Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 122(12): 1689-1702, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29545367

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PH) is a life-threatening condition associated with immune dysregulation and abnormal regulatory T cell (Treg) activity, but it is currently unknown whether and how abnormal Treg function differentially affects males and females. OBJECTIVE: To evaluate whether and how Treg deficiency differentially affects male and female rats in experimental PH. METHODS AND RESULTS: Male and female athymic rnu/rnu rats, lacking Tregs, were treated with the VEGFR2 (vascular endothelial growth factor receptor 2) inhibitor SU5416 or chronic hypoxia and evaluated for PH; some animals underwent Treg immune reconstitution before SU5416 administration. Plasma PGI2 (prostacyclin) levels were measured. Lung and right ventricles were assessed for the expression of the vasoprotective proteins COX-2 (cyclooxygenase 2), PTGIS (prostacyclin synthase), PDL-1 (programmed death ligand 1), and HO-1 (heme oxygenase 1). Inhibitors of these pathways were administered to athymic rats undergoing Treg immune reconstitution. Finally, human cardiac microvascular endothelial cells cocultured with Tregs were evaluated for COX-2, PDL-1, HO-1, and ER (estrogen receptor) expression, and culture supernatants were assayed for PGI2 and IL (interleukin)-10. SU5416-treatment and chronic hypoxia produced more severe PH in female than male athymic rats. Females were distinguished by greater pulmonary inflammation, augmented right ventricular fibrosis, lower plasma PGI2 levels, decreased lung COX-2, PTGIS, HO-1, and PDL-1 expression and reduced right ventricular PDL-1 levels. In both sexes, Treg immune reconstitution protected against PH development and raised levels of plasma PGI2 and cardiopulmonary COX-2, PTGIS, PDL-1, and HO-1. Inhibiting COX-2, HO-1, and PD-1 (programmed death 1)/PDL-1 pathways abrogated Treg protection. In vitro, human Tregs directly upregulated endothelial COX-2, PDL-1, HO-1, ERs and increased supernatant levels of PGI2 and IL-10. CONCLUSIONS: In 2 animal models of PH based on Treg deficiency, females developed more severe PH than males. The data suggest that females are especially reliant on the normal Treg function to counteract the effects of pulmonary vascular injury leading to PH.


Asunto(s)
Hipertensión Pulmonar/prevención & control , Factores Sexuales , Linfocitos T Reguladores/fisiología , Inhibidores de la Angiogénesis/farmacología , Animales , Antígeno B7-H1/análisis , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Enfermedad Crónica , Ciclooxigenasa 2/análisis , Ciclooxigenasa 2/metabolismo , Sistema Enzimático del Citocromo P-450/análisis , Sistema Enzimático del Citocromo P-450/metabolismo , Epoprostenol/antagonistas & inhibidores , Epoprostenol/sangre , Epoprostenol/metabolismo , Femenino , Hemo Oxigenasa (Desciclizante)/análisis , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Indoles/farmacología , Oxidorreductasas Intramoleculares/análisis , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/metabolismo , Pulmón/metabolismo , Masculino , Prostaglandinas I/biosíntesis , Pirroles/farmacología , Ratas , Ratas Desnudas , Receptores de Estrógenos/análisis , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/metabolismo , Linfocitos T Reguladores/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
2.
Br J Clin Pharmacol ; 83(1): 152-162, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27151091

RESUMEN

Bioactive peptides derived from milk proteins are food components that, in addition to their nutritional value, retain many biological properties and have therapeutic effects in several health disorders, including cardiovascular disease. Amongst these, atherosclerosis is the underlying cause of heart attack and strokes. It is a progressive dyslipidaemic and inflammatory disease where accumulation of oxidized lipids and inflammatory cells leads to the formation of an atherosclerotic plaque in the vessel wall. Milk-derived bioactive peptides can be released during gastrointestinal digestion, food processing or by enzymatic and bacterial fermentation and are considered to promote diverse beneficial effects such as lipid lowering, antihypertensive, immnomodulating, anti-inflammatory and antithrombotic effects. In this review, an overview of the diverse biological effects of these compounds is given, particularly focusing on their beneficial properties on cardiovascular disease and proposing novel mechanisms of action responsible for their bioactivity. Attempts to prevent cardiovascular diseases target modifications of several risk factors such as high blood pressure, obesity, high blood concentrations of lipids or insulin resistance. Milk-derived bioactive peptides are a source of health-enhancing components and the potential health benefit of these compounds has a growing commercial potential. Consequently, they have been incorporated as ingredients in functional foods, as dietary supplements and as pharmaceuticals to promote health and reduce risk of chronic diseases.


Asunto(s)
Aterosclerosis/prevención & control , Proteínas de la Leche/química , Péptidos/administración & dosificación , Péptidos/farmacología , Suplementos Dietéticos , Fermentación , Humanos , Péptidos/aislamiento & purificación
3.
Blood ; 121(1): 188-96, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23160460

RESUMEN

Wnt signaling is involved in numerous aspects of vertebrate development and homeostasis, including the formation and function of blood cells. Here, we show that canonical and noncanonical Wnt signaling pathways are present and functional in megakaryocytes (MKs), with several Wnt effectors displaying MK-restricted expression. Using the CHRF288-11 cell line as a model for human MKs, the canonical Wnt3a signal was found to induce a time and dose-dependent increase in ß-catenin expression. ß-catenin accumulation was inhibited by the canonical antagonist dickkopf-1 (DKK1) and by the noncanonical agonist Wnt5a. Whole genome expression analysis demonstrated that Wnt3a and Wnt5a regulated distinct patterns of gene expression in MKs, and revealed a further interplay between canonical and noncanonical Wnt pathways. Fetal liver cells derived from low-density-lipoprotein receptor-related protein 6-deficient mice (LRP6(-/-)), generated dramatically reduced numbers of MKs in culture of lower ploidy (2N and 4N) than wild-type controls, implicating LRP6-dependent Wnt signaling in MK proliferation and maturation. Finally, in wild-type mature murine fetal liver-derived MKs, Wnt3a potently induced proplatelet formation, an effect that could be completely abrogated by DKK1. These data identify novel extrinsic regulators of proplatelet formation, and reveal a profound role for Wnt signaling in platelet production.


Asunto(s)
Megacariocitos/citología , Trombopoyesis/fisiología , Vía de Señalización Wnt/fisiología , Animales , Plaquetas/citología , Línea Celular , Células Cultivadas/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Hígado/embriología , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Proteínas Recombinantes/farmacología , Trombopoyesis/genética , Proteínas Wnt/farmacología , Proteína Wnt3A/farmacología , beta Catenina/biosíntesis , beta Catenina/genética
4.
Circ Res ; 112(1): 174-94, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23287454

RESUMEN

The development of drugs that inhibit platelets has been driven by a combination of clinical insights, fundamental science, and sheer luck. The process has evolved as the days of stumbling on therapeutic gems, such as aspirin, have long passed and have been replaced by an arduous process in which a drug is designed to target a specific protein implicated in a well-characterized pathophysiological process, or so we would like to believe. The development of antiplatelet therapy illustrates the importance of understanding the mechanisms of disease and the pharmacology of the compounds we develop, coupled with careful clinical experimentation and observation and, yes, still, a fair bit of luck.


Asunto(s)
Plaquetas/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Inhibidores de la Ciclooxigenasa/uso terapéutico , Descubrimiento de Drogas , Inhibidores de Agregación Plaquetaria/uso terapéutico , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Investigación Biomédica Traslacional , Animales , Aspirina/uso terapéutico , Plaquetas/enzimología , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/historia , Clopidogrel , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/efectos adversos , Inhibidores de la Ciclooxigenasa/historia , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/historia , Resistencia a Medicamentos , Medicina Basada en la Evidencia , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Farmacogenética , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/historia , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/efectos adversos , Antagonistas del Receptor Purinérgico P2Y/historia , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico , Investigación Biomédica Traslacional/historia
5.
FASEB J ; 27(2): 499-510, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23070607

RESUMEN

Conjugated linoleic acid (CLA) induces regression of preestablished atherosclerosis in the ApoE(-/-) mouse. Understanding the mechanisms involved may help in identifying novel pathways associated with the regression of human disease. Animals were administered a 1% cholesterol diet for 12 wk, with 1% CLA supplementation from wk 8 to 12. ApoE(-/-) mice fed only the 1% cholesterol diet for 12 wk were employed as controls. Transcriptomic analysis of mouse aorta showed that many of the components of the IL-10 signaling pathway were modified during CLA-induced regression. Real-time PCR and Western blot analysis showed increased IL-10 receptor expression, phosphorylation of STAT3, and downstream target gene expression in the aorta, alongside an increase in serum IL-10 (79.8 ± 22.4 vs. 41.9 ± 5.5 pg/ml, n = 10; P < 0.01). CLA -supplementation also increased IL-10 production in bone marrow-derived macrophages (143.6 ± 28.6 vs. 94 ± 5.6 pg/ml, n = 5; P < 0.05). To explore the mechanisms for altered IL-10 production, we examined the profile of monocyte/macrophage phenotype in the vessel wall, bone marrow, and spleen. CLA increased macrophage polarization toward an anti-inflammatory M2 phenotype in vivo, increasing the population of Ly6C(lo) monocytes (29 vs. 77 ± 14, n=5, P < 0.05) in the aorta. CLA had similar effects on monocytes/macrophages differentiated from marrow-derived progenitor cells and on splenocytes. The induction of IL-10 on CLA supplementation in this model may reflect a systemic alteration toward an anti-inflammatory phenotype, which, in turn promotes increased vascular infiltration by Ly6C(lo) monocytes. These cells may contribute to CLA-induced disease regression.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Interleucina-10/inmunología , Ácidos Linoleicos Conjugados/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Interleucina-10/sangre , Interleucina-10/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
6.
Proteomics ; 13(14): 2135-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23606334

RESUMEN

15-Deoxy-delta12, 14-prostaglandin J(2) (15d-PGJ(2)) is an endogenous anti-inflammatory lipid derived from PGD(2). One potential mechanism for its activity is the covalent modification of cellular proteins, via a reactive α,ß-unsaturated carbonyl group in its cyclopentenone ring, which in turn alters protein function. In order to identify the candidate target proteins covalently modified by 15d-PGJ(2) in human aortic endothelial cell (EC), EC was treated with biotinylated-15d-PGJ(2), the modified proteins extracted by Neutravidin affinity-purification and the proteins identified by LTQ Orbitrap mass spectrometer. Classification of the 358 identified proteins was performed using PANTHER classification system (www.pantherdb.org), showing that the proteins mapped to metabolic process, cellular process, and transport activity. This protein data set highlights the potential for 15d-PGJ(2) to covalently modify cellular proteins and provides a source of data that will aid further studies on the mechanism of action of this endogenous regulator of inflammation.


Asunto(s)
Prostaglandina D2/análogos & derivados , Proteínas/análisis , Proteínas/clasificación , Proteoma/análisis , Proteómica/métodos , Línea Celular , Cromatografía de Fase Inversa , Células Endoteliales/química , Células Endoteliales/metabolismo , Humanos , Prostaglandina D2/química , Prostaglandina D2/metabolismo , Proteínas/química , Proteínas/metabolismo , Proteoma/química , Proteoma/metabolismo , Espectrometría de Masas en Tándem
7.
Circulation ; 123(17): 1854-63, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21502571

RESUMEN

BACKGROUND: Thrombin is a key mediator of platelet activation. Atopaxar is a reversible protease-activated receptor-1 antagonist that interferes with thrombin-mediated platelet effects. The phase II Lessons From Antagonizing the Cellular Effect of Thrombin-Coronary Artery Disease (LANCELOT-CAD) trial examined the safety and tolerability of prolonged therapy with atopaxar in subjects with CAD. METHODS AND RESULTS: Subjects with a qualifying history were randomized in a double-blind fashion to 3 dosing regimens of atopaxar (50, 100, or 200 mg daily) or matching placebo for 24 weeks and followed up for an additional 4 weeks. The key safety end points were bleeding according to the Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) and Thrombolysis in Myocardial Infarction (TIMI) classifications. Secondary objectives included platelet aggregation and major adverse cardiac events. Seven hundred and twenty subjects were randomized. Overall bleeding rates tended to be higher with atopaxar compared with placebo by CURE criteria (placebo, 0.6%; atopaxar, 3.9%; relative risk, 6.82, P=0.03; 50 mg, 3.9%; 100 mg, 1.7%; 200 mg, 5.9%; P for trend=0.01) and TIMI criteria (placebo, 6.8%; atopaxar, 10.3%; relative risk, 1.52, P=0.17; 50 mg, 9.9%; 100 mg, 8.1%; 200 mg, 12.9%; P for trend=0.07). There was no difference in major bleeding. Major adverse cardiac events were numerically lower in the atopaxar subjects. All atopaxar regimens achieved high levels of platelet inhibition. A transient elevation in liver transaminases and dose-dependent QTc prolongation without apparent complications were observed in higher-dose atopaxar treatment groups. CONCLUSIONS: In this dose-ranging study of patients with CAD, treatment with atopaxar resulted in platelet inhibition, more minor bleeding, and numerically but not statistically fewer ischemic events. Larger-scale trials are needed to determine whether these patterns translate into clinically meaningful effects. CLINICAL TRIAL REGISTRATION: URL: http://www.ClinicalTrials.gov. Unique identifier: NCT00312052.


Asunto(s)
Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Iminas/administración & dosificación , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Piridinas/administración & dosificación , Receptores de Trombina/antagonistas & inhibidores , Anciano , Biomarcadores , Plaquetas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Hemorragia/inducido químicamente , Humanos , Iminas/efectos adversos , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/efectos adversos , Piridinas/efectos adversos
8.
Circulation ; 123(17): 1843-53, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21502577

RESUMEN

BACKGROUND: Atopaxar (E5555) is a reversible protease-activated receptor-1 thrombin receptor antagonist that interferes with platelet signaling. The primary objective of the Lessons From Antagonizing the Cellular Effects of Thrombin-Acute Coronary Syndromes (LANCELOT­ACS) trial was to evaluate the safety and tolerability of atopaxar in patients with ACS. METHODS AND RESULTS: Six hundred and three subjects were randomized within 72 hours of non-ST-elevation ACS to 1 of 3 doses of atopaxar (400-mg loading dose followed by 50, 100, or 200 mg daily) or matching placebo. The incidence of Clopidogrel in Unstable Angina to Prevent Recurrent Events (CURE) major or minor bleeding did not differ significantly between the combined atopaxar and placebo groups (3.08% versus 2.17%, respectively; P=0.63), and there was no dose-related trend (P=0.80). The incidence of CURE major bleeding was numerically higher in the atopaxar group compared with the placebo group (1.8% versus 0%; P=0.12). The incidence of cardiovascular death, myocardial infarction, stroke, or recurrent ischemia was similar between the atopaxar and placebo arms (8.03% versus 7.75%; P=0.93). The incidence of CV death, MI, or stroke was 5.63% in the placebo group and 3.25% in the combined atopaxar group (P=0.20). Dose-dependent trends for efficacy were not seen. Atopaxar significantly reduced ischemia on continuous ECG monitoring (Holter) at 48 hours compared with placebo (relative risk, 0.67; P=0.02). Transient dose-dependent transaminase elevation and relative QTc prolongation were observed with the highest doses of atopaxar. CONCLUSION: In patients after ACS, atopaxar significantly reduced early ischemia on Holter monitoring without a significant increase in major or minor bleeding. Larger trials are required to fully establish the efficacy and safety of atopaxar. CLINICAL TRIAL REGISTRATION: URL: http://www.ClinicalTrials.gov. Unique identifier: NCT00548587.


Asunto(s)
Síndrome Coronario Agudo/tratamiento farmacológico , Iminas/administración & dosificación , Inhibidores de Agregación Plaquetaria/administración & dosificación , Piridinas/administración & dosificación , Receptores de Trombina/antagonistas & inhibidores , Síndrome Coronario Agudo/mortalidad , Anciano , Muerte Súbita Cardíaca/epidemiología , Femenino , Hemorragia/epidemiología , Humanos , Iminas/efectos adversos , Incidencia , Hepatopatías/epidemiología , Masculino , Persona de Mediana Edad , Infarto del Miocardio/epidemiología , Inhibidores de Agregación Plaquetaria/efectos adversos , Piridinas/efectos adversos , Factores de Riesgo , Prevención Secundaria , Accidente Cerebrovascular/epidemiología
9.
Blood ; 116(22): 4646-56, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20833976

RESUMEN

Within the healthy population, there is substantial, heritable, and interindividual variability in the platelet response. We explored whether a proportion of this variability could be accounted for by interindividual variation in gene expression. Through a correlative analysis of genome-wide platelet RNA expression data from 37 subjects representing the normal range of platelet responsiveness within a cohort of 500 subjects, we identified 63 genes in which transcript levels correlated with variation in the platelet response to adenosine diphosphate and/or the collagen-mimetic peptide, cross-linked collagen-related peptide. Many of these encode proteins with no reported function in platelets. An association study of 6 of the 63 genes in 4235 cases and 6379 controls showed a putative association with myocardial infarction for COMMD7 (COMM domain-containing protein 7) and a major deviation from the null hypo thesis for LRRFIP1 [leucine-rich repeat (in FLII) interacting protein 1]. Morpholino-based silencing in Danio rerio identified a modest role for commd7 and a significant effect for lrrfip1 as positive regulators of thrombus formation. Proteomic analysis of human platelet LRRFIP1-interacting proteins indicated that LRRFIP1 functions as a component of the platelet cytoskeleton, where it interacts with the actin-remodeling proteins Flightless-1 and Drebrin. Taken together, these data reveal novel proteins regulating the platelet response.


Asunto(s)
Plaquetas/metabolismo , Perfilación de la Expresión Génica , Proteínas de Unión al ARN/metabolismo , Animales , Silenciador del Gen , Genotipo , Humanos , Activación Plaquetaria , Proteoma/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Trombosis , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(47): 19836-41, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19901330

RESUMEN

Wnts regulate important intracellular signaling events, and dysregulation of the Wnt pathway has been linked to human disease. Here, we uncover numerous Wnt canonical effectors in human platelets where Wnts, their receptors, and downstream signaling components have not been previously described. We demonstrate that the Wnt3a ligand inhibits platelet adhesion, activation, dense granule secretion, and aggregation. Wnt3a also altered platelet shape change and inhibited the activation of the small GTPase RhoA. In addition, we found the Wnt-beta-catenin signaling pathway to be functional in platelets. Finally, disruption of the Wnt Frizzled 6 receptor in the mouse resulted in a hyperactivatory platelet phenotype and a reduced sensitivity to Wnt3a. Taken together our studies reveal a novel functional role for Wnt signaling in regulating anucleate platelet function and may provide a tractable target for future antiplatelet therapy.


Asunto(s)
Plaquetas/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , Animales , Plaquetas/citología , Calcio/metabolismo , Activación Enzimática , Receptores Frizzled/metabolismo , Humanos , Ratones , Adhesividad Plaquetaria/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Vesículas Secretoras/metabolismo , Proteína Wnt3 , Proteína Wnt3A , beta Catenina/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
11.
J Biol Chem ; 285(29): 22202-10, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20457605

RESUMEN

The signaling molecule 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) has been described as the "anti-inflammatory prostaglandin." Here we show that substrates of the nuclear export receptor CRM1 accumulate in the nucleus in the presence of 15d-PGJ(2), identifying this prostaglandin as a regulator of CRM1-dependent nuclear protein export that can be produced endogenously. Like leptomycin B (LMB), an established fungal CRM1-inhibitor, 15d-PGJ(2) reacts with a conserved cysteine residue in the CRM1 sequence. This covalent modification prevents the formation of nuclear export complexes. Cells that are transfected with mutant CRM1 (C528S) are resistant to the inhibitory effects of LMB and 15d-PGJ(2), demonstrating that the same single amino acid is targeted by the two compounds. Inhibition of the CRM1 pathway by endogenously produced prostaglandin and/or exogenously applied 15d-PGJ(2) may contribute to its anti-inflammatory, anti-proliferative, and anti-viral effects.


Asunto(s)
Antiinflamatorios/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Carioferinas/metabolismo , Prostaglandina D2/análogos & derivados , Receptores Citoplasmáticos y Nucleares/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Secuencia de Aminoácidos , Antiinflamatorios/química , Supervivencia Celular/efectos de los fármacos , Cisteína/metabolismo , Ácidos Grasos Insaturados/farmacología , Células HeLa , Humanos , Carioferinas/química , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Péptidos/química , Péptidos/metabolismo , Prostaglandina D2/química , Prostaglandina D2/farmacología , Receptores Citoplasmáticos y Nucleares/química , Proteína Exportina 1
12.
BMC Cancer ; 9: 207, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19558693

RESUMEN

BACKGROUND: Prostaglandin E2 (PGE2) is the major product of Cyclooxygenase-2 (COX-2) in colorectal cancer (CRC). We aimed to assess PGE2 cell surface receptors (EP 1-4) to examine the mechanisms by which PGE2 regulates tumour progression. METHODS: Gene expression studies were performed by quantitative RT-PCR. Cell cycle was analysed by flow cytometry with cell proliferation quantified by BrdU incorporation measured by enzyme immunoassay. Immunohistochemistry was employed for expression studies on formalin fixed paraffin embedded tumour tissue. RESULTS: EP4 was the most abundant subtype of PGE2 receptor in HT-29 and HCA7 cells (which show COX-2 dependent PGE2 generation) and was consistently the most abundant transcript in human colorectal tumours (n = 8) by qRT-PCR (ANOVA, p = 0.01). G0/G1 cell cycle arrest was observed in HT-29 cells treated with SC-236 5 microM (selective COX-2 inhibitor) for 24 hours (p = 0.02), an effect abrogated by co-incubation with PGE2 (1 microM). G0/G1 arrest was also seen with a specific EP4 receptor antagonist (EP4A, L-161982) (p = 0.01). Treatment of HT-29 cells with either SC-236 or EP4A caused reduction in intracellular cAMP (ANOVA, p = 0.01). Early induction in p21WAF1/CIP1 expression (by qRT-PCR) was seen with EP4A treatment (mean fold increase 4.4, p = 0.04) while other genes remained unchanged. Similar induction in p21WAF1/CIP1 was also seen with PD153025 (1 microM), an EGFR tyrosine kinase inhibitor, suggesting EGFR transactivation by EP4 as a potential mechanism. Additive inhibition of HCA7 proliferation was observed with the combination of SC-236 and neutralising antibody to amphiregulin (AR), a soluble EGFR ligand. Concordance in COX-2 and AR localisation in human colorectal tumours was noted. CONCLUSION: COX-2 regulates cell cycle transition via EP4 receptor and altered p21WAF1/CIP1 expression. EGFR pathways appear important. Specific targeting of the EP4 receptor or downstream targets may offer a safer alternative to COX-2 inhibition in the chemoprevention of CRC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Dinoprostona/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptores de Prostaglandina E/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Citometría de Flujo/métodos , Humanos , Inmunohistoquímica/métodos , Lesiones Precancerosas , Subtipo EP4 de Receptores de Prostaglandina E , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Pharmacol Exp Ther ; 326(1): 51-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18375790

RESUMEN

Exogenous prostacyclin is effective in reducing pulmonary vascular resistance in some forms of human pulmonary hypertension (PH). To explore whether endogenous prostaglandins played a similar role in pulmonary hypertension, we examined the effect of deleting cyclooxygenase (COX)-gene isoforms in a chronic hypoxia model of PH. Pulmonary hypertension, examined by direct measurement of right ventricular end systolic pressure (RVESP), right ventricular hypertrophy (n = 8), and hematocrit (n = 3), was induced by 3 weeks of hypobaric-hypoxia in wild-type and COX-knockout (KO) mice. RVESP was increased in wild-type hypoxic mice compared with normoxic controls (24.4 +/- 1.4 versus 13.8 +/- 1.9 mm Hg; n = 8; p < 0.05). COX-2 KO mice showed a greater increase in RVESP following hypoxia (36.8 +/- 2.7 mm Hg; p < 0.05). Urinary thromboxane (TX)B(2) excretion increased following hypoxia (44.6 +/- 11.1 versus 14.7 +/- 1.8 ng/ml; n = 6; p < 0.05), an effect that was exacerbated by COX-2 gene disruption (54.5 +/- 10.8 ng/ml; n = 6). In contrast, the increase in 6-keto-prostacyclin(1alpha) excretion following hypoxia was reduced by COX-2 gene disruption (29 +/- 3 versus 52 +/- 4.6 ng/ml; p < 0.01). Tail cut bleed times were lower following hypoxia, and there was evidence of intravascular thrombosis in lung vessels that was exacerbated by disruption of COX-2 and reduced by deletion of COX-1. The TXA(2)/endoperoxide receptor antagonist ifetroban (50 mg/kg/day) offset the effect of deleting the COX-2 gene, attenuating the hypoxia-induced rise in RVESP and intravascular thrombosis. COX-2 gene deletion exacerbates pulmonary hypertension, enhances sensitivity to TXA(2), and induces intravascular thrombosis in response to hypoxia. The data provide evidence that endogenous prostaglandins modulate the pulmonary response to hypoxia.


Asunto(s)
Ciclooxigenasa 2/fisiología , Hipertensión Pulmonar/enzimología , Hipoxia/enzimología , Trombosis de la Vena/enzimología , Animales , Ciclooxigenasa 2/genética , Femenino , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Hipoxia/complicaciones , Hipoxia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Trombosis de la Vena/etiología , Trombosis de la Vena/genética
14.
Anesth Analg ; 106(1): 294-300, table of contents, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18165593

RESUMEN

BACKGROUND: Cyclooxygenase 2 inhibition has proven analgesic efficacy in a variety of surgical procedures. We postulated that perioperative cyclooxygenase 2 inhibition significantly reduces postoperative morphine requirements after major thoracic surgery and investigated the site of this potential analgesic effect. METHODS: Ninety-two patients participated in this single-center, double-blind, randomized, placebo-controlled, parallel-group trial. Patients between the ages of 18 and 80 yr undergoing a thoracotomy or median sternotomy were randomized to receive either nimesulide or placebo in combination with a standard analgesic regimen perioperatively. Nimesulide was administered orally the evening before surgery and at 12-h intervals for 5 days postoperatively. The primary efficacy variables were morphine consumption and pain scores for the first 48 h postoperatively. The secondary efficacy variable was the effect of nimesulide on cyclooxygenase activity in cerebrospinal fluid (CSF). RESULTS: Pain scores at rest or with movement, and total morphine consumption for the first 48 h postoperatively, were not statistically different between the groups. The mean difference in total morphine consumption up to 48 h postoperatively between the nimesulide and placebo group was a 9.0 mg reduction (95% CI: -28.9 to 10.9 mg) (P = 0.37). Adjusted mean (se) CSF 6-keto-PGF1alpha (6-keto-PGF1alpha) concentrations increased by 54.7 (25.7) pg/mL from preoperatively to Day + 2 postoperatively in the placebo group, whereas adjusted mean (se) CSF 6-keto-PGF1alpha concentration decreased by 0.6 pg/mL (18.2 pg/mL) in the nimesulide group. These changes were not statistically different between the groups (P = 0.095). CONCLUSION: Nimesulide, at a dose of 90 mg twice daily in combination with a standard analgesic regimen, does not influence pain scores, morphine requirements, or CSF prostaglandin levels after major thoracic surgery.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Morfina/uso terapéutico , Dolor Postoperatorio/prevención & control , Esternón/cirugía , Sulfonamidas/uso terapéutico , Toracotomía , 6-Cetoprostaglandina F1 alfa/líquido cefalorraquídeo , Administración Oral , Anciano , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Método Doble Ciego , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Estudios Prospectivos , Prostaglandina-Endoperóxido Sintasas/líquido cefalorraquídeo , Sulfonamidas/administración & dosificación , Factores de Tiempo , Resultado del Tratamiento
15.
Curr Eye Res ; 33(2): 177-84, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18293189

RESUMEN

PURPOSE: The expression of cyclooxygenase-2 (COX-2) and its prognostic value in uveal melanoma was examined. METHODS: Paraffin-embedded sections from 32 clinicopathologically well-characterized cases of primary uveal melanoma were immunohistochemically stained for COX-2. COX-2 expression was evaluated in terms of both the intensity and the extent of staining for each tumor. A COX-2 score encompassing both intensity and extent was also calculated for each specimen. RESULTS: 29 specimens (90.6%) contained moderate or intense positive immunoreactivity for COX-2. A statistically significant association (p<0.05) between COX-2 expression (intensity and score) and metastatic death was established. CONCLUSION: Upregulation of COX-2 expression appears to be associated with poor prognosis in uveal melanoma.


Asunto(s)
Neoplasias de la Coroides/enzimología , Ciclooxigenasa 2/metabolismo , Melanoma/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Coroides/mortalidad , Neoplasias de la Coroides/patología , Femenino , Humanos , Técnicas para Inmunoenzimas , Masculino , Melanoma/mortalidad , Melanoma/secundario , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
16.
Clin Pharmacol Ther ; 103(6): 1047-1051, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28905988

RESUMEN

Low-dose aspirin (75 mg or 81 mg) is considered to be the lowest effective dose for cardiovascular protection; however, the use of enteric preparations has created a source of variability in bioavailability. As part of regulatory requirements, we carried out bioequivalence tests for two 75 mg enteric-coated aspirin preparations (Caprin and Protek) using Nu-Seals 75 mg aspirin as the comparator. The primary endpoint was serum thromboxane levels after 14 days of treatment. Protek failed to meet bioequivalence, as it was significantly less effective than Nu-Seals. In contrast, Caprin was not bioequivalent with Nu-Seals but as it was more effective it was granted approval. However, 75 mg plain aspirin was found to be more effective than Nu-Seals at inhibiting serum thromboxane production. Thus, there is significant variation in the ability of low-dose aspirin preparations to inhibit serum thromboxane production.


Asunto(s)
Aspirina/farmacocinética , Inhibidores de Agregación Plaquetaria/farmacocinética , Ácido Araquidónico/metabolismo , Aspirina/administración & dosificación , Relación Dosis-Respuesta a Droga , Humanos , Fragmentos de Péptidos/metabolismo , Inhibidores de Agregación Plaquetaria/administración & dosificación , Comprimidos Recubiertos , Equivalencia Terapéutica , Tromboxano A2/metabolismo
17.
Atherosclerosis ; 269: 42-49, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29258006

RESUMEN

BACKGROUND AND AIMS: Elevated urinary 11-dehydro thromboxane B2 (TxB2), a measure of thromboxane A2 formation in vivo, predicts future atherothrombotic events. To further understand this relationship, the genetic determinants of 11-dehydro TxB2 and their associations with cardiovascular morbidity were investigated in this study. METHODS: Genome-wide and targeted genetic association studies of urinary 11-dehydro TxB2 were conducted in 806 Anglo-Scandinavian Cardiac Outcomes Trial (ASCOT) participants. RESULTS: The strongest associations were in PPARGC1B (rs4235745, rs32582, rs10515638) and CNTN4 (rs10510230, rs4684343), these 5 single nucleotide polymorphisms (SNPs) were independently associated with 11-dehydro TxB2 formation. Haplotypes of 11-dehydro TxB2 increasing alleles for both PPARGC1B and CNTN4 were significantly associated with 11-dehydro TxB2, explaining 5.2% and 4.5% of the variation in the whole cohort, and 8.8% and 7.9% in participants not taking aspirin, respectively. In a second ASCOT population (n = 6199), addition of these 5 SNPs significantly improved the covariate-only Cox proportional hazards model for cardiovascular events (chisq = 14.7, p=0.01). Two of the risk alleles associated with increased urinary 11-dehydro TxB2 were individually associated with greater incidences of cardiovascular events - rs10515638 (HR = 1.31, p=0.01) and rs10510230 (HR = 1.25, p=0.007); effect sizes were larger in those not taking aspirin. CONCLUSIONS: PPARGC1B and CNTN4 genotypes are associated with elevated thromboxane A2 formation and with an excess of cardiovascular events. Aspirin appears to blunt these associations. If specific protection of PPARGC1B and CNTN4 variant carriers by aspirin is confirmed by additional studies, PPARGC1B and CNTN4 genotyping could potentially assist in clinical decision making regarding the use of aspirin in primary prevention.


Asunto(s)
Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/genética , Proteínas Portadoras/genética , Contactinas/genética , Polimorfismo de Nucleótido Simple , Tromboxano A2/metabolismo , Anciano , Aspirina/uso terapéutico , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/etnología , Enfermedades Cardiovasculares/prevención & control , Europa (Continente)/epidemiología , Femenino , Frecuencia de los Genes , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Estudios Multicéntricos como Asunto , Fenotipo , Prevención Primaria , Supervivencia sin Progresión , Proteínas de Unión al ARN , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Riesgo , Tromboxano B2/análogos & derivados , Tromboxano B2/orina , Factores de Tiempo , Población Blanca/genética
18.
Methods Mol Biol ; 357: 307-11, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17172696

RESUMEN

This chapter describes an approach to isolate, separate, and identify the contents of the platelet releasate, a fraction highly enriched for platelet granular and exosomal contents. Investigation into such a fraction will improve our understanding of platelet interactions with other cells, vascular remodeling, coagulation, and vessel growth.


Asunto(s)
Plaquetas/metabolismo , Proteínas/análisis , Proteómica/métodos , Plaquetas/citología , Humanos
19.
Can J Gastroenterol ; 21(6): 367-70, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17571170

RESUMEN

BACKGROUND AND AIMS: Helicobacter pylori is the major causative agent in peptic ulcer disease and is strongly implicated in the development of gastric cancer. It has also been linked, less strongly, to cardiovascular disease. The mechanisms by which certain strains of H pylori induce platelet aggregation through interactions with platelet glycoprotein Ib have been previously described. METHODS: In the present study, 21 different strains of H pylori, varying in their vacuolating toxin gene, cytotoxic-associated gene A status and other pathogenicity factors, were tested for their ability to induce platelet aggregation. RESULTS: Ten of the 21 strains induced platelet aggregation, a response that appeared to be independent of their vacuolating toxin gene and cytotoxic-associated gene A status. CONCLUSIONS: Platelet aggregation has been suggested to be one of the possible mechanisms involved in the effects on the cardiovascular system induced by H pylori. Our results suggest that any putative role H pylori plays in cardiovascular disease may be strain dependent. Further work to identify the H pylori factors involved in induction of platelet aggregation may allow for identification of 'higher risk' strains for cardiovascular disease.


Asunto(s)
Helicobacter pylori/fisiología , Agregación Plaquetaria , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Humanos , Técnicas In Vitro
20.
Stroke ; 37(8): 2153-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16794200

RESUMEN

BACKGROUND AND PURPOSE: Aspirin resistance may be relatively common and associated with adverse outcome. Meta-analysis has clearly shown that 75 mg plain aspirin is the lowest effective dose; however, it is not known whether the recent increased use of enteric-coated aspirin could account for aspirin resistance. This study was designed to determine whether enteric-coated aspirin is as effective as plain aspirin in healthy volunteers. METHODS: Seventy-one healthy volunteers were enrolled in 3 separate bioequivalence studies. Using a crossover design, each volunteer took 2 different aspirin preparations. Five aspirin preparations were evaluated, 3 different enteric-coated 75-mg aspirins, dispersible aspirin 75 mg and asasantin (25-mg standard release aspirin plus 200-mg modified-release dipyridamole given twice daily). Serum thromboxane (TX) B2 levels and arachidonic acid-induced platelet aggregation were measured before and after 14 days of treatment. RESULTS: All other aspirin preparations tested were inferior to dispersible aspirin (P<0.001) in their effect on serum TXB(2) level. Treatment failure (<95% inhibition serum TXB2 formation) occurred in 14 subjects, none of whom were taking dispersible aspirin. Mean weight for those demonstrating treatment failure was greater than those with complete TXB2 (>99%) inhibition (P<0.001). Using logistic regression analysis an 80-kg subject had a 20% probability of treatment failure. Asasantin was the most potent preparation in terms of inhibition of platelet aggregation. CONCLUSIONS: Equivalent doses of the enteric-coated aspirin were not as effective as plain aspirin. Lower bioavailability of these preparations and poor absorption from the higher pH environment of the small intestine may result in inadequate platelet inhibition, particularly in heavier subjects.


Asunto(s)
Aspirina/administración & dosificación , Aspirina/farmacocinética , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacocinética , Adulto , Aspirina/farmacología , Combinación Aspirina y Dipiridamol , Peso Corporal , Estudios Cruzados , Dipiridamol/farmacología , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Humanos , Modelos Logísticos , Persona de Mediana Edad , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Valores de Referencia , Comprimidos Recubiertos , Equivalencia Terapéutica , Tromboxano B2/sangre , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA