Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 187(6): 648-57, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23370913

RESUMEN

RATIONALE: Studies have demonstrated that angiotensin-converting enzyme 2 (ACE2) plays a protective role against lung diseases, including pulmonary hypertension (PH). Recently, an antitrypanosomal drug, diminazene aceturate (DIZE), was shown to exert an "off-target" effect of enhancing the enzymatic activity of ACE2 in vitro. OBJECTIVES: To evaluate the pharmacological actions of DIZE in experimental models of PH. METHODS: PH was induced in male Sprague Dawley rats by monocrotaline, hypoxia, or bleomycin challenge. Subsets of animals were simultaneously treated with DIZE. In a separate set of experiments, DIZE was administered after 3 weeks of PH induction to determine whether the drug could reverse PH. MEASUREMENTS AND MAIN RESULTS: DIZE treatment significantly prevented the development of PH in all of the animal models studied. The protective effects were associated with an increase in the vasoprotective axis of the lung renin-angiotensin system, decreased inflammatory cytokines, improved pulmonary vasoreactivity, and enhanced cardiac function. These beneficial effects were abolished by C-16, an ACE2 inhibitor. Initiation of DIZE treatment after the induction of PH arrested disease progression. Endothelial dysfunction represents a hallmark of PH pathophysiology, and growing evidence suggests that bone marrow-derived angiogenic progenitor cells contribute to endothelial homeostasis. We observed that angiogenic progenitor cells derived from the bone marrow of monocrotaline-challenged rats were dysfunctional and were repaired by DIZE treatment. Likewise, angiogenic progenitor cells isolated from patients with PH exhibited diminished migratory capacity toward the key chemoattractant stromal-derived factor 1α, which was corrected by in vitro DIZE treatment. CONCLUSIONS: Our results identify a therapeutic potential of DIZE in PH therapy.


Asunto(s)
Diminazeno/análogos & derivados , Hipertensión Pulmonar/prevención & control , Tripanocidas/farmacología , Animales , Ensayos de Migración Celular , Diminazeno/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio Vascular/fisiopatología , Hipertensión Pulmonar/fisiopatología , Masculino , Neovascularización Fisiológica/fisiología , Ratas , Ratas Sprague-Dawley , Sistema Renina-Angiotensina , Células Madre/fisiología
2.
J Cardiovasc Pharmacol ; 61(5): 401-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23364607

RESUMEN

Oxidative stress has been implicated in the pathogenesis of heart failure and atrial fibrillation and can result in increased peroxynitrite production in the myocardium. Atrial and ventricular canine cardiac myocytes were superfused with 3-morpholinosydnonimine-N-ethylcarbamide (SIN-1), a peroxynitrite donor, to evaluate the acute electrophysiologic effects of peroxynitrite. Perforated whole-cell patch clamp techniques were used to record action potentials. SIN-1 (200 µM) increased the action potential duration (APD) in atrial and ventricular myocytes; however, in the atria, APD prolongation was rate independent, whereas in the ventricle APD, prolongation was rate dependent. In addition to prolongation of the action potential, beat-to-beat variability of repolarization was significantly increased in ventricular but not in atrial myocytes. We examined the contribution of intracellular calcium cycling to the effects of SIN-1 by treating myocytes with the SERCA blocker, thapsigargin (5-10 µM). Inhibition of calcium cycling prevented APD prolongation in the atrial and ventricular myocytes, and prevented the SIN-1-induced increase in ventricular beat-to-beat APD variability. Collectively, these data demonstrate that peroxynitrite affects atrial and ventricular electrophysiology differentially. A detailed understanding of oxidative modulation of electrophysiology in specific chambers is critical to optimize therapeutic approaches for cardiac diseases.


Asunto(s)
Potenciales de Acción/fisiología , Función Atrial/fisiología , Molsidomina/análogos & derivados , Miocitos Cardíacos/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Función Ventricular/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Perros , Inhibidores Enzimáticos/farmacología , Femenino , Masculino , Molsidomina/farmacología , Tapsigargina/farmacología
3.
Biochemistry ; 50(36): 7774-86, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21823612

RESUMEN

Hyperkinetic Jak2 tyrosine kinase signaling has been implicated in several human diseases including leukemia, lymphoma, myeloma, and the myeloproliferative neoplasms. Using structure-based virtual screening, we previously identified a novel Jak2 inhibitor named G6. We showed that G6 specifically inhibits Jak2 kinase activity and suppresses Jak2-mediated cellular proliferation. To elucidate the molecular and biochemical mechanisms by which G6 inhibits Jak2-mediated cellular proliferation, we treated Jak2-V617F expressing human erythroleukemia (HEL) cells for 12 h with either vehicle control or 25 µM of the drug and compared protein expression profiles using two-dimensional gel electrophoresis. One differentially expressed protein identified by electrospray mass spectroscopy was the intermediate filament protein, vimentin. It was present in DMSO treated cells but absent in G6 treated cells. HEL cells treated with G6 showed both time- and dose-dependent cleavage of vimentin as well as a marked reorganization of vimentin intermediate filaments within intact cells. In a mouse model of Jak2-V617F mediated human erythroleukemia, G6 also decreased the levels of vimentin protein, in vivo. The G6-induced cleavage of vimentin was found to be Jak2-dependent and calpain-mediated. Furthermore, we found that intracellular calcium mobilization is essential and sufficient for the cleavage of vimentin. Finally, we show that the cleavage of vimentin intermediate filaments, per se, is sufficient to reduce HEL cell viability. Collectively, these results suggest that G6-induced inhibition of Jak2-mediated pathogenic cell growth is concomitant with the disruption of intracellular vimentin filaments. As such, this work describes a novel pathway for the targeting of Jak2-mediated pathological cell growth.


Asunto(s)
Janus Quinasa 2/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Estilbenos/farmacología , Vimentina/metabolismo , Animales , Calpaína/metabolismo , Muerte Celular , Línea Celular Tumoral , Humanos , Janus Quinasa 2/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Espectrometría de Masa por Ionización de Electrospray , Vimentina/química
4.
Am J Physiol Heart Circ Physiol ; 301(3): H903-11, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21724868

RESUMEN

In the vasculature, nitric oxide (NO) is generated by endothelial NO synthase (eNOS) in a calcium/calmodulin-dependent reaction. In the absence of the requisite eNOS cofactor tetrahydrobiopterin (BH(4)), NADPH oxidation is uncoupled from NO generation, leading to the production of superoxide. Although this phenomenon is apparent with purified enzyme, cellular studies suggest that formation of the BH(4) oxidation product, dihydrobiopterin, is the molecular trigger for eNOS uncoupling rather than BH(4) depletion alone. In the current study, we investigated the effects of both BH(4) depletion and oxidation on eNOS-derived superoxide production in endothelial cells in an attempt to elucidate the molecular mechanisms regulating eNOS oxidase activity. Results demonstrated that pharmacological depletion of endothelial BH(4) does not result in eNOS oxidase activity, whereas BH(4) oxidation gave rise to significant eNOS-oxidase activity. These findings suggest that the endothelium possesses regulatory mechanisms, which prevent eNOS oxidase activity from pterin-free eNOS. Using a combination of gene silencing and pharmacological approaches, we demonstrate that eNOS-caveolin-1 association is increased under conditions of reduced pterin bioavailability and that this sequestration serves to suppress eNOS uncoupling. Using small interfering RNA approaches, we demonstrate that caveolin-1 gene silencing increases eNOS oxidase activity to 85% of that observed under conditions of BH(4) oxidation. Moreover, when caveolin-1 silencing was combined with a pharmacological inhibitor of AKT, BH(4) depletion increased eNOS-derived superoxide to 165% of that observed with BH(4) oxidation. This study identifies a critical role of caveolin-1 in the regulation of eNOS uncoupling and provides new insight into the mechanisms through which disease-associated changes in caveolin-1 expression may contribute to endothelial dysfunction.


Asunto(s)
Biopterinas/análogos & derivados , Caveolina 1/metabolismo , Células Endoteliales/enzimología , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Superóxidos/metabolismo , Animales , Biopterinas/metabolismo , Bovinos , Caveolina 1/genética , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Cinética , Oxidación-Reducción , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transducción de Señal
5.
Am J Pathol ; 176(5): 2559-70, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20348244

RESUMEN

Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, is increasingly recognized as a novel biomarker in cardiovascular disease. To date, it remains unclear whether elevated ADMA levels are merely associated with cardiovascular risk or whether this molecule is of functional relevance in the pathogenesis of atherosclerotic vascular disease. To clarify this issue, we crossed dimethylarginine dimethylaminohydrolase (DDAH) transgenic mice that overexpress the human isoform 1 of the ADMA degrading enzyme DDAH into ApoE-deficient mice to generate ApoE(-/-)/hDDAH1(+/-) mice. In these mice, as well as ApoE(-/-) wild-type littermates, atherosclerosis within the aorta as well as vascular function of aortic ring preparations was assessed. We report here that overexpression of hDDAH1 reduces plaque formation in ApoE(-/-) mice by lowering ADMA. The extent of atherosclerosis closely correlated with plasma ADMA levels in male but not female mice fed either a standard rodent chow or an atherogenic diet. Functional analysis of aortic ring preparations revealed improved endothelial function in mice overexpressing hDDAH1. Our findings provide proof-of-principle that ADMA plays a causal role as a culprit molecule in atherosclerosis and support recent evidence indicating a functional relevance of DDAH enzymes in genetic mouse models. Together, these results demonstrate that pharmacological interventions targeting the ADMA/DDAH pathway may represent a novel approach in the prevention and management of cardiovascular diseases.


Asunto(s)
Amidohidrolasas/sangre , Apolipoproteínas E/metabolismo , Arginina/análogos & derivados , Aterosclerosis/enzimología , Animales , Aorta/patología , Arginina/sangre , Presión Sanguínea , Enfermedades Cardiovasculares/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos , Óxido Nítrico/metabolismo , Riesgo
6.
Circ Res ; 105(9): 897-905, 2009 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-19762684

RESUMEN

RATIONALE: Insulin-like growth factor binding protein (IGFBP)-3 modulates vascular development by regulating endothelial progenitor cell (EPC) behavior, specifically stimulating EPC cell migration. This study was undertaken to investigate the mechanism of IGFBP-3 effects on EPC function and how IGFBP-3 mediates cytoprotection following vascular injury. OBJECTIVE: To examine the mechanism of IGFBP-3-mediated repair following vascular injury. METHODS AND RESULTS: We used 2 complementary vascular injury models: laser occlusion of retinal vessels in adult green fluorescent protein (GFP) chimeric mice and oxygen-induced retinopathy in mouse pups. Intravitreal injection of IGFBP-3-expressing plasmid into lasered GFP chimeric mice stimulated homing of EPCs, whereas reversing ischemia induced increases in macrophage infiltration. IGFBP-3 also reduced the retinal ceramide/sphingomyelin ratio that was increased following laser injury. In the OIR model, IGFBP-3 prevented cell death of resident vascular endothelial cells and EPCs, while simultaneously increasing astrocytic ensheathment of vessels. For EPCs to orchestrate repair, these cells must migrate into ischemic tissue. This migratory ability is mediated, in part, by endogenous NO generation. Thus, we asked whether the migratory effects of IGFBP-3 were attributable to stimulation of NO generation. IGFBP-3 increased endothelial NO synthase expression in human EPCs leading to NO generation. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO regulated protein critical for cell migration. IGFBP-3-mediated NO generation required high-density lipoprotein receptor activation and stimulation of phosphatidylinositol 3-kinase/Akt pathway. CONCLUSION: These studies support consideration of IGFBP-3 as a novel agent to restore the function of injured vasculature and restore NO generation.


Asunto(s)
Movimiento Celular , Células Endoteliales/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Óxido Nítrico/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo , Retinopatía de la Prematuridad/metabolismo , Células Madre/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Muerte Celular , Proliferación Celular , Células Cultivadas , Ceramidas/metabolismo , Arterias Cerebrales/metabolismo , Arterias Cerebrales/fisiopatología , Citoprotección , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Recién Nacido , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Masculino , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Neovascularización Retiniana/patología , Neovascularización Retiniana/fisiopatología , Vasos Retinianos/patología , Vasos Retinianos/fisiopatología , Retinopatía de la Prematuridad/patología , Retinopatía de la Prematuridad/fisiopatología , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal , Esfingomielinas/metabolismo , Células Madre/patología , Vasodilatación
7.
J Biol Chem ; 284(51): 35338-47, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19820234

RESUMEN

Reduced NO is a hallmark of endothelial dysfunction, and among the mechanisms for impaired NO synthesis is the accumulation of the endogenous nitric-oxide synthase inhibitor asymmetric dimethylarginine (ADMA). Free ADMA is actively metabolized by the intracellular enzyme dimethylarginine dimethylaminohydrolase (DDAH), which catalyzes the conversion of ADMA to citrulline. Decreased DDAH expression/activity is evident in disease states associated with endothelial dysfunction and is believed to be the mechanism responsible for increased methylarginines and subsequent ADMA-mediated endothelial nitric-oxide synthase impairment. Two isoforms of DDAH have been identified; however, it is presently unclear which is responsible for endothelial ADMA metabolism and NO regulation. The current study investigated the effects of both DDAH-1 and DDAH-2 in the regulation of methylarginines and endothelial NO generation. Results demonstrated that overexpression of DDAH-1 and DDAH-2 increased endothelial NO by 24 and 18%, respectively. Moreover, small interfering RNA-mediated down-regulation of DDAH-1 and DDAH-2 reduced NO bioavailability by 27 and 57%, respectively. The reduction in NO production following DDAH-1 gene silencing was associated with a 48% reduction in l-Arg/ADMA and was partially restored with l-Arg supplementation. In contrast, l-Arg/ADMA was unchanged in the DDAH-2-silenced cells, and l-Arg supplementation had no effect on NO. These results clearly demonstrate that DDAH-1 and DDAH-2 manifest their effects through different mechanisms, the former of which is largely ADMA-dependent and the latter ADMA-independent. Overall, the present study demonstrates an important regulatory role for DDAH in the maintenance of endothelial function and identifies this pathway as a potential target for treating diseases associated with decreased NO bioavailability.


Asunto(s)
Amidohidrolasas/metabolismo , Arginina/análogos & derivados , Citrulina/biosíntesis , Endotelio Vascular/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Amidohidrolasas/genética , Animales , Arginina/genética , Arginina/metabolismo , Bovinos , Citrulina/genética , Expresión Génica , Isoenzimas/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética
8.
Circ Res ; 103(12): 1466-72, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-19008475

RESUMEN

Abnormal cardiac ryanodine receptor (RyR2) function is recognized as an important factor in the pathogenesis of heart failure (HF). However, the specific molecular causes underlying RyR2 defects in HF remain poorly understood. In the present study, we used a canine model of chronic HF to test the hypothesis that the HF-related alterations in RyR2 function are caused by posttranslational modification by reactive oxygen species generated in the failing heart. Experimental approaches included imaging of cytosolic ([Ca(2+)](c)) and sarcoplasmic reticulum (SR) luminal Ca(2+) ([Ca(2+)]SR) in isolated intact and permeabilized ventricular myocytes and single RyR2 channel recording using the planar lipid bilayer technique. The ratio of reduced to oxidized glutathione, as well as the level of free thiols on RyR2 decreased markedly in failing versus control hearts consistent with increased oxidative stress in HF. RyR2-mediated SR Ca(2+) leak was significantly enhanced in permeabilized myocytes, resulting in reduced [Ca(2+)](SR) in HF compared to control cells. Both SR Ca(2+) leak and [Ca(2+)](SR) were partially normalized by treating HF myocytes with reducing agents. Conversely, oxidizing agents accelerated SR Ca(2+) leak and decreased [Ca(2+)](SR) in cells from normal hearts. Moreover, exposure to antioxidants significantly improved intracellular Ca(2+)-handling parameters in intact HF myocytes. Single RyR2 channel activity was significantly higher in HF versus control because of increased sensitivity to activation by luminal Ca(2+) and was partially normalized by reducing agents through restoring luminal Ca(2+) sensitivity oxidation of control RyR2s enhanced their luminal Ca(2+) sensitivity, thus reproducing the HF phenotype. These findings suggest that redox modification contributes to abnormal function of RyR2s in HF, presenting a potential therapeutic target for treating HF.


Asunto(s)
Calcio/metabolismo , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Perros , Insuficiencia Cardíaca/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Oxidación-Reducción , Estrés Oxidativo/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Especies Reactivas de Oxígeno/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/efectos adversos , Retículo Sarcoplasmático/patología
9.
Clin Sci (Lond) ; 116(5): 423-31, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18785879

RESUMEN

The signalling pathway CD40/CD40L (CD40 ligand) plays an important role in atherosclerotic plaque formation and rupture. AngII (angiotensin II), which induces oxidative stress and inflammation, is also implicated in the progression of atherosclerosis. In the present study, we tested the hypothesis that AngII increases CD40/CD40L activity in vascular cells and that ROS (reactive oxygen species) are part of the signalling cascade that controls CD40/CD40L expression. Human CASMCs (coronary artery smooth muscle cells) in culture exposed to IL (interleukin)-1beta or TNF-alpha (tumour necrosis factor-alpha) had increased superoxide generation and enhanced CD40 expression, detected by EPR (electron paramagnetic resonance) and immunoblotting respectively. Both phenomena were abolished by previous incubation with membrane-permeant antioxidants or cell transfection with p22(phox)antisense. AngII (50-200 nmol/l) induced an early and sustained increase in CD40 mRNA and protein expression in CASMCs, which was blocked by treatment with antioxidants. Increased CD40 expression led to enhanced activity of the pathway, as AngII-treated cells stimulated with recombinant CD40L released higher amounts of IL-8 and had increased COX-2 (cyclo-oxygenase-2) expression. We conclude that AngII stimulation of vascular cells leads to a ROS-dependent increase in CD40/CD40L signalling pathway activity. This phenomenon may be an important mechanism modulating the arterial injury observed in atherosclerosis-related vasculopathy.


Asunto(s)
Angiotensina II/farmacología , Antígenos CD40/biosíntesis , Músculo Liso Vascular/efectos de los fármacos , Antioxidantes/farmacología , Antígenos CD40/genética , Ligando de CD40/metabolismo , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Citocinas/farmacología , Espectroscopía de Resonancia por Spin del Electrón/métodos , Humanos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos
10.
Pharmacol Res ; 60(6): 461-5, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19682581

RESUMEN

There is abundant evidence that the endothelium plays a crucial role in the maintenance of vascular tone and structure. One of the major endothelium-derived vasoactive mediators is nitric oxide (NO), formed in healthy vascular endothelium from the amino acid precursor l-arginine. Endothelial dysfunction is increased by various cardiovascular risk factors, metabolic diseases, and systemic or local inflammation. One mechanism that has been implicated in the development of endothelial dysfunction is the presence of elevated levels of asymmetric dimethylarginine (ADMA). Free ADMA, which is formed during proteolysis, is actively degraded by the intracellular enzyme dimethylarginine dimethylaminohydrolase (DDAH) which catalyzes the conversion of ADMA to citrulline and dimethylamine. It has been estimated that more than 70% of ADMA is metabolized by DDAH (Achan et al. [1]). Decreased DDAH expression/activity is evident in disease states associated with endothelial dysfunction and is believed to be the mechanism responsible for increased methylarginines and subsequent ADMA mediated eNOS impairment. However, recent studies suggest that DDAH may regulate eNOS activity and endothelial function through both ADMA-dependent and -independent mechanisms. In this regard, elevated plasma ADMA may serve as a marker of impaired methylarginine metabolism and the pathology previously attributed to elevated ADMA may be manifested, at least in part, through altered activity of the enzymes involved in ADMA regulation, specifically DDAH and PRMT.


Asunto(s)
Amidohidrolasas/fisiología , Arginina/análogos & derivados , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico/biosíntesis , Proteína-Arginina N-Metiltransferasas/fisiología , Animales , Arginina/fisiología , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/patología , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Óxido Nítrico/fisiología
11.
Arterioscler Thromb Vasc Biol ; 28(10): 1760-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18599801

RESUMEN

OBJECTIVE: Fine particulate matter <2.5 microm (PM(2.5)) has been implicated in vasoconstriction and potentiation of hypertension in humans. We investigated the effects of short-term exposure to PM(2.5) in the angiotensin II (AII) infusion model. METHODS AND RESULTS: Sprague-Dawley rats were exposed to PM(2.5) or filtered air (FA) for 10 weeks. At week 9, minipumps containing AII were implanted and the responses studied over a week. Mean concentration of PM(2.5) inside the chamber was 79.1+/-7.4 microg/m(3). After AII infusion, mean arterial pressure was significantly higher in PM(2.5)-AII versus FA-AII group. Aortic vasoconstriction to phenylephrine was potentiated with exaggerated relaxation to the Rho-kinase (ROCK) inhibitor Y-27632 and increase in ROCK-1 mRNA levels in the PM(2.5)-AII group. Superoxide (O(2).(-)) production in aorta was increased in the PM(2.5)-AII compared to the FA group, inhibitable by apocynin and L-NAME with coordinate upregulation of NAD(P)H oxidase subunits p22(phox) and p47(phox) and depletion of tetrahydrobiopterin. In vitro exposure to ultrafine particles (UFP) and PM(2.5) was associated with an increase in ROCK activity, phosphorylation of myosin light chain, and myosin phosphatase target subunit (MYPT1). Pretreatment with the nonspecific antioxidant N-acetylcysteine and the Rho kinase inhibitors (Fasudil and Y-27632) prevented MLC and MYPT-1 phosphorylation by UFP suggesting a O(2)(.-)-mediated mechanism for PM(2.5) and UFP effects. CONCLUSIONS: Short-term air pollution exaggerates hypertension through O(2)(.-)-mediated upregulation of the Rho/ROCK pathway.


Asunto(s)
Hipertensión/inducido químicamente , Exposición por Inhalación , Material Particulado/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Angiotensina II/administración & dosificación , Animales , Antioxidantes/farmacología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/enzimología , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Hipertensión/enzimología , Hipertensión/fisiopatología , Bombas de Infusión Implantables , Masculino , Cadenas Ligeras de Miosina/metabolismo , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa/metabolismo , Tamaño de la Partícula , Fosforilación , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética
12.
Biochemistry ; 47(27): 7256-63, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18553936

RESUMEN

The endogenous methylarginines, asymmetric dimethylarginine (ADMA) and N (G)-monomethyl- l-arginine (L-NMMA) regulate nitric oxide (NO) production from endothelial NO synthase (eNOS). Under conditions of tetrahydrobiopterin (BH 4) depletion eNOS also generates (*)O 2 (-); however, the effects of methylarginines on eNOS-derived (*)O 2 (-) generation are poorly understood. Therefore, using electron paramagnetic resonance spin trapping techniques we measured the dose-dependent effects of ADMA and L-NMMA on (*)O 2 (-) production from eNOS under conditions of BH 4 depletion. In the absence of BH 4, ADMA dose-dependently increased NOS-derived (*)O 2 (-) generation, with a maximal increase of 151% at 100 microM ADMA. L-NMMA also dose-dependently increased NOS-derived (*)O 2 (-), but to a lesser extent, demonstrating a 102% increase at 100 microM L-NMMA. Moreover, the native substrate l-arginine also increased eNOS-derived (*)O 2 (-), exhibiting a similar degree of enhancement as that observed with ADMA. Measurements of NADPH consumption from eNOS demonstrated that binding of either l-arginine or methylarginines increased the rate of NADPH oxidation. Spectrophotometric studies suggest, just as for l-arginine and L-NMMA, the binding of ADMA shifts the eNOS heme to the high-spin state, indicative of a more positive heme redox potential, enabling enhanced electron transfer from the reductase to the oxygenase site. These results demonstrate that the methylarginines can profoundly shift the balance of NO and (*)O 2 (-) generation from eNOS. These observations have important implications with regard to the therapeutic use of l-arginine and the methylarginine-NOS inhibitors in the treatment of disease.


Asunto(s)
Arginina/análogos & derivados , Óxido Nítrico Sintasa de Tipo III/metabolismo , Superóxidos/metabolismo , omega-N-Metilarginina/farmacología , Arginina/farmacología , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Hemo/metabolismo , Humanos , NADP/metabolismo
13.
Front Oncol ; 8: 97, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29682483

RESUMEN

Heat-shock factor-1 (HSF-1) is an important transcription factor that regulates pathogenesis of many human diseases through its extensive transcriptional regulation. Especially, it shows pleiotropic effects in human cancer, and hence it has recently received increased attention of cancer researchers. After myriad investigations on HSF-1, the field has advanced to the phase where there is consensus that finding a potent and selective pharmacological inhibitor for this transcription factor will be a major break-through in the treatment of various human cancers. Presently, all reported inhibitors have their limitations, made evident at different stages of clinical trials. This brief account summarizes the advances with tested natural products as HSF-1 inhibitors and highlights the necessity of phytochemistry in this endeavor of discovering a potent pharmacological HSF-1 inhibitor.

14.
Antioxid Redox Signal ; 9(4): 447-55, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17280486

RESUMEN

The current study aims to characterize the alterations of in vivo tissue redox status, oxygenation, formation of reactive oxygen species (ROS), and their effects on the postischemic heart. Mouse heart was subjected to 30 min LAD occlusion, followed by 60 min reperfusion. In vivo myocardial redox status and oxygenation were measured with electron paramagnetic resonance (EPR). In vivo tissue NAD(P)H and formation of ROS were monitored with fluorometry. Tissue glutathione/glutathione disulfide (GSH/GSSG) levels were detected with high-performance liquid chromatography (HPLC). These experiments demonstrated that tissue reduction rate of nitroxide was increased 100% during ischemia and decreased 33% after reperfusion compared to the nonischemic tissue. There was an overshoot of tissue oxygenation after reperfusion. Tissue NAD(P)H levels were increased during and after ischemia. There was a burst formation of ROS at the beginning of reperfusion. Tissue GSH/GSSG level showed a 48% increase during ischemia and 29% decrease after reperfusion. In conclusion, the hypoxia during ischemia limited mitochondrial respiration and caused a shift of tissue redox status to a more reduced state. ROS generated at the beginning of reperfusion caused a shift of redox status to a more oxidized state, which may contribute to the postischemic myocardial injury.


Asunto(s)
Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Espectroscopía de Resonancia por Spin del Electrón , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADP/metabolismo , Oxidación-Reducción
15.
Int J Parasitol ; 37(7): 743-61, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17307183

RESUMEN

Trichinella spiralis infection causes hyperexcitability in enteric after-hyperpolarising (AH) sensory neurons that is mimicked by neural, immune or inflammatory mediators known to stimulate adenylyl cyclase (AC)/cyclic 3',5'-adenosine monophosphate (cAMP) signaling. The hypothesis was tested that ongoing modulation and sustained amplification in the AC/cAMP/phosphorylated cAMP related element binding protrein (pCREB) signaling pathway contributes to hyperexcitability and neuronal plasticity in gut sensory neurons after nematode infection. Electrophysiological, immunological, molecular biological or immunochemical studies were done in T. spiralis-infected guinea-pigs (8000 larvae or saline) after acute-inflammation (7 days) or 35 days p.i., after intestinal clearance. Acute-inflammation caused AH-cell hyperexcitability and elevated mucosal and neural tissue levels of myeloperoxidase, mast cell tryptase, prostaglandin E2, leukotrine B4, lipid peroxidation, nitric oxide and gelatinase; lower level inflammation persisted 35 days p.i. Acute exposure to blockers of AC, histamine, cyclooxygenase or leukotriene pathways suppressed AH-cell hyperexcitability in a reversible manner. Basal cAMP responses or those evoked by forskolin (FSK), Ro-20-1724, histamine or substance P in isolated myenteric ganglia were augmented after T. spiralis infection; up-regulation also occurred in AC expression and AC-immunoreactivity in calbindin (AH) neurons. The cAMP-dependent slow excitatory synaptic transmission-like responses to histamine (mast cell mediator) or substance P (neurotransmitter) acting via G-protein coupled receptors (GPCR) in AH neurons were augmented by up to 2.5-fold after T. spiralis infection. FSK, histamine, substance P or T. spiralis acute infection caused a 5- to 30-fold increase in cAMP-dependent nuclear CREB phosphorylation in isolated ganglia or calbindin (AH) neurons. AC and CREB phosphorylation remained elevated 35 days p.i.. Ongoing immune activation, AC up-regulation, enhanced phosphodiesterase IV activity and facilitation of the GPCR-AC/cAMP/pCREB signaling pathway contributes to T. spiralis-induced neuronal plasticity and AH-cell hyperexcitability. This may be relevant in gut nematode infections and inflammatory bowel diseases, and is a potential therapeutic target.


Asunto(s)
AMP Cíclico/metabolismo , Mucosa Intestinal/inervación , Plasticidad Neuronal/fisiología , Neuronas Aferentes/fisiología , Trichinella spiralis/fisiología , Triquinelosis/metabolismo , Animales , Colforsina/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Didesoxiadenosina/análogos & derivados , Didesoxiadenosina/farmacología , Dinoprostona/metabolismo , Cobayas , Histamina/farmacología , Imidazoles/farmacología , Técnicas In Vitro , Mucosa Intestinal/efectos de los fármacos , Leucotrieno B4/metabolismo , Lisina/análogos & derivados , Lisina/farmacología , Masculino , Potenciales de la Membrana/fisiología , Músculo Liso/efectos de los fármacos , Músculo Liso/inervación , Plasticidad Neuronal/efectos de los fármacos , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Neuronas Aferentes/parasitología , Óxido Nítrico/metabolismo , Peroxidasa/metabolismo , Transducción de Señal , Sustancia P/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Trichinella spiralis/metabolismo , Triquinelosis/parasitología , Triptasas/metabolismo
16.
Cardiovasc Res ; 71(2): 322-30, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16626670

RESUMEN

OBJECTIVE: Research in the pathophysiology of ischemia/reperfusion or redox signaling is hindered by lack of simple methodology to measure short-lived oxygen radicals. In the presence of hydroxyl radical ((*)OH), d-phenylalanine (d-Phe) yields para-, meta- and ortho-tyrosine. We have previously demonstrated that d-Phe can accurately detect (*)OH formation in chemical, enzymatic and cellular systems by simple HPLC methodology [Anal Biochem 290:138;2001]. In the present study, we tested whether d-Phe hydroxylation can be used to detect (*)OH formation in intact organs. METHODS: Rat hearts were perfused with buffer containing 5 mM d-Phe and subjected to 30 min of total global ischemia at 37 degrees C followed by 45 min of reperfusion. Quantitative analysis of the three hydroxytyrosine isomers was achieved by HPLC-based electrochemical detection of cardiac venous effluent, with the analytical cells operating in the oxidative mode. The detection limit of this assay was <10 fmol. RESULTS: Under baseline conditions, hydroxytyrosine release from the heart was very low ( congruent with0.8 nmol/min/g). However, a prominent tyrosine burst occurred immediately upon post-ischemic reflow. In cardiac effluent collected 40 s into reperfusion, the hydroxytyrosine concentration was more than 40 times greater than at baseline; hydroxytyrosine concentration then progressively declined, to return to pre-ischemic values by 5 min of reperfusion. In parallel experiments, formation of hydroxytyrosines was markedly reduced in hearts reperfused in the presence of the (*)OH scavenger mannitol. Inclusion of 5 mm d-Phe in the perfusion medium altered neither basal cardiac function nor coronary vascular tone, but it enhanced recovery of myocardial function during post-ischemic reperfusion, consistent with direct reaction with (*)OH. CONCLUSION: Our results demonstrate that d-Phe is a sensitive method for detection of (*)OH generation in the heart. Since d-Phe is not a substrate for endogenous enzymes, it can be exploited as a reliable method to measure (*)OH formation under a variety of pathophysiological conditions.


Asunto(s)
Radical Hidroxilo/análisis , Daño por Reperfusión Miocárdica/diagnóstico , Miocardio/metabolismo , Fenilalanina/metabolismo , Isoformas de Proteínas/análisis , Animales , Biomarcadores/análisis , Cromatografía Líquida de Alta Presión/métodos , Depuradores de Radicales Libres/farmacología , Radical Hidroxilo/metabolismo , Hidroxilación , Masculino , Manitol/farmacología , Daño por Reperfusión Miocárdica/metabolismo , Oxidación-Reducción , Perfusión , Ratas , Ratas Sprague-Dawley , Tirosina/análisis , Tirosina/metabolismo
17.
Inflamm Bowel Dis ; 12(8): 766-89, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16917233

RESUMEN

Adenosine A3 receptors (ADOA3Rs) are emerging as novel purinergic targets for treatment of inflammatory diseases. Our goal was to assess the protective effect of the ADOA3R agonist N(6)-(3-iodobenzyl)-adenosine-5-N-methyluronamide (IB-MECA) on gene dysregulation and injury in a rat chronic model of 2,4,6-trinitrobenzene sulfonic acid (TNBS)--induced colitis. It was necessary to develop and validate a microarray technique for testing the protective effects of purine-based drugs in experimental inflammatory bowel disease. High-density oligonucleotide microarray analysis of gene dysregulation was assessed in colons from normal, TNBS-treated (7 days), and oral IB-MECA-treated rats (1.5 mg/kg b.i.d.) using a rat RNU34 neural GeneChip of 724 genes and SYBR green polymerase chain reaction. Analysis included clinical evaluation, weight loss assessment, and electron paramagnetic resonance imaging/spin-trap monitoring of free radicals. Remarkable colitis-induced gene dysregulation occurs in the most exceptional cluster of 5.4% of the gene pool, revealing 2 modes of colitis-related dysregulation. Downregulation occurs in membrane transporter, mitogen-activated protein (MAP) kinase, and channel genes. Upregulation occurs in chemokine, cytokine/inflammatory, stress, growth factor, intracellular signaling, receptor, heat shock protein, retinoid metabolism, neural, remodeling, and redox-sensitive genes. Oral IB-MECA prevented dysregulation in 92% of these genes, histopathology, gut injury, and weight loss. IB-MECA or adenosine suppressed elevated free radicals in ex vivo inflamed gut. Oral IB-MECA blocked the colitis-induced upregulation (90% of genes tested (33 of 37 genes). We conclude that our validated high-density oligonucleotide microarray analysis is a powerful technique for molecular gene dysregulation studies to assess the beneficial effects of purine-based or other drugs in experimental colitis. ADOA3R is new potential therapeutic target for inflammatory bowel disease.


Asunto(s)
Agonistas del Receptor de Adenosina A3 , Adenosina/análogos & derivados , Colitis/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Adenosina/farmacología , Adenosina/uso terapéutico , Animales , Colitis/inducido químicamente , Colitis/enzimología , Colitis/genética , Modelos Animales de Enfermedad , Radicales Libres/metabolismo , Glutatión Peroxidasa/metabolismo , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo , Ácido Trinitrobencenosulfónico
18.
Biochem Pharmacol ; 72(6): 681-92, 2006 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-16844095

RESUMEN

Apigenin, a flavone abundantly found in fruits and vegetables, exhibits antiproliferative, anti-inflammatory, and antimetastatic activities through poorly defined mechanisms. In the present study, the treatment of different cell lines with apigenin resulted in selective antiproliferative and apoptotic effect in monocytic and lymphocytic leukemias. Apigenin-induced-apoptosis was mediated by the activation of caspase-9 and caspase-3. Apigenin was found intracellularly and localized to the mitochondria. Treatment of monocytic cells with apigenin was accompanied by an increase in reactive oxygen species (ROS) and phosphorylation of the MAPKs, p38 and ERK. However, the inhibition of ROS, p38 or ERK failed to block apoptosis, suggesting that these cellular responses induced by apigenin are not essential for the induction of apoptosis. In addition, apigenin induced the activation of PKCdelta. Pharmacological inhibition of PKCdelta, the expression of dominant-negative PKCdelta and silencing of PKCdelta in leukemia cells showed that apigenin-induced-apoptosis requires PKCdelta activity. Together, these results indicate that this flavonoid provides selective activity to promote caspase-dependent-apoptosis of leukemia cells and uncover an essential role of PKCdelta during the induction of apoptosis by apigenin.


Asunto(s)
Apigenina/farmacología , Apoptosis/efectos de los fármacos , Caspasas/fisiología , Proteína Quinasa C-delta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células 3T3 , Animales , Apoptosis/fisiología , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Células HL-60 , Humanos , Células Jurkat , Células K562 , Leucemia/patología , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Tumorales Cultivadas , Células U937
19.
Cancer Res ; 62(1): 307-12, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11782393

RESUMEN

Therapeutic regimens such as radiation or chemotherapy attempt to exploit the physiological differences between normal and malignant tissue. Tissue redox status and pO(2) are two factors that are hypothesized to be different in normal and malignant tissues. Methods that can detect subtle differences in the above physiological parameters would greatly aid in devising appropriate treatment strategies. We have previously used in vivo electron paramagnetic resonance (EPR) spectroscopy and imaging techniques and shown that tumor tissues are highly reducing and hypoxic compared with normal tissues (P. Kuppusamy et al., Cancer Res., 58: 1562-1568, 1998). The purpose of the present study was to obtain spatially resolved redox data from normal and tumor tissues of radiation-induced fibrosarcoma (RIF-1) tumor-bearing mice and to examine the role of intracellular glutathione (GSH) on the tissue redox status. Experiments were performed using low-frequency (1.3 GHz) in vivo EPR spectroscopy and imaging techniques with a nitroxide redox probe. L-buthionine-S,R-sulfoximine (BSO), an inhibitor of GSH synthesis, was used to deplete tissue GSH levels. The results show the existence of significant heterogeneity of redox status in the tumor tissue compared with normal tissue. The tumor tissues show at least 4-fold higher concentrations of GSH levels compared with normal tissues in the tumor-bearing mice. Also BSO treatment showed a differential depletion of GSH and reducing equivalents in the tumor tissue. Thus, it appears that there is significant heterogeneity of tumor redox status and that manipulation of the tumor redox status may be important in tumor growth and therapy.


Asunto(s)
Fibrosarcoma/metabolismo , Glutatión/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Butionina Sulfoximina/farmacología , Espectroscopía de Resonancia por Spin del Electrón/métodos , Femenino , Ratones , Ratones Endogámicos C3H , Óxidos de Nitrógeno/farmacocinética , Oxidación-Reducción
20.
Biochem Biophys Rep ; 5: 96-104, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26709389

RESUMEN

In humans, sulfite is generated endogenously by the metabolism of sulfur containing amino acids such as methionine and cysteine. Sulfite is also formed from exposure to sulfur dioxide, one of the major environmental pollutants. Sulfite is used as an antioxidant and preservative in dried fruits, vegetables, and beverages such as wine. Sulfite is also used as a stabilizer in many drugs. Sulfite toxicity has been associated with allergic reactions characterized by sulfite sensitivity, asthma, and anaphylactic shock. Sulfite is also toxic to neurons and cardiovascular cells. Recent studies suggest that the cytotoxicity of sulfite is mediated by free radicals; however, molecular mechanisms involved in sulfite toxicity are not fully understood. Cytochrome c (cyt c) is known to participate in mitochondrial respiration and has antioxidant and peroxidase activities. Studies were performed to understand the related mechanism of oxidation of sulfite and radical generation by ferric cytochrome c (Fe3+ cyt c) in the absence and presence of H2O2. Electron paramagnetic resonance (EPR) spin trapping studies using 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) were performed with sulfite, Fe3+ cyt c, and H2O2. An EPR spectrum corresponding to the sulfite radical adducts of DMPO (DMPO-SO3-) was obtained. The amount of DMPO-SO3- formed from the oxidation of sulfite by the Fe3+ cyt c increased with sulfite concentration. In addition, the amount of DMPO-SO3- formed by the peroxidase activity of Fe3+ cyt c also increased with sulfite and H2O2 concentration. From these results, we propose a mechanism in which the Fe3+ cyt c and its peroxidase activity oxidizes sulfite to sulfite radical. Our results suggest that Fe3+ cyt c could have a novel role in the deleterious effects of sulfite in biological systems due to increased production of sulfite radical. It also shows that the increased production of sulfite radical may be responsible for neurotoxicity and some of the injuries which occur to humans born with molybdenum cofactor and sulfite oxidase deficiencies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA