Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Biochem Biophys Res Commun ; 464(4): 1254-1259, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212441

RESUMEN

Laminar shear flow triggers a signaling cascade that maintains the integrity of endothelial cells (ECs). Hydrogen sulfide (H2S), a new gasotransmitter is regarded as an upstream regulator of nitric oxide (NO). Whether the H2S-generating enzymes are correlated to the enzymes involved in NO production under shear flow conditions remains unclear as yet. In the present study, the cultured ECs were subjected to a constant shear flow (12 dyn/cm(2)) in a parallel flow chamber system. We investigated the expression of three key enzymes for H2S biosynthesis, cystathionine-γ-lyase (CSE), cystathionine-ß-synthase (CBS), and 3-mercapto-sulfurtransferase (3-MST). Shear flow markedly increased the level of 3-MST. Shear flow enhanced the production of H2S was determined by NBD-SCN reagent that can bind to cysteine/homocystein. Exogenous treatment of NaHS that can release gaseous H2S, ECs showed an increase of phosphorylation in Akt(S473), ERK(T202/Y204) and eNOS(S1177). This indicated that H2S can trigger the NO-production signaling cascade. Silencing of CSE, CBS and 3-MST genes by siRNA separately attenuated the phosphorylation levels of Akt(S473) and eNOS(S1177) under shear flow conditions. The particular mode of shear flow increased H2S production. The interplay between H2S and NO-generating enzymes were discussed in the present study.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Células Endoteliales/fisiología , Sulfuro de Hidrógeno/metabolismo , Mecanotransducción Celular/fisiología , Óxido Nítrico/metabolismo , Células Cultivadas , Humanos , Resistencia al Corte/fisiología , Estrés Mecánico
2.
Biochem J ; 464(1): 157-68, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25162939

RESUMEN

The endothelial cells (ECs) that line the vascular lumen are exposed to a wide variety of environmental stresses, such as hypoxia. Maladaptation to stress in ECs is a key event in the development of cardiovascular disease. Sirtuin 3 (SIRT3) is an NAD+-dependent protein deacetylase that modulates various proteins to control mitochondrial function and metabolism. We found that hypoxia elicits an increase in SIRT3 mRNA and protein expression in ECs. Under the same hypoxic conditions, the forkhead box class O transcription factor FOXO3 is deacetylated by SIRT3. The SIRT3-mediated deacetylation of FOXO3 further reduces FOXO3 phosphorylation, ubiquitination and degradation, thereby stabilizing FOXO3 proteins. As a result, the level of FOXO3 protein is increased during hypoxia. Moreover, a set of FOXO3-dependent mitochondrial antioxidant enzymes, including manganese superoxide dismutase (MnSOD), peroxiredoxin 3 (Prx3), Prx5 and thioredoxin 2 (Trx2), are up-regulated in ECs to facilitate ROS detoxification in response to hypoxia. The SIRT3-mediated deacetylation of FOXO3 preserves mitochondrial bioenergetic function and increases cell survival under hypoxic conditions. These results indicate that SIRT3 stabilizes FOXO3 via deacetylation, which enhances the mitochondrial antioxidant defence system to increase the adaptive capacity of ECs during hypoxia. This finding provides a direction for ameliorating the development of cardiovascular diseases.


Asunto(s)
Células Endoteliales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Sirtuina 3/metabolismo , Acetilación , Hipoxia de la Célula/fisiología , Supervivencia Celular/fisiología , Proteína Forkhead Box O3 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Fosforilación/fisiología , Unión Proteica/fisiología , Ubiquitinación/fisiología
3.
J Biomed Sci ; 21: 3, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24410814

RESUMEN

Hemodynamic shear stress, the blood flow-generated frictional force acting on the vascular endothelial cells, is essential for endothelial homeostasis under normal physiological conditions. Mechanosensors on endothelial cells detect shear stress and transduce it into biochemical signals to trigger vascular adaptive responses. Among the various shear-induced signaling molecules, reactive oxygen species (ROS) and nitric oxide (NO) have been implicated in vascular homeostasis and diseases. In this review, we explore the molecular, cellular, and vascular processes arising from shear-induced signaling (mechanotransduction) with emphasis on the roles of ROS and NO, and also discuss the mechanisms that may lead to excessive vascular remodeling and thus drive pathobiologic processes responsible for atherosclerosis. Current evidence suggests that NADPH oxidase is one of main cellular sources of ROS generation in endothelial cells under flow condition. Flow patterns and magnitude of shear determine the amount of ROS produced by endothelial cells, usually an irregular flow pattern (disturbed or oscillatory) producing higher levels of ROS than a regular flow pattern (steady or pulsatile). ROS production is closely linked to NO generation and elevated levels of ROS lead to low NO bioavailability, as is often observed in endothelial cells exposed to irregular flow. The low NO bioavailability is partly caused by the reaction of ROS with NO to form peroxynitrite, a key molecule which may initiate many pro-atherogenic events. This differential production of ROS and RNS (reactive nitrogen species) under various flow patterns and conditions modulates endothelial gene expression and thus results in differential vascular responses. Moreover, ROS/RNS are able to promote specific post-translational modifications in regulatory proteins (including S-glutathionylation, S-nitrosylation and tyrosine nitration), which constitute chemical signals that are relevant in cardiovascular pathophysiology. Overall, the dynamic interplay between local hemodynamic milieu and the resulting oxidative and S-nitrosative modification of regulatory proteins is important for ensuing vascular homeostasis. Based on available evidence, it is proposed that a regular flow pattern produces lower levels of ROS and higher NO bioavailability, creating an anti-atherogenic environment. On the other hand, an irregular flow pattern results in higher levels of ROS and yet lower NO bioavailability, thus triggering pro-atherogenic effects.


Asunto(s)
Hemodinámica/genética , Óxido Nítrico/metabolismo , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Humanos , Mecanotransducción Celular/genética , Procesamiento Proteico-Postraduccional/genética , Especies de Nitrógeno Reactivo/metabolismo , Transducción de Señal , Estrés Mecánico
4.
ScientificWorldJournal ; 2014: 480387, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24971375

RESUMEN

Hydrogen sulfide (H2S) and nitric oxide (NO), two endogenous gaseous molecules in endothelial cells, got increased attention with respect to their protective roles in the cardiovascular system. However, the details of the signaling pathways between H2S and NO in endothelia cells remain unclear. In this study, a treatment with NaHS profoundly increased the expression and the activity of endothelial nitric oxide synthase. Elevated gaseous NO levels were observed by a novel and specific fluorescent probe, 5-amino-2-(6-hydroxy-3-oxo-3H-xanthen-9-yl)benzoic acid methyl ester (FA-OMe), and quantified by flow cytometry. Further study indicated an increase of upstream regulator for eNOS activation, AMP-activated protein kinase (AMPK), and protein kinase B (Akt). By using a biotin switch, the level of NO-mediated protein S-nitrosylation was also enhanced. However, with the addition of the NO donor, NOC-18, the expressions of cystathionine-γ-lyase, cystathionine-ß-synthase, and 3-mercaptopyruvate sulfurtransferase were not changed. The level of H2S was also monitored by a new designed fluorescent probe, 4-nitro-7-thiocyanatobenz-2-oxa-1,3-diazole (NBD-SCN) with high specificity. Therefore, NO did not reciprocally increase the expression of H2S-generating enzymes and the H2S level. The present study provides an integrated insight of cellular responses to H2S and NO from protein expression to gaseous molecule generation, which indicates the upstream role of H2S in modulating NO production and protein S-nitrosylation.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Sulfuro de Hidrógeno/farmacología , Óxido Nítrico/biosíntesis , Proteínas Quinasas Activadas por AMP/metabolismo , Línea Celular , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Proteome Sci ; 10(1): 43, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22799578

RESUMEN

BACKGROUND: The pleiotropic effects of 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), which are independent from their cholesterol-lowering action, have been widely recognized in various biological systems. Statins can affect endothelial homeostasis, which is partly modulated by the production of nitric oxide (NO). However, it is unclear how statin/NO-mediated posttranslational S-nitrosylation of endothelial proteins and changes in translational profiles may benefit endothelial integrity. Therefore, it is important to understand the statin/NO-mediated S-nitrosylation in endothelial cells. RESULTS: Rosuvastatin treatment of human umbilical vein endothelial cells (ECs) enhanced the enzymatic activity of endothelial nitric oxide synthase (eNOS) and the expression of 78 S-nitrosoproteins. Among these S-nitrosoproteins, we identified 17 proteins, including protein disulfide bond isomerase, phospholipase C, transaldolase and heat shock proteins. Furthermore, a hydrophobic Cys66 was determined as the S-nitrosylation site of the mitochondrial HSP70. In addition to the statin-modulated posttranslational S-nitrosylation, changes in the NO-mediated translational proteome were also observed. Seventeen major proteins were significantly upregulated after rosuvastatin treatment. However, 12 of these proteins were downregulated after pretreating ECs with an eNOS inhibitor (L-NAME), which indicated that their expression was modulated by NO. CONCLUSIONS: ECs treated with rosuvastatin increase eNOS activation. The increased NO production is involved in modulating S-nitrosylation and translation of proteins. We provide further evidence of the pleiotropic effect of rosuvastatin on endothelial physiology.

6.
J Thromb Thrombolysis ; 29(1): 52-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19333555

RESUMEN

Carvedilol, a nonselective beta-adrenoceptor antagonist, has been shown to possess antioxidant effects and reduce the risk of hospitalization and death in patients with severe congestive heart failure, which is featured by the activation of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-alpha), and leads to thrombotic complications. Thrombomodulin (TM) plays protective roles against thrombosis. Treatment of ECs with TNF-alpha resulted in a down-regulation in the TM expression in a time-dependent manner. Pre-treatment of ECs with carvedilol (1 and 10 microM) for 1 h significantly up-regulated the TM expression in ECs in response to TNF-alpha. When ECs were pre-treated with a nuclear factor-kappaB (NF-kappaB) inhibitor, i.e., parthenolide, their TNF-alpha-mediated down-regulation of TM expression was inhibited. Pre-treatment of ECs with carvedilol inhibited the NF-kappaB-DNA binding activity in ECs induced by TNF-alpha. Our findings provide insights into the mechanisms by which carvedilol exerts anti-thrombotic effects by inducing TM expression in ECs in response to pro-inflammatory stimulation.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Carbazoles/farmacología , FN-kappa B/metabolismo , Propanolaminas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Trombomodulina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Carvedilol , Bovinos , Células Cultivadas , Células Endoteliales/metabolismo
7.
J Proteome Res ; 8(10): 4835-43, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19673540

RESUMEN

NO-mediated S-nitrosation of cysteine residues has been recognized as a fundamental post-translational modification. S-Nitrosation of endothelial cell (EC) proteins can alter function and affect vascular homeostasis. Trace amounts of S-nitrosoproteins in endothelial cells (ECs) in vivo coupled with lability of the S-nitroso bond have hindered a comprehensive characterization. We demonstrate a convenient and reliable method, requiring minimal sample, for the screening and identification of S-nitrosoproteins. ECs treated with the NO donor S-nitroso-N-acetylpenicillamine (SNAP) were subjected to the biotin switch method of labeling, then detected by analytical Western blot-based two-dimensional gel electrophoresis (2-DE). More than 89 SNAP-increased S-nitrosoproteins were detected and 28 of these were successfully excised from preparative 2-DE gel and identified by LC-MS/MS. Moreover, the nitrosocysteine residue for each protein (HSPA9/368, beta-actin/16, TMP3/170, vimentin/328) was also determined, and the relative ratio of S-nitrosation/non-S-nitrosation for Cys328 of vimentin was estimated using MASIC software. By the combination of the biotin switch method with 2-DE and Western blot analysis, S-nitrosoproteins can be screened and characterized by MS, providing a basis for further study of the physiological significance of each S-nitrosoproteins.


Asunto(s)
Biotina/metabolismo , Western Blotting/métodos , Electroforesis en Gel Bidimensional/métodos , Células Endoteliales/metabolismo , Proteínas/metabolismo , S-Nitrosotioles/metabolismo , Avidina/metabolismo , Línea Celular , Humanos , Espectrometría de Masas , Óxido Nítrico/metabolismo , Nitrosación , Fragmentos de Péptidos/análisis , Proteínas/química , Reproducibilidad de los Resultados , S-Nitrosotioles/química
8.
Circ Res ; 100(10): 1476-85, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17463319

RESUMEN

Angiotensin II (Ang II) is involved in the pathogenesis of atrial fibrillation (AF). L-type calcium channel (LCC) expression is altered in AF remodeling. We investigated whether Ang II modulates LCC current through transcriptional regulation, by using murine atrial HL-1 cells, which have a spontaneous calcium transient, and an in vivo rat model. Ang II increased LCC alpha1C subunit mRNA and protein levels and LCC current density, which resulted in an augmented calcium transient in atrial myocytes. An approximately 2-kb promoter region of LCC alpha1C subunit gene was cloned to the pGL3 luciferase vector. Ang II significantly increased promoter activity in a concentration- and time-dependent manner. Truncation and mutational analysis of the LCC alpha1C subunit gene promoter showed that cAMP response element (CRE) (-1853 to -1845) was an important cis element in Ang II-induced LCC alpha1C subunit gene expression. Transfection of dominant-negative CRE binding protein (CREB) (pCMV-CREBS133A) abolished the Ang II effect. Ang II (1 micromol/L, 2 hours) induced serine 133 phosphorylation of CREB and binding of CREB to CRE and increased LCC alpha1C subunit gene promoter activity through a protein kinase C/NADPH oxidase/reactive oxygen species pathway, which was blocked by the Ang II type 1 receptor blocker losartan and the antioxidant simvastatin. In the rat model, Ang II infusion increased LCC alpha1C subunit expression and serine 133 phosphorylation of CREB, which were attenuated by oral losartan and simvastatin. In summary, Ang II induced LCC alpha1C subunit expression via a protein kinase C-, reactive oxygen species-, and CREB-dependent pathway and was blocked by losartan and simvastatin.


Asunto(s)
Angiotensina II/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/fisiología , Células Cultivadas , ADN/metabolismo , Losartán/farmacología , NADPH Oxidasas/fisiología , Proteína Quinasa C/fisiología , Ratas , Ratas Wistar , Elementos de Respuesta/fisiología , Simvastatina/farmacología , Superóxidos/metabolismo , Transcripción Genética
9.
Biochem Biophys Res Commun ; 374(4): 662-7, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18664364

RESUMEN

Discoidin domain receptor-2 (DDR2) is a receptor tyrosine kinase that binds to the extracellular matrix. We investigated the role of hypoxia in DDR2 expression in vascular smooth muscle cells (VSMCs) and the underlying mechanism. Subjecting VSMCs to hypoxia (2.5% O(2)) induced DDR2 expression; treatments with a specific inhibitor (SB203580) of p38 mitogen-activated protein kinase (MAPK) or p38-specific small interference RNA (siRNA) abolished this hypoxia-induced DDR2 expression. Gel shifting assays showed that hypoxia increased the Myc-Max-DNA binding activity in the promoter region of DDR2; inhibition of p38 MAPK activation by SB203580 and p38-specific siRNA blocked hypoxia-induced DDR2 promoter activity. Hypoxia also induced matrix metalloproteinase-2 (MMP-2) activity in VSMCs and increased their migration. These VSMC responses to hypoxia were inhibited by DDR2- and p38-specific siRNAs. Our results suggested that hypoxia induces DDR2 expression in VSMCs at the transcriptional level, which is mediated by the p38 MAPK pathway and contributes to VSMC migration.


Asunto(s)
Movimiento Celular , Hipoxia/enzimología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores Mitogénicos/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , ADN/metabolismo , Receptores con Dominio Discoidina , Ensayo de Cambio de Movilidad Electroforética , Hipoxia/genética , Imidazoles/farmacología , Masculino , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piridinas/farmacología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Mitogénicos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
10.
Biochem Biophys Res Commun ; 377(4): 1274-8, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18992711

RESUMEN

Hypoxia-induced responses are frequently encountered during cardiovascular injuries. Hypoxia triggers intracellular reactive oxygen species/nitric oxide (NO) imbalance. Recent studies indicate that NO-mediated S-nitrosylation (S-NO) of cysteine residue is a key posttranslational modification of proteins. We demonstrated that acute hypoxia to endothelial cells (ECs) transiently increased the NO levels via endothelial NO synthase (eNOS) activation. A modified biotin-switch method coupled with Western blot on 2-dimensional electrophoresis (2-DE) demonstrated that at least 11 major proteins have significant increase in S-NO after acute hypoxia. Mass analysis by CapLC/Q-TOF identified those as Ras-GTPase-activating protein, protein disulfide-isomerase, human elongation factor-1-delta, tyrosine 3/tryptophan 5-monooxygenase activating protein, and several cytoskeleton proteins. The S-nitrosylated cysteine residue on tropomyosin (Cys 170) and beta-actin (Cys 285) was further verified with the trypsic peptides analyzed by MASCOT search program. Further understanding of the functional relevance of these S-nitrosylated proteins may provide a molecular basis for treating ischemia-induced vascular disorders.


Asunto(s)
Células Endoteliales/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Secuencia de Aminoácidos , Biotina/química , Biotina/metabolismo , Western Blotting , Hipoxia de la Célula , Línea Celular , Electroforesis en Gel Bidimensional , Humanos , Isquemia/metabolismo , Datos de Secuencia Molecular , Fosforilación , Proteómica , Serina/metabolismo
11.
Cardiovasc Res ; 74(3): 497-505, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17383622

RESUMEN

OBJECTIVE: Atherosclerosis is a chronic disease that involves inflammation, in which cytokines, including interferon-gamma (IFNgamma), participate. Endothelial cells (ECs) exposed to IFNgamma increase the expression of CXC chemokines. ECs subjected to laminar flow (LF) are atheroprotective, despite an unclear mechanism. This study was conducted to analyze whether ECs under LF were protected from IFNgamma-induced responses. METHODS: IFNgamma-treated human umbilical cord ECs were subjected to LF in a well-defined flow chamber system. IFNgamma-induced STAT1 activation and downstream target genes were examined. RESULTS: ECs exposed to IFNgamma triggered STAT1 activation via the phosphorylation of Tyr701 and Ser727 in STAT1. ECs exposed to LF alone did not activate STAT1. LF exposure of IFNgamma-treated ECs significantly attenuated IFNgamma-induced Tyr701 phosphorylation in a shear-force- and time-dependent manner, whereas Ser727 phosphorylation was unaffected. Consistently, LF inhibited IFNgamma-induced STAT1 binding to DNA. ECs treated with IFNgamma induced the expression of three T-cell-specific CXC chemokines (CXCL9, CXCL10 and CXCL11) as well as CIITA, a transcriptional regulator of major histocompatibility complex class II (MHCII). Consistently, LF exposure of IFNgamma-treated ECs reduced the expression of CXC chemokines and CIITA. CONCLUSIONS: LF attenuates IFNgamma-induced responses via the suppression of STAT1 activation. Inhibition by LF of the interferon-induced ECs' response may explain some aspects of LF's atheroprotective effects on the endothelium.


Asunto(s)
Aterosclerosis/inmunología , Quimiocinas CXC/metabolismo , Células Endoteliales/metabolismo , Interferón gamma/farmacología , Western Blotting , Movimiento Celular , Células Cultivadas , Quimiocina CXCL10 , Quimiocina CXCL11 , Quimiocina CXCL9 , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Proteínas Nucleares/metabolismo , Fosforilación , Interferencia de ARN , Proteínas Recombinantes , Flujo Sanguíneo Regional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/metabolismo , Estrés Mecánico , Factores de Tiempo , Transactivadores/metabolismo
12.
Chem Biol Interact ; 281: 60-68, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29273567

RESUMEN

The autophagy of human prostate cancer cells (PC3 cells) induced by a new anthraquinone derivative, 1-Hydroxy-3-[(E)-4-(piperazine-diium)but-2-enyloxy]-9,10-anthraquinone ditrifluoroactate (PA) was investigated, and the relationship between autophagy and reactive oxygen species (ROS) generation was studied. The results indicated that PA induced PC3 cell death in a time- and dose-dependent manner, could inhibit PC3 cell growth by G1 phase cell cycle arrest and corresponding decrease in the G2/M cell population and induced S-phase arrest accompanied by a significant decrease G2/M and G1 phase numbers after PC3 cells treated with PA for 48 h, and increased the accumulation of autophagolysosomes and microtubule-associated protein LC3-ll, a marker of autophagy. However, these phenomenon were not observed in the group pretreated with the autophagy inhibitor 3-MA or Bafilomycin A1 (BAF), suggesting that PA induced PC3 cell autophagy. In addition, we found that PA triggered ROS generation in cells, while the levels of ROS decreased in the N-acetylcysteine (NAC) co-treatment, indicating that PA-mediated autophagy was partly blocked by NAC. In summary, the autophagic cell death of human PC3 cells mediated by PA-triggered ROS generation.


Asunto(s)
Antraquinonas/toxicidad , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Acetilcisteína/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Antraquinonas/síntesis química , Antraquinonas/química , Caspasa 3/metabolismo , Línea Celular Tumoral , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Macrólidos/farmacología , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Thromb Haemost ; 97(4): 658-64, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17393030

RESUMEN

Thrombomodulin (TM) is an endothelial glycoprotein that functions as a thrombin cofactor in the activation of protein C. Recent evidence has revealed that TM has unique effects on cellular proliferation, adhesion, and inflammation. We examined TM expression in the arterial remodeling process with different shear conditions. Quantitative real-time reverse transcription-PCR (Q-PCR) revealed that shear stress (25 dyne/cm(2) for 6 hours) induced a 2.6 +/- 0.4-fold increase in TM mRNA levels in endothelial cell culture. Adult FVB (Friend leukemia virus B strain) mice underwent transverse aortic constriction (TAC) between the right (RCA) and left carotid artery (LCA). Doppler (n = 8), morphometric (n = 8), and Q-PCR (n = 8 or 10) studies were performed on carotid arteries at different time points. The RCA lumen and media area increased. The LCA wall shear stress decreased after TAC. RCA wall shear stress increased at day 7 followed by a decrease to the baseline at day 28. TM mRNA level in the LCA was decreased by 61% at day 7 after TAC (0.39 +/- 0.04; p < 0.05 vs. baseline). It progressively returned to the baseline at day 14 (0.85 +/- 0.12) and day 28 (1.48 +/- 0.05; all p = NS). TM appeared in the media of the RCA; TM mRNA level in the RCA was increased by 11-fold at day 14 after TAC (11.0 +/- 0.22) and progressively decreased at day 28 (5.34 +/- 0.25, all p < 0.05 vs. baseline). Our studies suggested that altered shear stress induced significant TM gene expression changes during the arterial remodeling process.


Asunto(s)
Enfermedades de la Aorta/complicaciones , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/etiología , Trombomodulina/metabolismo , Animales , Aorta Torácica/cirugía , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/fisiopatología , Velocidad del Flujo Sanguíneo , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/fisiopatología , Células Cultivadas , Constricción Patológica/complicaciones , Constricción Patológica/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Humanos , Inmunohistoquímica , Flujometría por Láser-Doppler , Ligadura , Ratones , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Mecánico , Trombomodulina/genética , Factores de Tiempo , Transcripción Genética , Regulación hacia Arriba
14.
Br J Pharmacol ; 148(2): 226-32, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16520748

RESUMEN

Magnolol (Mag), an active constituent isolated from the Chinese herb Hou p'u (Magnolia officinalis) has long been used to suppress inflammatory processes. Chronic inflammation is well known to be involved in vascular injuries such as atherosclerosis in which interleukin (IL)-6 may participate. Signal transducer and activator of transcription protein 3 (STAT3), a transcription factor involved in inflammation and the cell cycle, is activated by IL-6. In this study, we evaluated whether Mag can serve as an anti-inflammatory agent during endothelial injuries. The effects of Mag on IL-6-induced STAT3 activation and downstream target gene induction in endothelial cells (ECs) were examined. Pretreatment of ECs with Mag dose dependently inhibited IL-6-induced Tyr705 and Ser727 phosphorylation in STAT3 without affecting the phosphorylation of JAK1, JAK2, and ERK1/2. Mag pretreatment of these ECs dose dependently suppressed IL-6-induced promoter activity of intracellular cell adhesion molecule (ICAM)-1 that contains functional IL-6 response elements (IREs). An electrophoretic mobility shift assay (EMSA) revealed that Mag treatment significantly reduced STAT3 binding to the IRE region. Consistently, Mag treatment markedly inhibited ICAM-1 expression on the endothelial surface. As a result, reduced monocyte adhesion to IL-6-activated ECs was observed. Furthermore, Mag suppressed IL-6-induced promoter activity of cyclin D1 and monocyte chemotactic protein (MCP)-1 for which STAT3 activation plays a role. In conclusion, our results indicate that Mag inhibits IL-6-induced STAT3 activation and subsequently results in the suppression of downstream target gene expression in ECs. These results provide a therapeutic basis for the development of Mag as an anti-inflammatory agent for vascular disorders including atherosclerosis.


Asunto(s)
Compuestos de Bifenilo/farmacología , Células Endoteliales/efectos de los fármacos , Interleucina-6/farmacología , Lignanos/farmacología , Factor de Transcripción STAT3/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Western Blotting , Bovinos , Adhesión Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Quimiocina CCL2/genética , Ciclina D1/genética , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
15.
J Am Coll Cardiol ; 42(10): 1845-54, 2003 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-14642698

RESUMEN

OBJECTIVES: The aim of this study was to investigate the effects of angiotensin II (Ang II) on fibroblast proliferation and endothelin-1 (ET-1) gene induction, focusing especially on reactive oxygen species (ROS)-mediated signaling in cardiac fibroblasts. BACKGROUND: Angiotensin II increases ET-1 expression, which plays an important role in Ang II-induced fibroblast proliferation. Angiotensin II also stimulates ROS generation in cardiac fibroblasts. However, whether ROS are involved in Ang II-induced proliferation and ET-1 expression remains unknown. METHODS: Cultured neonatal rat cardiac fibroblasts were stimulated with Ang II, and then [(3)H]thymidine incorporation and the ET-1 gene expression were examined. We also examined the effects of antioxidants on Ang II-induced proliferation and mitogen-activated protein kinase (MAPK) phosphorylation to elucidate the redox-sensitive pathway in fibroblast proliferation and ET-1 gene expression. RESULTS: Both AT(1) receptor antagonist (losartan) and ET(A) receptor antagonist (BQ485) inhibited Ang II-increased DNA synthesis. Endothelin-1 gene was induced with Ang II as revealed by Northern blotting and promoter activity assay. Angiotensin II increased intracellular ROS levels, which were inhibited with losartan and antioxidants. Antioxidants further suppressed Ang II-induced ET-1 gene expression, DNA synthesis, and MAPK phosphorylation. PD98059, but not SB203580, fully inhibited Ang II-induced ET-1 expression. Truncation and mutational analysis of the ET-1 gene promoter showed that AP-1 binding site was an important cis-element in Ang II-induced ET-1 gene expression. CONCLUSIONS: Our data suggest that ROS are involved in Ang II-induced proliferation and ET-1 gene expression. Our findings imply that the combination of AT(I) and ET(A) receptor antagonists plus antioxidants may be beneficial in preventing the formation of excessive cardiac fibrosis.


Asunto(s)
Angiotensina II/metabolismo , Endotelina-1/genética , Fibroblastos/fisiología , Expresión Génica/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Antagonistas de Receptores de Angiotensina , Animales , Antihipertensivos/farmacología , Azepinas/farmacología , Técnicas de Cultivo de Célula , División Celular/fisiología , Antagonistas de los Receptores de Endotelina , Endotelina-1/metabolismo , Corazón/fisiología , Losartán/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Oligopéptidos/farmacología , Oxidación-Reducción , Ratas , Receptores de Angiotensina/metabolismo , Receptores de Endotelina/metabolismo , Regulación hacia Arriba
16.
FEMS Microbiol Lett ; 249(2): 247-54, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16046081

RESUMEN

Extracted polysaccharides from medicinal fungi, including Antrodia cinnamomea, Antrodia malicola, Antrodia xantha, Antrodiella liebmannii, Agaricus murrill, and Rigidoporus ulmarius, were investigated for their effects on vascular endothelial growth factor (VEGF)-induced tube formation in endothelial cells (ECs). Chemical analysis revealed that myo-inositol, sorbitol, fucose, galactosamine, glucosamine, galactose, glucose, and mannose were the neutral sugars in these polysaccharides. These fungal polysaccharides showed no toxicity to ECs. For the inhibition of endothelial tube formation, extracted polysaccharides from A. xantha and R. ulmarius were shown to produce greater inhibition compared to those from other fungi. Fucose, glucose and mannose were the predominant monosaccharides from these two fungi. These results suggest that monosaccharides may play a role in the inhibitory effect of these fungi on endothelial tube formation. In contrast to the inhibition on tube formation from polysaccharides of A. cinnamomea and A. malicola, polysaccharides from A. xantha and R. ulmarius, with molecular weight between 2693-2876 and 304-325 kDa, were critical for this inhibitory activity. Our results show that polysaccharides isolated from A. xantha and R. ulmarius provide greater antiangiogenesis than those from commercialized A. murrill (Brazilian mushroom) and A. cinnamomea. These studies provide a basis for the potential development of these polysaccharides for antiangiogenesis usage.


Asunto(s)
Inhibidores de la Angiogénesis/aislamiento & purificación , Inhibidores de la Angiogénesis/farmacología , Basidiomycota/química , Polisacáridos/aislamiento & purificación , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/toxicidad , Animales , Aorta , Bovinos , Cromatografía en Gel , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Monosacáridos/análisis , Polisacáridos/farmacología
17.
Life Sci ; 76(26): 3029-42, 2005 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-15850596

RESUMEN

The main purposes of this study were to investigate the regulation of polysaccharides isolated from A. cinnamomea on vascular endothelial growth factor (VEGF)-induced cyclin D1 expression and down stream signaling pathway that may correlate with their anti-angiogenc effects in endothelial cells (ECs). Crude and fractionated polysaccharides (Fra-1 to Fra-4) of A. cinnamomea showed slightly toxicity to ECs as compared with their inhibition concentration on angiogenic-related gene expression. The crude extract and fractionated fractions, except for Fra-2, of A. cinnamomea polysaccharides significantly decreased VEGFR2 phosphorylation on tyrosine 1054/1059, cyclin D1 promotor activity, and protein expression induced by VEGF. Crude extract of A. cinnamomea polysaccharides inhibited the binding of VEGF to KDR/flk-1 in a dose-dependent manner. These results indicated that inhibition of VEGF interaction with VEGF receptor 2 is the mechanism serves A. cinnamomea as a protective mechanism composing the anti-angiogenesis function. Furthermore, A. cinnamomea polysaccharides also blocked VEGF-induced migration and capillary-like tube formation of ECs on Matrigel. Taken together, these results indicate that A. cinnamomea polysaccharides inhibit cyclin D1 expression through inhibition of VEGF receptor signaling, leading to the suppression of angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Medicina Tradicional de Asia Oriental , Polyporales/química , Polisacáridos/farmacología , Inhibidores de la Angiogénesis/química , Animales , Aorta/citología , Aorta/efectos de los fármacos , Bovinos , Células Cultivadas , Fraccionamiento Químico , Ciclina D1/genética , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/metabolismo , Fosforilación , Extractos Vegetales/farmacología , Polisacáridos/química , Regiones Promotoras Genéticas , Tirosina , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Free Radic Biol Med ; 63: 222-34, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23665396

RESUMEN

Progressive accumulation of defective mitochondria is a common feature of aged cells. SIRT3 is a NAD(+)-dependent protein deacetylase that regulates mitochondrial function and metabolism in response to caloric restriction and stress. FOXO3 is a direct target of SIRT3 and functions as a forkhead transcription factor to govern diverse cellular responses to stress. Here we show that hydrogen peroxide induces SIRT3 to deacetylate FOXO3 at K271 and K290, followed by the upregulation of a set of genes that are essential for mitochondrial homeostasis (mitochondrial biogenesis, fission/fusion, and mitophagy). Consequently, SIRT3-mediated deacetylation of FOXO3 modulates mitochondrial mass, ATP production, and clearance of defective mitochondria. Thus, mitochondrial quantity and quality are ensured to maintain mitochondrial reserve capacity in response to oxidative damage. Maladaptation to oxidative stress is a major risk factor underlying aging and many aging-related diseases. Hence, our finding that SIRT3 deacetylates FOXO3 to protect mitochondria against oxidative stress provides a possible direction for aging-delaying therapies and disease intervention.


Asunto(s)
Acetilación , Factores de Transcripción Forkhead/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Sirtuina 3/metabolismo , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Aorta/citología , Aorta/metabolismo , Restricción Calórica , Bovinos , Proteína Forkhead Box O3 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mitocondrias/patología , Oxidación-Reducción , Mapas de Interacción de Proteínas , Especies Reactivas de Oxígeno/metabolismo
19.
J Clin Invest ; 123(3): 1057-67, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23426184

RESUMEN

Despite a general repression of translation under hypoxia, cells selectively upregulate a set of hypoxia-inducible genes. Results from deep sequencing revealed that Let-7 and miR-103/107 are hypoxia-responsive microRNAs (HRMs) that are strongly induced in vascular endothelial cells. In silico bioinformatics and in vitro validation showed that these HRMs are induced by HIF1α and target argonaute 1 (AGO1), which anchors the microRNA-induced silencing complex (miRISC). HRM targeting of AGO1 resulted in the translational desuppression of VEGF mRNA. Inhibition of HRM or overexpression of AGO1 without the 3' untranslated region decreased hypoxia-induced angiogenesis. Conversely, AGO1 knockdown increased angiogenesis under normoxia in vivo. In addition, data from tumor xenografts and human cancer specimens indicate that AGO1-mediated translational desuppression of VEGF may be associated with tumor angiogenesis and poor prognosis. These findings provide evidence for an angiogenic pathway involving HRMs that target AGO1 and suggest that this pathway may be a suitable target for anti- or proangiogenesis strategies.


Asunto(s)
Proteínas Argonautas/genética , Factores Eucarióticos de Iniciación/genética , MicroARNs/genética , Neovascularización Patológica/metabolismo , Regiones no Traducidas 3' , Animales , Proteínas Argonautas/metabolismo , Proteínas Argonautas/fisiología , Secuencia de Bases , Sitios de Unión , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Hipoxia de la Célula , Línea Celular Tumoral , Factores Eucarióticos de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/fisiología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones SCID , MicroARNs/metabolismo , Trasplante de Neoplasias , Especificidad de Órganos , Interferencia de ARN , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Thromb Res ; 128(6): 560-4, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21992896

RESUMEN

INTRODUCTION: Dysfunction of the endothelium contributes to pathological conditions of the arterial wall including atherosclerosis as a result of immunological and/or inflammatory responses. Salvianolic acid B (Sal B), a pure and active compound extracted from the Chinese herb Salvia miltiorrhizae (SM) was characterized for its anti-inflammatory and anti-oxidant properties on vascular system. METHODS AND RESULTS: Sal B pretreatment significantly inhibited the IFN-γ-induced phosphorylations of JAK2 (Tyr 1007/1008) and STAT1 (Tyr701 and Ser727). Consistently, IFN-γ-induced STAT1 downstream targets CXC chemokines' IP-10, Mig, and I-TAC were suppressed by Sal B pretreatment. Sal B inhibited promoter activities of IP-10 and the secretion of IP-10 protein. The monocyte adhesion to IFN-γ-treated ECs was observed to be reduced after Sal B pretreatment. ECs treated with Sal B alone also increased the expression of PIAS1 and SOCS1 which may also contribute to its inhibitory effect on JAK-STAT1 signaling pathways. CONCLUSIONS: The anti-inflammatory properties of Sal B on IFN-γ-induced JAK-STAT1 activation were demonstrated in the present study which provides a molecular basis for possible therapeutic usage on vascular disorders.


Asunto(s)
Benzofuranos/farmacología , Interferón gamma/farmacología , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Bovinos , Interacciones Farmacológicas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Interferón gamma/antagonistas & inhibidores , Células Jurkat , Fosforilación , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda