Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circulation ; 148(13): 1023-1034, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37548012

RESUMEN

BACKGROUND: The major cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) has emerged as a key mediator of inflammation that underlies cardiovascular disease. On interaction with double-stranded DNA, cGAS generates the second messenger 2',3'-cyclic GMP-AMP (cGAMP) that directly binds to and activates the stimulator of interferon genes, which in turn leads to enhanced expression of genes encoding interferons and proinflammatory cytokines. Here, we show that cGAMP generated by cGAS also directly activates PKGI (cGMP-dependent protein kinase 1), a mechanism that underlies crosstalk between inflammation and blood pressure regulation. METHODS: The ability of cGAS and cGAMP to activate PKGI was assessed using molecular, cellular, and biochemical analyses, and in myography experiments, as well. The release of cGAMP from the endothelium was measured using an ELISA, and its uptake into the vascular smooth muscle was assessed using molecular and biochemical approaches, including the identification and targeting of specific cGAMP transporters. The blood pressure of wild-type and cGAS-/- mice was assessed using implanted telemetry probes. cGAS was activated by in vivo transfection with G3-YSD or mice were made septic by administration of lipopolysaccharide. RESULTS: The detection of cytosolic DNA by cGAS within the vascular endothelium leads to formation of cGAMP that was found to be actively extruded by MRP1 (multidrug resistance protein 1). Once exported, this cGAMP is then imported into neighboring vascular smooth muscle cells through the volume-regulated anion channel, where it can directly activate PKGI. The activation of PKGI by cGAMP mediates vasorelaxation that is dependent on the activity of MRP1 and volume-regulated anion channel, but independent of the canonical nitric oxide pathway. This mechanism of PKGI activation mediates lowering of blood pressure and contributes to hypotension and tissue hypoperfusion during sepsis. CONCLUSIONS: The activation of PKGI by cGAMP enables the coupling of blood pressure to cytosolic DNA sensing by cGAS, which plays a key role during sepsis by mediating hypotension and tissue hypoperfusion.


Asunto(s)
ADN , Hipotensión , Animales , Ratones , Presión Sanguínea , ADN/metabolismo , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Inflamación
2.
Nat Commun ; 14(1): 2123, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37055412

RESUMEN

Redox signaling and cardiac function are tightly linked. However, it is largely unknown which protein targets are affected by hydrogen peroxide (H2O2) in cardiomyocytes that underly impaired inotropic effects during oxidative stress. Here, we combine a chemogenetic mouse model (HyPer-DAO mice) and a redox-proteomics approach to identify redox sensitive proteins. Using the HyPer-DAO mice, we demonstrate that increased endogenous production of H2O2 in cardiomyocytes leads to a reversible impairment of cardiac contractility in vivo. Notably, we identify the γ-subunit of the TCA cycle enzyme isocitrate dehydrogenase (IDH)3 as a redox switch, linking its modification to altered mitochondrial metabolism. Using microsecond molecular dynamics simulations and experiments using cysteine-gene-edited cells reveal that IDH3γ Cys148 and 284 are critically involved in the H2O2-dependent regulation of IDH3 activity. Our findings provide an unexpected mechanism by which mitochondrial metabolism can be modulated through redox signaling processes.


Asunto(s)
Peróxido de Hidrógeno , Mitocondrias , Ratones , Animales , Peróxido de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Metabolismo Energético , Miocitos Cardíacos/metabolismo , Estrés Oxidativo
4.
Circulation ; 140(2): 126-137, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31116951

RESUMEN

BACKGROUND: The health-promoting and disease-limiting abilities of resveratrol, a natural polyphenol, has led to considerable interest in understanding the mechanisms of its therapeutic actions. The polyphenolic rings of resveratrol enable it to react with and detoxify otherwise injurious oxidants. Whilst the protective actions of resveratrol are commonly ascribed to its antioxidant activity, here we show that this is a misconception. METHODS: The ability of resveratrol to oxidize cGMP-dependent PKG1α (protein kinase 1α) was assessed in isolated rat aortic smooth muscle cells, and the mechanism of action of this polyphenol was characterized using in vitro experiments, mass spectrometry and electron paramagnetic resonance. The blood pressure of wild-type and C42S knock-in mice was assessed using implanted telemetry probes. Mice were made hypertensive by administration of angiotensin II via osmotic mini-pumps and blood pressure monitored during 15 days of feeding with chow diet containing vehicle or resveratrol. RESULTS: Oxidation of the phenolic rings of resveratrol paradoxically leads to oxidative modification of proteins, explained by formation of a reactive quinone that oxidizes the thiolate side chain of cysteine residues; events that were enhanced in cells under oxidative stress. Consistent with these observations and its ability to induce vasodilation, resveratrol induced oxidative activation of PKG1α and lowered blood pressure in hypertensive wild-type mice, but not C42S PKG1α knock-in mice that are resistant to disulfide activation. CONCLUSIONS: Resveratrol mediates lowering of blood pressure by paradoxically inducing protein oxidation, especially during times of oxidative stress, a mechanism that may be a common feature of antioxidant molecules.


Asunto(s)
Antioxidantes/farmacología , Presión Sanguínea/efectos de los fármacos , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Estrés Oxidativo/efectos de los fármacos , Resveratrol/farmacología , Animales , Presión Sanguínea/fisiología , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/fisiología , Ratas , Telemetría/métodos
5.
Mol Cell ; 69(3): 438-450.e5, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29358077

RESUMEN

S-nitrosation, commonly referred to as S-nitrosylation, is widely regarded as a ubiquitous, stable post-translational modification that directly regulates many proteins. Such a widespread role would appear to be incompatible with the inherent lability of the S-nitroso bond, especially its propensity to rapidly react with thiols to generate disulfide bonds. As anticipated, we observed robust and widespread protein S-nitrosation after exposing cells to nitrosocysteine or lipopolysaccharide. Proteins detected using the ascorbate-dependent biotin switch method are typically interpreted to be directly regulated by S-nitrosation. However, these S-nitrosated proteins are shown to predominantly comprise transient intermediates leading to disulfide bond formation. These disulfides are likely to be the dominant end effectors resulting from elevations in nitrosating cellular nitric oxide species. We propose that S-nitrosation primarily serves as a transient intermediate leading to disulfide formation. Overall, we conclude that the current widely held perception that stable S-nitrosation directly regulates the function of many proteins is significantly incorrect.


Asunto(s)
Disulfuros/metabolismo , Nitrosación/fisiología , Procesamiento Proteico-Postraduccional/fisiología , S-Nitrosotioles/metabolismo , Cisteína/metabolismo , Humanos , Óxido Nítrico/metabolismo , Oxidación-Reducción , Proteínas/metabolismo , Proteolisis , Proteómica/métodos , Compuestos de Sulfhidrilo/metabolismo
6.
J Mol Cell Cardiol ; 115: 20-31, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29294329

RESUMEN

BACKGROUND: Type 2A protein phosphatase (PP2A) enzymes are serine/threonine phosphatases which comprise a scaffold A subunit, a regulatory B subunit and a catalytic C subunit, and have been implicated in the dephosphorylation of multiple cardiac phosphoproteins. B subunits determine subcellular targeting, substrate specificity and catalytic activity, and can themselves be regulated by post-translational modifications. We explored potential ß-adrenergic regulation of PP2A in cardiomyocytes through phosphorylation of the regulatory B subunit isoform B56δ. METHODS AND RESULTS: Phosphate affinity SDS-PAGE and immunoblot analysis revealed increased phosphorylation of B56δ in adult rat ventricular myocytes (ARVM) exposed to the ß-adrenergic receptor (ßAR) agonist isoprenaline (ISO). Phosphorylation of B56δ occurred at S573, primarily through stimulation of the ß1AR subtype, and was dependent on PKA activity. The functional role of the phosphorylation was explored in ARVM transduced with adenoviruses expressing wild type (WT) or non-phosphorylatable (S573A) B56δ, fused to GFP at the N-terminus. C subunit expression was increased in ARVM expressing GFP-B56δ-WT or GFP-B56δ-S573A, both of which co-immunoprecipitated with endogenous C and A subunits. PP2A activity in cell lysates was increased in response to ISO in ARVM expressing GFP-B56δ-WT but not GFP-B56δ-S573A. Immunoblot analysis of the phosphoproteome in ARVM expressing GFP-B56δ-WT or GFP-B56δ-S573A with antibodies detecting (i) phospho-serine/threonine residues in distinct kinase substrate motifs or (ii) specific phosphorylated residues of functional importance in selected proteins revealed a comparable phosphorylation profile in the absence or presence of ISO stimulation. CONCLUSIONS: In cardiomyocytes, ßAR stimulation induces PKA-mediated phosphorylation of the PP2A regulatory subunit isoform B56δ at S573, which increases associated PP2A catalytic activity. This is likely to regulate the phosphorylation status of specific B56δ-PP2A substrates, which remain to be identified.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Miocardio/enzimología , Fosfoserina/metabolismo , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína/metabolismo , Adenoviridae/metabolismo , Secuencia de Aminoácidos , Animales , Cardiomegalia/enzimología , Cardiomegalia/patología , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Isoproterenol/farmacología , Masculino , Ratones , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/química , Subunidades de Proteína/química , Ratas Wistar
7.
Hypertension ; 70(3): 577-586, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28716990

RESUMEN

Arterial hypertension continues to be a major health burden. Development of new antihypertensive drugs that engage vasodilatory mechanisms not harnessed by available therapies offer therapeutic potential. Oxidants induce an interprotein disulfide in PKG Iα (protein kinase G Iα) at C42, which is associated with its targeting and activation, resulting in vasodilation and blood pressure lowering. Consequently, we developed an assay and screened for electrophilic drugs that activate PKG Iα by selectively targeting C42, as such compounds have potential as novel antihypertensives with a mechanism of action that differs from current therapies. In this way, a drug that we termed G1 was identified, which targets C42 of PKG Iα to induce vasodilation of isolated resistance blood vessels and blood pressure lowering in a mouse model of angiotensin II-induced hypertension. In contrast, these antihypertensive effects were deficient in angiotensin II-induced hypertensive C42S PKG Iα knockin mice. These transgenic mice were engineered to have the reactive cysteinyl thiol replaced with a hydroxyl so that it cannot react with endogenous vasodilatory oxidants or electrophiles such as drug G1. These studies, therefore, provide validation of PKG Iα C42 as the target of G1, as well as proof-of-principle for a new class of antihypertensive drugs that have potential for further development for clinical use in humans.


Asunto(s)
Antihipertensivos/farmacología , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Hipertensión , Músculo Liso Vascular , Vasodilatación/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Hipertensión/fisiopatología , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Oxidación-Reducción/efectos de los fármacos , Resultado del Tratamiento , Vasodilatación/fisiología
8.
J Biol Chem ; 292(39): 16161-16173, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28739872

RESUMEN

The kinase p38α MAPK (p38α) plays a pivotal role in many biological processes. p38α is activated by canonical upstream kinases that phosphorylate the activation region. The purpose of our study was to determine whether such activation may depend on redox-sensing cysteines within p38α. p38α was activated and formed a disulfide-bound heterodimer with MAP2K3 (MKK3) in rat cardiomyocytes and isolated hearts exposed to H2O2 This disulfide heterodimer was sensitive to reduction by mercaptoethanol and was enhanced by the thioredoxin-reductase inhibitor auranofin. We predicted that Cys-119 or Cys-162 of p38α, close to the known MKK3 docking domain, were relevant for these redox characteristics. The C119S mutation decreased whereas the C162S mutation increased the dimer formation, suggesting that these two Cys residues act as vicinal thiols, consistent with C119S/C162S being incapable of sensing H2O2 Similarly, disulfide heterodimer formation was abolished in H9C2 cells expressing both MKK3 and p38α C119S/C162S and subjected to simulated ischemia and reperfusion. However, the p38α C119S/C162S mutants did not exhibit appreciable alteration in activating dual phosphorylation. In contrast, the anti-inflammatory agent 10-nitro-oleic acid (NO2-OA), a component of the Mediterranean diet, reduced p38α activation and covalently modified Cys-119/Cys-162, probably obstructing MKK3 access. Moreover, NO2-OA reduced the dephosphorylation of p38α by hematopoietic tyrosine phosphatase (HePTP). Furthermore, steric obstruction of Cys-119/Cys-162 by NO2-OA pretreatment in Langendorff-perfused murine hearts prevented the p38-MKK3 disulfide dimer formation and attenuated H2O2-induced contractile dysfunction. Our findings suggest that cysteine residues within p38α act as redox sensors that can dynamically regulate the association between p38 and MKK3.


Asunto(s)
Cistina/metabolismo , Ventrículos Cardíacos/enzimología , MAP Quinasa Quinasa 3/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Moleculares , Miocitos Cardíacos/enzimología , Estrés Oxidativo , Sustitución de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Cisteína/química , Cisteína/metabolismo , Cistina/química , Activación Enzimática , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Técnicas In Vitro , MAP Quinasa Quinasa 3/química , MAP Quinasa Quinasa 3/genética , Masculino , Ratones Endogámicos C57BL , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Conformación Proteica , Multimerización de Proteína , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo
9.
Biochemistry ; 55(5): 751-61, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26784639

RESUMEN

8-Nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is a nitrated derivative of guanosine 3',5'-cyclic monophosphate (cGMP) formed endogenously under conditions associated with production of both reactive oxygen species and nitric oxide. It acts as an electrophilic second messenger in the regulation of cellular signaling by inducing a post-translational modification of redox-sensitive protein thiols via covalent adduction of cGMP moieties to protein thiols (protein S-guanylation). Here, we demonstrate that 8-nitro-cGMP potentially S-guanylates thiol groups of cGMP-dependent protein kinase (PKG), the enzyme that serves as one of the major receptor proteins for intracellular cGMP and controls a variety of cellular responses. S-Guanylation of PKG was found to occur in a site specific manner; Cys42 and Cys195 were the susceptible residues among 11 Cys residues. Importantly, S-guanylation at Cys195, which is located in the high-affinity cGMP binding domain of PKG, causes persistent enzyme activation as determined by in vitro kinase assay as well as by an organ bath assay. In vivo, S-guanylation of PKG was demonstrated to occur in mice without any specific treatment and was significantly enhanced by lipopolysaccharide administration. These findings warrant further investigation in terms of the physiological and pathophysiological roles of S-guanylation-dependent persistent PKG activation.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Guanina/metabolismo , Nucleótidos Cíclicos/metabolismo , Proteínas/metabolismo , Animales , Activación Enzimática , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/enzimología , Miocardio/metabolismo
10.
J Biol Chem ; 291(4): 1774-1788, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26620565

RESUMEN

The gasotransmitter, hydrogen sulfide (H2S) is recognized as an important mediator of endothelial cell homeostasis and function that impacts upon vascular tone and blood pressure. Cystathionine-γ-lyase (CSE) is the predominant endothelial generator of H2S, and recent evidence suggests that its transcriptional expression is regulated by the reactive oxygen species, H2O2. However, the cellular source of H2O2 and the redox-dependent molecular signaling pathway that modulates this is not known. We aimed to investigate the role of Nox4, an endothelial generator of H2O2, in the regulation of CSE in endothelial cells. Both gain- and loss-of-function experiments in human endothelial cells in vitro demonstrated Nox4 to be a positive regulator of CSE transcription and protein expression. We demonstrate that this is dependent upon a heme-regulated inhibitor kinase/eIF2α/activating transcription factor 4 (ATF4) signaling module. ATF4 was further demonstrated to bind directly to cis-regulatory sequences within the first intron of CSE to activate transcription. Furthermore, CSE expression was also increased in cardiac microvascular endothelial cells, isolated from endothelial-specific Nox4 transgenic mice, compared with wild-type littermate controls. Using wire myography we demonstrate that endothelial-specific Nox4 transgenic mice exhibit a hypo-contractile phenotype in response to phenylephrine that was abolished when vessels were incubated with a CSE inhibitor, propargylglycine. We, therefore, conclude that Nox4 is a positive transcriptional regulator of CSE in endothelial cells and propose that it may in turn contribute to the regulation of vascular tone via the modulation of H2S production.


Asunto(s)
Cistationina gamma-Liasa/genética , Regulación Enzimológica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/enzimología , NADPH Oxidasas/metabolismo , Transcripción Genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Cistationina gamma-Liasa/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Transducción de Señal
11.
Front Pharmacol ; 6: 166, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26321952

RESUMEN

Homeostatic cardiac function is maintained by a complex network of interdependent signaling pathways which become compromised during disease progression. Excitation-contraction-coupling, the translation of an electrical signal to a contractile response is critically dependent on a tightly controlled sequence of events culminating in a rise in intracellular Ca(2+) and subsequent contraction of the myocardium. Dysregulation of this Ca(2+) handling system as well as increases in the production of reactive oxygen species (ROS) are two major contributing factors to myocardial disease progression. ROS, generated by cellular oxidases and by-products of cellular metabolism, are highly reactive oxygen derivatives that function as key secondary messengers within the heart and contribute to normal homeostatic function. However, excessive production of ROS, as in disease, can directly interact with kinases critical for Ca(2+) regulation. This post-translational oxidative modification therefore links changes in the redox status of the myocardium to phospho-regulated pathways essential for its function. This review aims to describe the oxidative regulation of the Ca(2+)/calmodulin-dependent kinase II (CaMKII) and cAMP-dependent protein kinase A (PKA), and the subsequent impact this has on Ca(2+) handling within the myocardium. Elucidating the impact of alterations in intracellular ROS production on Ca(2+) dynamics through oxidative modification of key ROS sensing kinases, may provide novel therapeutic targets for preventing myocardial disease progression.

12.
Nat Commun ; 6: 7920, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26258640

RESUMEN

Angiogenesis is essential for tissue development, wound healing and tissue perfusion, with its dysregulation linked to tumorigenesis, rheumatoid arthritis and heart disease. Here we show that pro-angiogenic stimuli couple to NADPH oxidase-dependent generation of oxidants that catalyse an activating intermolecular-disulphide between regulatory-RIα subunits of protein kinase A (PKA), which stimulates PKA-dependent ERK signalling. This is crucial to blood vessel growth as 'redox-dead' Cys17Ser RIα knock-in mice fully resistant to PKA disulphide-activation have deficient angiogenesis in models of hind limb ischaemia and tumour-implant growth. Disulphide-activation of PKA represents a new therapeutic target in diseases with aberrant angiogenesis.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Neovascularización Fisiológica/genética , Animales , Aorta/fisiología , Bovinos , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Células Endoteliales , Técnicas de Sustitución del Gen , Miembro Posterior , Inmunoprecipitación , Isquemia , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/irrigación sanguínea , Oxidación-Reducción , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/farmacología
13.
Hypertension ; 64(6): 1344-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25267801

RESUMEN

Dysregulated blood pressure control leading to hypertension is prevalent and is a risk factor for several common diseases. Fully understanding blood pressure regulation offers the possibility of developing rationale therapies to alleviate hypertension and associated disease risks. Although hydrogen sulfide (H2S) is a well-established endogenous vasodilator, the molecular basis of its blood-pressure lowering action is incompletely understood. H2S-dependent vasodilation and blood pressure lowering in vivo was mediated by it catalyzing formation of an activating interprotein disulfide within protein kinase G (PKG) Iα. However, this oxidative activation of PKG Iα is counterintuitive because H2S is a thiol-reducing molecule that breaks disulfides, and so it is not generally anticipated to induce their formation. This apparent paradox was explained by H2S in the presence of molecular oxygen or hydrogen peroxide rapidly converting to polysulfides, which have oxidant properties that in turn activate PKG by inducing the disulfide. These observations are relevant in vivo because transgenic knockin mice in which the cysteine 42 redox sensor within PKG has been systemically replaced with a redox-dead serine residue are resistant to H2S-induced blood pressure lowering. Thus, a primary mechanism by which the reductant molecule H2S lowers blood pressure is mediated somewhat paradoxically by the oxidative activation of PKG.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Sulfuro de Hidrógeno/farmacología , Hipertensión/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Hipertensión/metabolismo , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sustancias Reductoras
14.
Methods Enzymol ; 528: 111-28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23849862

RESUMEN

Hydrogen peroxide regulates intracellular signaling by oxidatively converting susceptible cysteine thiols to a modified state, which includes the formation of intermolecular disulfides. This type of oxidative modification can occur within the cAMP- and cGMP-dependent protein kinases often referred to as PKA and PKG, which have important roles in regulating cardiac contractility and systemic blood pressure. Both kinases are stimulated through conical pathways that elevate their respective cyclic nucleotides leading to direct kinase stimulation. However, PKA and PKG can also be functionally modulated independently of cyclic nucleotide stimulation through direct cysteine thiol oxidation leading to intermolecular disulfide formation. In the case of PKG, the formation of an intermolecular disulfide between two parallel dimeric subunits leads to enhanced kinase affinity for substrate. For PKA, the formation of two intermolecular disulfides between antiparallel dimeric regulatory RI subunits increases the affinity of this kinase for its binding partners, the A-kinase anchoring proteins, leading to increased PKA localization to its substrates. In this chapter, we describe the methods for detecting intermolecular disulfide-bound proteins and monitoring PKA and PKG oxidation within biological samples.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Disulfuros/análisis , Células Endoteliales/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Procesamiento Proteico-Postraduccional , Células Cultivadas , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/química , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/química , Cisteína/metabolismo , Electroforesis en Gel de Poliacrilamida , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Inmunohistoquímica , Oxidación-Reducción , Fosforilación , Unión Proteica , Transducción de Señal/efectos de los fármacos
15.
Proc Natl Acad Sci U S A ; 110(24): 9909-13, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23716652

RESUMEN

Sepsis is a common life-threatening clinical syndrome involving complications as a result of severe infection. A cardinal feature of sepsis is inflammation that results in oxidative stress. Sepsis in wild-type mice induced oxidative activation of cGMP-dependent protein kinase 1 alpha (PKG Iα), which increased blood vessel dilation and permeability, and also lowered cardiac output. These responses are typical features of sepsis and their combined effect is a lowering of blood pressure. This hypotension, a hallmark of sepsis, resulted in underperfusion of end organs, resulting in their damage. A central role for PKG Iα oxidative activation in injury is supported by oxidation-resistant Cys42Ser PKG Iα knock-in mice being markedly protected from these clinical indices of injury during sepsis. We conclude that oxidative activation of PKG Iα is a key mediator of hypotension and consequential organ injury during sepsis.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Hipotensión/fisiopatología , Insuficiencia Multiorgánica/fisiopatología , Sepsis/fisiopatología , Sustitución de Aminoácidos , Animales , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Activación Enzimática/genética , Hipotensión/enzimología , Hipotensión/genética , Immunoblotting , L-Lactato Deshidrogenasa/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Insuficiencia Multiorgánica/enzimología , Insuficiencia Multiorgánica/genética , Oxidación-Reducción , Sepsis/enzimología , Sepsis/genética
16.
J Biol Chem ; 288(21): 15380-9, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23548900

RESUMEN

We demonstrate for the first time that endomembrane-delimited H-Ras mediates VEGF-induced activation of endothelial nitric-oxide synthase (eNOS) and migratory response of human endothelial cells. Using thiol labeling strategies and immunofluorescent cell staining, we found that only 31% of total H-Ras is S-palmitoylated, tethering the small GTPase to the plasma membrane but leaving the function of the large majority of endomembrane-localized H-Ras unexplained. Knockdown of H-Ras blocked VEGF-induced PI3K-dependent Akt (Ser-473) and eNOS (Ser-1177) phosphorylation and nitric oxide-dependent cell migration, demonstrating the essential role of H-Ras. Activation of endogenous H-Ras led to recruitment and phosphorylation of eNOS at endomembranes. The loss of migratory response in cells lacking endogenous H-Ras was fully restored by modest overexpression of an endomembrane-delimited H-Ras palmitoylation mutant. These studies define a newly recognized role for endomembrane-localized H-Ras in mediating nitric oxide-dependent proangiogenic signaling.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Cultivadas , Células Endoteliales/citología , Inducción Enzimática/fisiología , Humanos , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Factor A de Crecimiento Endotelial Vascular/genética
18.
Antioxid Redox Signal ; 18(9): 1042-52, 2013 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22867279

RESUMEN

SIGNIFICANCE: Oxidants were once principally considered perpetrators of injury and disease. However, this has become an antiquated view, with cumulative evidence showing that the oxidant hydrogen peroxide serves as a signaling molecule. Hydrogen peroxide carries vital information about the redox state of the cell and is crucial for homeostatic regulation during health and adaptation to stress. RECENT ADVANCES: In this review, we examine the contemporary concepts for how hydrogen peroxide is sensed and transduced into a biological response by introducing post-translational oxidative modifications on select proteins. Oxidant sensing and signaling by kinases are of particular importance as they integrate oxidant signals into phospho-regulated pathways. We focus on CAMKII, PKA, and PKG, kinases whose redox regulation has notable impact on cardiovascular function. CRITICAL ISSUES: In addition, we examine the mechanism for regulating intracellular hydrogen peroxide, considering the net concentrations that may accumulate. The effects of endogenously generated oxidants are often modeled by applying exogenous hydrogen peroxide to cells or tissues. Here we consider whether model systems exposed to exogenous hydrogen peroxide have relevance to systems where the oxidant is generated endogenously, and if so, what concentration can be justified in terms of relevance to health and disease. FUTURE DIRECTIONS: Improving our understanding of hydrogen peroxide signaling and the sensor proteins that it can modify will help us develop new strategies to regulate intracellular signaling to prevent disease.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Sistema Cardiovascular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Peróxido de Hidrógeno/metabolismo , Transducción de Señal/fisiología , Animales , Cisteína/metabolismo , Homeostasis , Humanos , Peróxido de Hidrógeno/farmacología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Metionina/metabolismo , Modelos Cardiovasculares , Oxidación-Reducción , Oxidorreductasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Sistemas de Mensajero Secundario/fisiología , Compuestos de Sulfhidrilo/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Superóxidos/metabolismo
19.
Circ Res ; 111(8): 1091-106, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23023511

RESUMEN

Redox signaling refers to the specific and usually reversible oxidation/reduction modification of molecules involved in cellular signaling pathways. In the heart, redox signaling regulates several physiological processes (eg, excitation-contraction coupling) and is involved in a wide variety of pathophysiological and homoeostatic or stress response pathways. Reactive oxygen species involved in cardiac redox signaling may derive from many sources, but NADPH oxidases, as dedicated sources of signaling reactive oxygen species, seem to be especially important. An increasing number of specific posttranslational oxidative modifications involved in cardiac redox signaling are being defined, along with the reactive oxygen species sources that are involved. Here, we review current knowledge on the molecular targets of signaling reactive oxygen species in cardiac cells and their involvement in cardiac physiopathology. Advances in this field may allow the development of targeted therapeutic strategies for conditions such as heart failure as opposed to the general antioxidant approaches that have failed to date.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/patología , Contracción Miocárdica/fisiología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Animales , Cardiomegalia/fisiopatología , Humanos , NADPH Oxidasas/metabolismo , Oxidación-Reducción
20.
Circulation ; 126(3): 287-95, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22685118

RESUMEN

BACKGROUND: Although nitroglycerin has remained in clinical use since 1879, the mechanism by which it relaxes blood vessels to lower blood pressure remains incompletely understood. Nitroglycerin undergoes metabolism that generates several reaction products, including oxidants, and this bioactivation process is essential for vasodilation. Protein kinase G (PKG) mediates classic nitric oxide-dependent vasorelaxation, but the 1α isoform is also independently activated by oxidation that involves interprotein disulfide formation within this homodimeric protein complex. We hypothesized that nitroglycerin-induced vasodilation is mediated by disulfide activation of PKG1α. METHODS AND RESULTS: Treating smooth muscle cells or isolated blood vessels with nitroglycerin caused PKG1α disulfide dimerization. PKG1α disulfide formation was increased in wild-type mouse aortas by in vivo nitroglycerin treatment, but this oxidation was lost as tolerance developed. To establish whether kinase oxidation underlies nitroglycerin-induced vasodilation in vivo, we used a Cys42Ser PKG1α knock-in mouse that cannot transduce oxidant signals because it does not contain the vital redox-sensing thiol. This redox-dead knock-in mouse was substantively deficient in hypotensive response to nitroglycerin compared with wild-type littermates as measured in vivo by radiotelemetry. Resistance blood vessels from knock-ins were markedly less sensitive to nitroglycerin-induced vasodilation (EC(50)=39.2 ± 10.7 µmol/L) than wild-types (EC(50)=12.1 ± 2.9 µmol/L). Furthermore, after ≈24 hours of treatment, wild-type controls stopped vasodilating to nitroglycerin, and the vascular sensitivity to nitroglycerin was decreased, whereas this tolerance phenomenon, which routinely hampers the management of hypertensive patients, was absent in knock-ins. CONCLUSIONS: PKG1α disulfide formation is a significant mediator of nitroglycerin-induced vasodilation, and tolerance to nitroglycerin is associated with loss of kinase oxidation.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Hipertensión , Nitroglicerina/farmacología , Sustitución de Aminoácidos/genética , Animales , Aorta/citología , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/química , Dimerización , Disulfuros/química , Disulfuros/metabolismo , Técnicas de Sustitución del Gen , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Hipertensión/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Liso Vascular/citología , Oxidación-Reducción/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Compuestos de Sulfhidrilo/metabolismo , Telemetría , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...