP stimulation of SERT surface trafficking, we document expression of PKGI, and to a lower extent, PKGII. Quantitative analysis of staining profiles using permeabilized or nonpermeabilized conditions reveals that SERT colocalizes with PKGI in both intracellular and cell surface domains of RN46A cell bodies, and exhibits a more restricted, intracellular pattern of colocalization in neuritic processes. In the same cells, SERT demonstrates a lack of colocalization with PKGII in either intracellular or surface membranes. In keeping with the ability of the membrane permeant kinase inhibitor DT-2 to block 8-Br-cGMP stimulation of SERT, we found that DT-2 treatment eliminated cGMP-dependent kinase activity in PKGI-immunoreactive extracts resolved by liquid chromatography. Similarly, treatment of SERT-transfected HeLa cells with small interfering RNAs targeting endogenous PKGI eliminated 8-Br-cGMP-induced regulation of SERT activity. Co-immunoprecipitation studies show that, in transporter/kinase co-transfected cells, PKGIalpha specifically associates with hSERT. CONCLUSION: Our findings provide evidence of a physical and compartmentalized association between SERT and PKGIalpha that supports rapid, 8-Br-cGMP-induced regulation of SERT. We discuss a model wherein SERT-associated PKGIalpha supports sequentially the mobilization of intracellular transporter-containing vesicles, leading to enhanced surface expression, and the production of catalytic-modulatory SERT phosphorylation, leading to a maximal enhancement of 5-HT clearance capacity.


Asunto(s)
Antidepresivos/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Animales , Especificidad de Anticuerpos/efectos de los fármacos , Línea Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas , Transfección
18.
J Biol Chem ; 284(24): 16354-16368, 2009 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-19369251

RESUMEN

The inhibitor peptide DT-2 (YGRKKRRQRRRPPLRKKKKKH) is the most potent and selective inhibitor of the cGMP-dependent protein kinase (PKG) known today. DT-2 is a construct of a PKG tight binding sequence (W45, LRKKKKKH, KI=0.8 microM) and a membrane translocating sequence (DT-6, YGRKKRRQRRRPP, KI=1.1 microM), that combined strongly inhibits PKG catalyzed phosphorylation (KI=12.5 nM) with approximately 1000-fold selectivity toward PKG over protein kinase A, the closest relative of PKG. However, the molecular mechanism behind this inhibition is not entirely understood. Using a combination of photoaffinity labeling, stable isotope labeling, and mass spectrometry, we have located the binding sites of PKG-specific substrate and inhibitor peptides. Covalent linkage of a PKG-specific substrate analogue was localized in the catalytic core on residues 356-372, also known as the glycine-rich loop, essential for ATP binding. By analogy, the individual inhibitor peptides W45 and DT-6 were also found to cross-link near the glycine-rich loop, suggesting these are both substrate competitive inhibitors. A bifunctional photoreactive analogue of DT-2 was found to generate dimers of PKG. This cross-linking induced covalent PKG dimerization was not observed for an N-terminal deletion mutant of PKG, which lacks the dimerization domain. In addition, non-covalent mass spectrometry was used to determine binding stoichiometry and binding order of the inhibitor peptides. Dimeric PKG binds two W45 and DT-6 peptides, whereas only one DT-2 molecule was observed to bind to the dimeric PKG. Taken together, these findings imply that (i) the two individual components making up DT-2 are both targeted against the substrate-binding site and (ii) binding of a single DT-2 molecule inactivates both PKG monomers simultaneously, which is an indication that (iii) in cGMP-activated PKG the catalytic centers of both subunits may be in each other's proximity.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Fluoresceínas/farmacología , Espectrometría de Masas/métodos , Fragmentos de Péptidos/farmacología , Etiquetas de Fotoafinidad/farmacología , Animales , Sitios de Unión , Células Cultivadas , Reactivos de Enlaces Cruzados/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de GMP Cíclico/química , Dimerización , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Spodoptera
19.
Proc Natl Acad Sci U S A ; 105(1): 365-70, 2008 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-18165313

RESUMEN

Here, we report the design of unprecedented, non-FRET based cGMP-biosensors, named FlincGs, to assess the dynamics of nitric oxide (NO) and atrial natriuretic peptide (ANP) induced synthesis of intracellular cGMP, [cGMP](i). Regulatory fragments of PKG I alpha, PKG I beta, and an N-terminal deletion mutant of PKG I alpha were fused to circular permutated EGFP to generate alpha-, beta-, and delta-FlincG, with high dynamic ranges and apparent K(D,cGMP) values of 35 nM, 1.1 microM, and 170 nM, respectively. All indicators displayed significant selectivity for cGMP over cAMP, and 1.5- to 2.1-fold increases in fluorescence intensity at 510 nm when excited at 480 nm. Surprisingly, FlincGs displayed an additional excitation peak at 410 nm. delta-FlincG permitted ratiometric (480/410 nm) measurements, with a cGMP-specific 3.5-fold ratio change. In addition, delta-FlincG presented cGMP association and dissociation kinetics sufficiently fast to monitor rapid changes of [cGMP](i) in intact cells. In unpassaged, adenoviral transfected vascular smooth muscle (VSM) cells, delta-FlincG had an EC(50,cGMP) of 150 nM, and revealed transient global cGMP elevations to sustained physiological NO (EC(50,DEA/NO) = 4 nM), and the decay phase depended on the activity of PDE-5. In contrast, ANP elicited sustained submembrane elevations in [cGMP](i), which were converted to global cGMP elevations by inhibition of PDE-5 by sildenafil. These results indicate that FlincG is an innovative tool to elucidate the dynamics of a central biological signal, cGMP, and that NO and natriuretic peptides induce distinct cGMP patterning under the regulation of PDE-5, and therefore likely differentially engage cGMP targets.


Asunto(s)
Técnicas Biosensibles , GMP Cíclico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Animales , Aorta/patología , Calibración , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Cinética , Microscopía Confocal/métodos , Péptidos Natriuréticos/química , Óxido Nítrico/química , Ratas
20.
FEBS J ; 274(9): 2274-86, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17403045

RESUMEN

The molecular mechanism of cGMP-dependent protein kinase activation by its allosteric regulator cyclic-3',5'-guanosine monophosphate (cGMP) has been intensely studied. However, the structural as well as thermodynamic changes upon binding of cGMP to type I cGMP-dependent protein kinase are not fully understood. Here we report a cGMP-induced shift of Gibbs free enthalpy (DeltaDeltaGD) of 2.5 kJ.mol-1 as determined from changes in tryptophan fluorescence using urea-induced unfolding for bovine PKG Ialpha. However, this apparent increase in overall stability specifically excluded the N-terminal region of the kinase. Analyses of tryptic cleavage patterns using liquid chromatography-coupled ESI-TOF mass spectrometry and SDS/PAGE revealed that cGMP binding destabilizes the N-terminus at the hinge region, centered around residue 77, while the C-terminus was protected from degradation. Furthermore, two recombinantly expressed mutants: the deletion fragment Delta1-77 and the trypsin resistant mutant Arg77Leu (R77L) revealed that the labile nature of the N-terminus is primarily associated with the hinge region. The R77L mutation not only stabilized the N-terminus but extended a stabilizing effect on the remaining domains of the enzyme as well. These findings support the concept that the hinge region of PKG acts as a stability switch.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/química , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Termodinámica , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Bovinos , Línea Celular , GMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Estabilidad de Enzimas/genética , Fragmentos de Péptidos/genética , Unión Proteica , Estructura Terciaria de Proteína/genética , Eliminación de Secuencia , Triptófano/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...