Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(1): 271-286, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823267

RESUMEN

BACKGROUND: Prostacyclin is a fundamental signaling pathway traditionally associated with the cardiovascular system and protection against thrombosis but which also has regulatory functions in fibrosis, proliferation, and immunity. Prevailing dogma states that prostacyclin is principally derived from vascular endothelium, although it is known that other cells can also synthesize it. However, the role of nonendothelial sources in prostacyclin production has not been systematically evaluated resulting in an underappreciation of their importance relative to better characterized endothelial sources. METHODS: To address this, we have used novel endothelial cell-specific and fibroblast-specific COX (cyclo-oxygenase) and prostacyclin synthase knockout mice and cells freshly isolated from mouse and human lung tissue. We have assessed prostacyclin release by immunoassay and thrombosis in vivo using an FeCl3-induced carotid artery injury model. RESULTS: We found that in arteries, endothelial cells are the main source of prostacyclin but that in the lung, and other tissues, prostacyclin production occurs largely independently of endothelial and vascular smooth muscle cells. Instead, in mouse and human lung, prostacyclin production was strongly associated with fibroblasts. By comparison, microvascular endothelial cells from the lung showed weak prostacyclin synthetic capacity compared with those isolated from large arteries. Prostacyclin derived from fibroblasts and other nonendothelial sources was seen to contribute to antithrombotic protection. CONCLUSIONS: These observations define a new paradigm in prostacyclin biology in which fibroblast/nonendothelial-derived prostacyclin works in parallel with endothelium-derived prostanoids to control thrombotic risk and potentially a broad range of other biology. Although generation of prostacyclin by fibroblasts has been shown previously, the scale and systemic activity was unappreciated. As such, this represents a basic change in our understanding and may provide new insight into how diseases of the lung result in cardiovascular risk.


Asunto(s)
Epoprostenol , Trombosis , Ratones , Humanos , Animales , Fibrinolíticos , Células Endoteliales/metabolismo , Prostaglandinas I/metabolismo , Prostaglandinas I/farmacología , Endotelio Vascular/metabolismo , Ratones Noqueados , Fibroblastos/metabolismo , Trombosis/genética , Trombosis/prevención & control , Trombosis/metabolismo
2.
FASEB J ; 34(1): 1745-1754, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914612

RESUMEN

Blood vessels are comprised of endothelial and smooth muscle cells. Obtaining both types of cells from vessels of living donors is not possible without invasive surgery. To address this, we have devised a strategy whereby human endothelial and smooth muscle cells derived from blood progenitors from the same donor could be cultured with autologous leukocytes to generate a same donor "vessel in a dish" bioassay. Autologous sets of blood outgrowth endothelial cells (BOECs), smooth muscle cells (BO-SMCs), and leukocytes were obtained from four donors. Cells were treated in monoculture and cumulative coculture conditions. The endothelial specific mediator endothelin-1 along with interleukin (IL)-6, IL-8, tumor necrosis factor α, and interferon gamma-induced protein 10 were measured under control culture conditions and after stimulation with cytokines. Cocultures remained viable throughout. The profile of individual mediators released from cells was consistent with what we know of endothelial and smooth muscle cells cultured from blood vessels. For the first time, we report a proof of concept study where autologous blood outgrowth "vascular" cells and leukocytes were studied alone and in coculture. This novel bioassay has usefulness in vascular biology research, patient phenotyping, drug testing, and tissue engineering.


Asunto(s)
Células Endoteliales/fisiología , Leucocitos/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Bioensayo/métodos , Células Cultivadas , Técnicas de Cocultivo/métodos , Citocinas/metabolismo , Descubrimiento de Drogas/métodos , Células Endoteliales/metabolismo , Humanos , Interleucina-6/metabolismo , Leucocitos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , Ingeniería de Tejidos/métodos , Factor de Necrosis Tumoral alfa/metabolismo
3.
FASEB J ; 34(8): 10027-10040, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32592197

RESUMEN

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Asunto(s)
Aspirina/farmacología , Plaquetas/metabolismo , Ciclooxigenasa 1/fisiología , Inhibidores de la Ciclooxigenasa/farmacología , Eicosanoides/metabolismo , Proteínas de la Membrana/fisiología , Trombosis/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , Plaquetas/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombosis/tratamiento farmacológico , Trombosis/patología
4.
Circ Res ; 125(9): 847-854, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31510878

RESUMEN

RATIONALE: Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo. OBJECTIVE: We sought to identify the role, if any, of COX1-derived prostacyclin in antithrombotic protection in vivo and compare this to the established protective role of COX2. METHODS AND RESULTS: We developed vascular-specific COX1 knockout mice and studied them alongside endothelial-specific COX2 knockout mice. COX1 immunoreactivity and prostacyclin production were primarily associated with the endothelial layer of aortae; freshly isolated aortic ECs released >10-fold more prostacyclin than smooth muscle cells. Moreover, aortic prostacyclin production, the ability of aortic rings to inhibit platelet aggregation and plasma prostacyclin levels were reduced when COX1 was knocked out in ECs but not in smooth muscle cells. When thrombosis was measured in vivo after FeCl3 carotid artery injury, endothelial COX1 deletion accelerated thrombosis to a similar extent as prostacyclin receptor blockade. However, this effect was lost when COX1 was deleted from both ECs and platelets. Deletion of COX2 from ECs also resulted in a prothrombotic phenotype that was independent of local vascular prostacyclin production. CONCLUSIONS: These data demonstrate for the first time that, in healthy animals, endothelial COX1 provides an essential antithrombotic tone, which is masked when COX1 activity is lost in both ECs and platelets. These results help us define a new 2-component paradigm wherein thrombotic tone is regulated by both COX1 and COX2 through complementary but mechanistically distinct pathways.


Asunto(s)
Ciclooxigenasa 1/deficiencia , Endotelio Vascular/metabolismo , Epoprostenol/metabolismo , Fibrinolíticos/metabolismo , Eliminación de Gen , Proteínas de la Membrana/deficiencia , Agregación Plaquetaria/fisiología , Animales , Aorta/metabolismo , Ciclooxigenasa 1/genética , Epoprostenol/genética , Femenino , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos
5.
Br J Clin Pharmacol ; 87(3): 776-784, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32805057

RESUMEN

AIMS: In light of the recent safety concerns relating to NSAID use in COVID-19, we sought to evaluate cardiovascular and respiratory complications in patients taking NSAIDs during acute lower respiratory tract infections. METHODS: We carried out a systematic review of randomised controlled trials and observational studies. Studies of adult patients with short-term NSAID use during acute lower respiratory tract infections, including bacterial and viral infections, were included. Primary outcome was all-cause mortality. Secondary outcomes were cardiovascular, renal and respiratory complications. RESULTS: In total, eight studies including two randomised controlled trials, three retrospective and three prospective observational studies enrolling 44 140 patients were included. Five of the studies were in patients with pneumonia, two in patients with influenza, and one in a patient with acute bronchitis. Meta-analysis was not possible due to significant heterogeneity. There was a trend towards a reduction in mortality and an increase in pleuro-pulmonary complications. However, all studies exhibited high risks of bias, primarily due to lack of adjustment for confounding variables. Cardiovascular outcomes were not reported by any of the included studies. CONCLUSION: In this systematic review of NSAID use during acute lower respiratory tract infections in adults, we found that the existing evidence for mortality, pleuro-pulmonary complications and rates of mechanical ventilation or organ failure is of extremely poor quality, very low certainty and should be interpreted with caution. Mechanistic and clinical studies addressing the captioned subject are urgently needed, especially in relation to COVID-19.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Ibuprofeno/uso terapéutico , COVID-19/complicaciones , COVID-19/mortalidad , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/mortalidad , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto
6.
Circ Res ; 122(4): 555-559, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29298774

RESUMEN

RATIONALE: The balance between vascular prostacyclin, which is antithrombotic, and platelet thromboxane A2, which is prothrombotic, is fundamental to cardiovascular health. Prostacyclin and thromboxane A2 are formed after the concerted actions of cPLA2α (cytosolic phospholipase A2) and COX (cyclooxygenase). Urinary 2,3-dinor-6-keto-PGF1α (PGI-M) and 11-dehydro-TXB2 (TX-M) have been taken as biomarkers of prostacyclin and thromboxane A2 formation within the circulation and used to explain COX biology and patient phenotypes, despite concerns that urinary PGI-M and TX-M originate in the kidney. OBJECTIVE: We report data from a remarkable patient carrying an extremely rare genetic mutation in cPLA2α, causing almost complete loss of prostacyclin and thromboxane A2, who was transplanted with a normal kidney resulting in an experimental scenario of whole-body cPLA2α knockout, kidney-specific knockin. By studying this patient, we can determine definitively the contribution of the kidney to the productions of PGI-M and TX-M and test their validity as markers of prostacyclin and thromboxane A2 in the circulation. METHODS AND RESULTS: Metabolites were measured using liquid chromatography-tandem mass spectrometry. Endothelial cells were grown from blood progenitors. Before kidney transplantation, the patient's endothelial cells and platelets released negligible levels of prostacyclin (measured as 6-keto-prostaglandin F1α) and thromboxane A2 (measured as TXB2), respectively. Likewise, the urinary levels of PGI-M and TX-M were very low. After transplantation and the establishment of normal renal function, the levels of PGI-M and TX-M in the patient's urine rose to within normal ranges, whereas endothelial production of prostacyclin and platelet production of thromboxane A2 remained negligible. CONCLUSIONS: These data show that PGI-M and TX-M can be derived exclusively from the kidney without contribution from prostacyclin made by endothelial cells or thromboxane A2 by platelets in the general circulation. Previous work relying on urinary metabolites of prostacyclin and thromboxane A2 as markers of whole-body endothelial and platelet function now requires reevaluation.


Asunto(s)
6-Cetoprostaglandina F1 alfa/análogos & derivados , Aloinjertos/metabolismo , Trasplante de Riñón , Riñón/metabolismo , Mutación con Pérdida de Función , Fosfolipasas A2 Citosólicas/genética , Tromboxano B2/análogos & derivados , 6-Cetoprostaglandina F1 alfa/metabolismo , 6-Cetoprostaglandina F1 alfa/orina , Biomarcadores/orina , Células Cultivadas , Femenino , Humanos , Persona de Mediana Edad , Fenotipo , Fosfolipasas A2 Citosólicas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Tromboxano B2/metabolismo , Tromboxano B2/orina
7.
Proc Natl Acad Sci U S A ; 113(2): 434-9, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26712011

RESUMEN

Cyclooxygenase-2 (COX-2) is an inducible enzyme that drives inflammation and is the therapeutic target for widely used nonsteroidal antiinflammatory drugs (NSAIDs). However, COX-2 is also constitutively expressed, in the absence of overt inflammation, with a specific tissue distribution that includes the kidney, gastrointestinal tract, brain, and thymus. Constitutive COX-2 expression is therapeutically important because NSAIDs cause cardiovascular and renal side effects in otherwise healthy individuals. These side effects are now of major concern globally. However, the pathways driving constitutive COX-2 expression remain poorly understood. Here we show that in the kidney and other sites, constitutive COX-2 expression is a sterile response, independent of commensal microorganisms and not associated with activity of the inflammatory transcription factor NF-κB. Instead, COX-2 expression in the kidney but not other regions colocalized with nuclear factor of activated T cells (NFAT) transcription factor activity and was sensitive to inhibition of calcineurin-dependent NFAT activation. However, calcineurin/NFAT regulation did not contribute to constitutive expression elsewhere or to inflammatory COX-2 induction at any site. These data address the mechanisms driving constitutive COX-2 and suggest that by targeting transcription it may be possible to develop antiinflammatory therapies that spare the constitutive expression necessary for normal homeostatic functions, including those important to the cardiovascular-renal system.


Asunto(s)
Ciclooxigenasa 2/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Transducción de Señal , Transcripción Genética , Animales , Ciclooxigenasa 2/metabolismo , Ciclosporina/farmacología , Citocinas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Vida Libre de Gérmenes , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipopolisacáridos/farmacología , Luciferasas/metabolismo , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Distribución Tisular/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
8.
FASEB J ; 31(6): 2439-2445, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28246166

RESUMEN

IL-1ß release is integral to the innate immune system. The release of mature IL-1ß depends on 2 regulated events: the de novo induction of pro-IL-1ß, generally via NF-κB-dependent transduction pathways; and the assembly and activation of the NLRP3 inflammasome. This latter step is reliant on active caspase-1, pannexin-1, and P2X7 receptor activation. Pathogen-associated molecular patterns in gram-positive and gram-negative bacteria activate IL-1ß release from immune cells via TLR2 and TLR4 receptors, respectively. We found that pro-IL-1ß and mature IL-1ß release from human monocytes is stimulated by the TLR2 agonists Pam3CSK4 or FSL-1, as well as the TLR4 agonist LPS in the absence of additional ATP. TLR2 agonists required pannexin-1 and P2X7 receptor activation to stimulate IL-1ß release. In contrast, IL-1ß release stimulated by the TLR4 agonist LPS is independent of both pannexin-1 and P2X7 activation. In the absence of exogenous ATP, P2X7 activation requires endogenous ATP release, which occurs in some cells via pannexin-1. In line with this, we found that LPS-stimulated human monocytes released relatively low levels of ATP, whereas cells stimulated with TLR2 agonists released high levels of ATP. These findings suggest that in human monocytes, both TLR2 and TLR4 signaling induce pro-IL-1ß expression, but the mechanism by which they activate caspase-1 diverges at the level of the pannexin-1/ATP/P2X7 axis.-Parzych, K., Zetterqvist, A. V., Wright, W. R., Kirkby, N. S., Mitchell, J. A., Paul-Clark, M. J. Differential role of pannexin-1/ATP/P2X7 axis in IL-1ß release by human monocytes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Interleucina-1beta/metabolismo , Monocitos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular Tumoral , Conexinas/genética , Diglicéridos/farmacología , Regulación de la Expresión Génica/fisiología , Humanos , Interleucina-1beta/genética , Lipopéptidos/farmacología , Lipopolisacáridos/farmacología , Proteínas del Tejido Nervioso/genética , Oligopéptidos/farmacología , Receptores Purinérgicos P2X7/genética , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
Blood ; 126(10): e11-8, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26215112

RESUMEN

Testing of platelet function is central to the cardiovascular phenotyping of genetically modified mice. Traditional platelet function tests have been developed primarily for testing human samples and the volumes required make them highly unsuitable for the testing of mouse platelets. This limits research in this area. To address this problem, we have developed a miniaturized whole blood aggregometry assay, based on a readily accessible 96-well plate format coupled with quantification of single platelet depletion by flow cytometric analysis. Using this approach, we observed a concentration-dependent loss of single platelets in blood exposed to arachidonic acid, collagen, U46619 or protease activated receptor 4 activating peptide. This loss was sensitive to well-established antiplatelet agents and genetic manipulation of platelet activation pathways. Observations were more deeply analyzed by flow cytometric imaging, confocal imaging, and measurement of platelet releasates. Phenotypic analysis of the reactivity of platelets taken from mice lacking intercellular adhesion molecule (ICAM)-1 identified a marked decrease in fibrinogen-dependent platelet-monocyte interactions, especially under inflammatory conditions. Such findings exemplify the value of screening platelet phenotypes of genetically modified mice and shed further light upon the roles and interactions of platelets in inflammation.


Asunto(s)
Plaquetas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Monocitos/metabolismo , Agregación Plaquetaria/fisiología , Pruebas de Función Plaquetaria/métodos , Animales , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fenotipo , Sensibilidad y Especificidad
10.
FASEB J ; 30(12): 4256-4266, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27633788

RESUMEN

Regular consumption of low-dose aspirin reduces the occurrence of colorectal, esophageal, stomach, and gastrointestinal cancers. The underlying mechanism is unknown but may be linked to inhibition of angiogenesis. Because the effective doses of aspirin are consistent with the inhibition of cyclooxygenase-1 in platelets, we used liquid chromatography with tandem mass spectrometry analyses and immunoassays of human platelet releasates coupled with angiogenesis assays to search for the mediators of these effects. Blood or platelet-rich plasma from healthy volunteers stimulated with platelet activators produced a broad range of eicosanoids. Notably, preincubation of platelets with aspirin, but not with a P2Y12 receptor antagonist, caused a marked reduction in the production of 11-hydroxyeicosatetraenoic acid (HETE) and 15(S)-HETE, in addition to prostanoids such as thromboxane A2 Releasates from activated platelets caused cell migration and tube formation in cultured human endothelial cells and stimulated the sprouting of rat aortic rings in culture. These proangiogenic effects were absent when platelets were treated with aspirin but returned by coincubation with exogenous 15(S)-HETE. These results reveal 15(S)-HETE as a major platelet cyclooxygenase-1 product with strong proangiogenic effects. Thus, 15(S)-HETE represents a potential target for the development of novel antiangiogenic therapeutics, and blockade of its production may provide a mechanism for the anticancer effects of aspirin.-Rauzi, F., Kirkby, N. S., Edin, M. L., Whiteford, J. Zeldin, D. C., Mitchell, J. A., Warner, T. D. Aspirin inhibits the production of proangiogenic 15(S)-HETE by platelet cyclooxygenase-1.


Asunto(s)
Aspirina/farmacología , Plaquetas/efectos de los fármacos , Ciclooxigenasa 1/metabolismo , Endotelio/efectos de los fármacos , Ácidos Hidroxieicosatetraenoicos/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Células Cultivadas , Ciclooxigenasa 1/efectos de los fármacos , Eicosanoides/metabolismo , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo
11.
FASEB J ; 30(12): 4172-4179, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27601438

RESUMEN

Nonsteroidal antiinflammatory drugs, including ibuprofen, are among the most commonly used medications and produce their antiinflammatory effects by blocking cyclooxygenase (COX)-2. Their use is associated with increased risk of heart attacks caused by blocking COX-2 in the vasculature and/or kidney, with our recent work implicating the endogenous NOS inhibitor asymmetric dimethylarginine (ADMA), a cardiotoxic hormone whose effects can be prevented by l-arginine. The ibuprofen salt ibuprofen arginate (Spididol) was created to increase solubility but we suggest that it could also augment the NO pathway through codelivery of arginine. Here we investigated the idea that ibuprofen arginate can act to simultaneously inhibit COX-2 and preserve the NO pathway. Ibuprofen arginate functioned similarly to ibuprofen sodium for inhibition of mouse/human COX-2, but only ibuprofen arginate served as a substrate for NOS. Ibuprofen arginate but not ibuprofen sodium also reversed the inhibitory effects of ADMA and NG-nitro-l-arginine methyl ester on inducible NOS (macrophages) and endothelial NOS in vitro (aorta) and in vivo (blood pressure). These observations show that ibuprofen arginate provides, in one preparation, a COX-2 inhibitor and NOS substrate that could act to negate the harmful cardiovascular consequences mediated by blocking renal COX-2 and increased ADMA. While remarkably simple, our findings are potentially game-changing in the nonsteroidal antiinflammatory drug arena.-Kirkby, N. S., Tesfai, A., Ahmetaj-Shala, B., Gashaw, H. H., Sampaio, W., Etelvino, G., Leão, N. M., Santos, R. A., Mitchell, J. A. Ibuprofen arginate retains eNOS substrate activity and reverses endothelial dysfunction: implications for the COX-2/ADMA axis.


Asunto(s)
Arginina/análogos & derivados , Ciclooxigenasa 2/metabolismo , Ibuprofeno/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Arginina/metabolismo , Arginina/farmacología , Combinación de Medicamentos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Riñón/metabolismo , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Especificidad por Sustrato
12.
Circulation ; 131(7): 633-42, 2015 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-25492024

RESUMEN

BACKGROUND: Cardiovascular side effects associated with cyclooxygenase-2 inhibitor drugs dominate clinical concern. Cyclooxygenase-2 is expressed in the renal medulla where inhibition causes fluid retention and increased blood pressure. However, the mechanisms linking cyclooxygenase-2 inhibition and cardiovascular events are unknown and no biomarkers have been identified. METHODS AND RESULTS: Transcriptome analysis of wild-type and cyclooxygenase-2(-/-) mouse tissues revealed 1 gene altered in the heart and aorta, but >1000 genes altered in the renal medulla, including those regulating the endogenous nitric oxide synthase inhibitors asymmetrical dimethylarginine (ADMA) and monomethyl-l-arginine. Cyclo-oxygenase-2(-/-) mice had increased plasma levels of ADMA and monomethyl-l-arginine and reduced endothelial nitric oxide responses. These genes and methylarginines were not similarly altered in mice lacking prostacyclin receptors. Wild-type mice or human volunteers taking cyclooxygenase-2 inhibitors also showed increased plasma ADMA. Endothelial nitric oxide is cardio-protective, reducing thrombosis and atherosclerosis. Consequently, increased ADMA is associated with cardiovascular disease. Thus, our study identifies ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction with nonsteroidal anti-inflammatory drug usage. CONCLUSIONS: We identify the endogenous endothelial nitric oxide synthase inhibitor ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction.


Asunto(s)
Antiinflamatorios/efectos adversos , Arginina/análogos & derivados , Enfermedades Cardiovasculares/sangre , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Ciclooxigenasa 2/deficiencia , Adulto , Animales , Arginina/sangre , Biomarcadores/sangre , Enfermedades Cardiovasculares/tratamiento farmacológico , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Adulto Joven
13.
Blood ; 123(8): e11-22, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24408324

RESUMEN

Up to 1% of the population have mild bleeding disorders, but these remain poorly characterized, particularly with regard to the roles of platelets. We have compared the usefulness of Optimul, a 96-well plate-based assay of 7 distinct pathways of platelet activation to characterize inherited platelet defects in comparison with light transmission aggregometry (LTA). Using Optimul and LTA, concentration-response curves were generated for arachidonic acid, ADP, collagen, epinephrine, Thrombin receptor activating-peptide, U46619, and ristocetin in samples from (1) healthy volunteers (n = 50), (2) healthy volunteers treated with antiplatelet agents in vitro (n = 10), and (3) patients with bleeding of unknown origin (n = 65). The assays gave concordant results in 82% of cases (κ = 0.62, P < .0001). Normal platelet function results were particularly predictive (sensitivity, 94%; negative predictive value, 91%), whereas a positive result was not always substantiated by LTA (specificity, 67%; positive predictive value, 77%). The Optimul assay was significantly more sensitive at characterizing defects in the thromboxane pathway, which presented with normal responses with LTA. The Optimul assay is sensitive to mild platelet defects, could be used as a rapid screening assay in patients presenting with bleeding symptoms, and detects changes in platelet function more readily than LTA. This trial was registered at www.isrctn.org as #ISRCTN 77951167.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/diagnóstico , Monitoreo de Drogas/métodos , Hemorragia/diagnóstico , Ensayos Analíticos de Alto Rendimiento/métodos , Activación Plaquetaria/fisiología , Inhibidores de Agregación Plaquetaria/farmacología , Adulto , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/genética , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Femenino , Estudios de Asociación Genética , Voluntarios Sanos , Hemorragia/sangre , Hemorragia/fisiopatología , Humanos , Masculino , Activación Plaquetaria/efectos de los fármacos , Valor Predictivo de las Pruebas , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Sensibilidad y Especificidad , Adulto Joven
14.
FASEB J ; 29(11): 4568-78, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26183771

RESUMEN

Eicosanoids are important vascular regulators, but the phospholipase A2 (PLA2) isoforms supporting their production within the cardiovascular system are not fully understood. To address this, we have studied platelets, endothelial cells, and leukocytes from 2 siblings with a homozygous loss-of-function mutation in group IVA cytosolic phospholipase A2 (cPLA2α). Chromatography/mass spectrometry was used to determine levels of a broad range of eicosanoids produced by isolated vascular cells, and in plasma and urine. Eicosanoid release data were paired with studies of cellular function. Absence of cPLA2α almost abolished eicosanoid synthesis in platelets (e.g., thromboxane A2, control 20.5 ± 1.4 ng/ml vs. patient 0.1 ng/ml) and leukocytes [e.g., prostaglandin E2 (PGE2), control 21.9 ± 7.4 ng/ml vs. patient 1.9 ng/ml], and this was associated with impaired platelet activation and enhanced inflammatory responses. cPLA2α-deficient endothelial cells showed reduced, but not absent, formation of prostaglandin I2 (prostacyclin; control 956 ± 422 pg/ml vs. patient 196 pg/ml) and were primed for inflammation. In the urine, prostaglandin metabolites were selectively influenced by cPLA2α deficiency. For example, prostacyclin metabolites were strongly reduced (18.4% of control) in patients lacking cPLA2α, whereas PGE2 metabolites (77.8% of control) were similar to healthy volunteer levels. These studies constitute a definitive account, demonstrating the fundamental role of cPLA2α to eicosanoid formation and cellular responses within the human circulation.


Asunto(s)
Antígenos de Plaqueta Humana/genética , Plaquetas/enzimología , Dinoprostona/genética , Células Endoteliales/enzimología , Epoprostenol/genética , Leucocitos/enzimología , Mutación , Adulto , Plaquetas/patología , Dinoprostona/biosíntesis , Células Endoteliales/patología , Epoprostenol/biosíntesis , Femenino , Humanos , Leucocitos/patología , Masculino , Activación Plaquetaria/genética
15.
Circ Res ; 114(4): 677-88, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24334027

RESUMEN

RATIONALE: Evidence is increasing of a link between interferon (IFN) and pulmonary arterial hypertension (PAH). Conditions with chronically elevated endogenous IFNs such as systemic sclerosis are strongly associated with PAH. Furthermore, therapeutic use of type I IFN is associated with PAH. This was recognized at the 2013 World Symposium on Pulmonary Hypertension where the urgent need for research into this was highlighted. OBJECTIVE: To explore the role of type I IFN in PAH. METHODS AND RESULTS: Cells were cultured using standard approaches. Cytokines were measured by ELISA. Gene and protein expression were measured using reverse transcriptase polymerase chain reaction, Western blotting, and immunohistochemistry. The role of type I IFN in PAH in vivo was determined using type I IFN receptor knockout (IFNAR1(-/-)) mice. Human lung cells responded to types I and II but not III IFN correlating with relevant receptor expression. Type I, II, and III IFN levels were elevated in serum of patients with systemic sclerosis associated PAH. Serum interferon γ inducible protein 10 (IP10; CXCL10) and endothelin 1 were raised and strongly correlated together. IP10 correlated positively with pulmonary hemodynamics and serum brain natriuretic peptide and negatively with 6-minute walk test and cardiac index. Endothelial cells grown out of the blood of PAH patients were more sensitive to the effects of type I IFN than cells from healthy donors. PAH lung demonstrated increased IFNAR1 protein levels. IFNAR1(-/-) mice were protected from the effects of hypoxia on the right heart, vascular remodeling, and raised serum endothelin 1 levels. CONCLUSIONS: These data indicate that type I IFN, via an action of IFNAR1, mediates PAH.


Asunto(s)
Hipertensión Pulmonar/inmunología , Interferón-alfa/inmunología , Interferón beta/inmunología , Receptor de Interferón alfa y beta/inmunología , Esclerodermia Sistémica/inmunología , Animales , Células Cultivadas , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Células Endoteliales/inmunología , Endotelina-1/inmunología , Endotelina-1/metabolismo , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/metabolismo , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Interferón beta/metabolismo , Interferón beta/farmacología , Interferón gamma/inmunología , Interferón gamma/farmacología , Pulmón/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Esclerodermia Sistémica/metabolismo , Transducción de Señal/inmunología
16.
Br J Clin Pharmacol ; 81(4): 621-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26561399

RESUMEN

AIMS: In vivo platelet function is a product of intrinsic platelet reactivity, modifiable by dual antiplatelet therapy (DAPT), and the extrinsic inhibitory endothelial mediators, nitric oxide (NO) and prostacyclin (PGI2 ), that are powerfully potentiated by P2Y12 receptor blockade. This implies that for individual patients endothelial mediator production is an important determinant of DAPT effectiveness. Here, we have investigated this idea using platelets taken from healthy volunteers treated with anti-platelet drugs. METHODS: Three groups of male volunteers (n = 8) received either prasugrel (10 mg), aspirin (75 mg) or DAPT (prasugrel + aspirin) once daily for 7 days. Platelet reactivity in the presence of diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA/NONOate) and PGI2 was studied before and following treatment. RESULTS: Ex vivo, PGI2 and/or DEA/NONOate had little inhibitory effect on TRAP-6-induced platelet reactivity in control conditions. However, in the presence of DAPT, combination of DEA/NONOate + PGI2 reduced platelet aggregation (74 ± 3% to 19 ± 6%, P < 0.05). In vitro studies showed even partial (25%) P2Y12 receptor blockade produced a significant (67 ± 2% to 39 ± 10%, P < 0.05) inhibition when DEA/NONOate + PGI2 was present. CONCLUSIONS: We have demonstrated that PGI2 and NO synergize with P2Y12 receptor antagonists to produce powerful platelet inhibition. Furthermore, even with submaximal P2Y12 blockade the presence of PGI2 and NO greatly enhances platelet inhibition. Our findings highlight the importance of endothelial mediator in vivo modulation of P2Y12 inhibition and introduces the concept of refining ex vivo platelet function testing by incorporating an assessment of endothelial function to predict thrombotic outcomes better and adjust therapy to prevent adverse outcomes in individual patients.


Asunto(s)
Aspirina/farmacología , Epoprostenol/farmacología , Óxido Nítrico/farmacología , Activación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Adolescente , Adulto , Aspirina/administración & dosificación , Plaquetas/efectos de los fármacos , Sinergismo Farmacológico , Epoprostenol/administración & dosificación , Epoprostenol/metabolismo , Voluntarios Sanos , Humanos , Técnicas In Vitro , Masculino , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel/administración & dosificación , Antagonistas del Receptor Purinérgico P2Y/administración & dosificación , Adulto Joven
17.
Arterioscler Thromb Vasc Biol ; 35(10): 2122-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26272940

RESUMEN

OBJECTIVE: Reduced antiplatelet drug efficacy occurs in conditions of increased platelet turnover, associated with increased proportions of drug-free, that is, uninhibited, platelets. Here, we detail mechanisms by which drug-free platelets promote platelet aggregation in the face of standard antiplatelet therapy. APPROACH AND RESULTS: To model standard antiplatelet therapy, platelets were treated in vitro with aspirin, the P2Y12 receptor blocker prasugrel active metabolite, or aspirin plus prasugrel active metabolite. Different proportions of uninhibited platelets were then introduced. Light transmission aggregometry analysis demonstrated clear positive associations between proportions of drug-free platelets and percentage platelet aggregation in response to a range of platelet agonists. Using differential platelet labeling coupled with advanced flow cytometry and confocal imaging we found aggregates formed in mixtures of aspirin-inhibited platelets together with drug-free platelets were characterized by intermingled platelet populations. This distribution is in accordance with the ability of drug-free platelets to generate thromboxane A2 and so drive secondary platelet activation. Conversely, aggregates formed in mixtures of prasugrel active metabolite-inhibited or aspirin plus prasugrel active metabolite-inhibited platelets together with drug-free platelets were characterized by distinct cores of drug-free platelets. This distribution is consistent with the ability of drug-free platelets to respond to the secondary activator ADP. CONCLUSIONS: These experiments are the first to image the interactions of inhibited and uninhibited platelets in the formation of platelet aggregates. They demonstrate that a general population of platelets can contain subpopulations that respond strikingly differently to overall stimulation of the population and so act as the seed for platelet aggregation.


Asunto(s)
Aspirina/farmacología , Plaquetas/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel/farmacología , Quimioterapia Combinada , Citometría de Flujo , Humanos , Técnicas In Vitro , Activación Plaquetaria/efectos de los fármacos , Sensibilidad y Especificidad , Tromboxanos/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(39): 15782-7, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-24003163

RESUMEN

Circulating platelets are constantly exposed to nitric oxide (NO) released from the vascular endothelium. This NO acts to reduce platelet reactivity, and in so doing blunts platelet aggregation and thrombus formation. For successful hemostasis, platelet activation and aggregation must occur at sites of vascular injury despite the constant presence of NO. As platelets aggregate, they release secondary mediators that drive further aggregation. Particularly significant among these secondary mediators is ADP, which, acting through platelet P2Y12 receptors, strongly amplifies aggregation. Platelet P2Y12 receptors are the targets of very widely used antithrombotic drugs such as clopidogrel, prasugrel, and ticagrelor. Here we show that blockade of platelet P2Y12 receptors dramatically enhances the antiplatelet potency of NO, causing a 1,000- to 100,000-fold increase in inhibitory activity against platelet aggregation and release reactions in response to activation of receptors for either thrombin or collagen. This powerful synergism is explained by blockade of a P2Y12 receptor-dependent, NO/cGMP-insensitive phosphatidylinositol 3-kinase pathway of platelet activation. These studies demonstrate that activation of the platelet ADP receptor, P2Y12, severely blunts the inhibitory effects of NO. The powerful antithrombotic effects of P2Y12 receptor blockers may, in part, be mediated by profound potentiation of the effects of endogenous NO.


Asunto(s)
Óxido Nítrico/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/enzimología , Proteína C-Reactiva/farmacología , Calcio/metabolismo , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Epoprostenol/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Trombina/farmacología , Proteínas de Unión al GTP rap1/metabolismo
19.
Proc Natl Acad Sci U S A ; 110(39): 15812-7, 2013 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-24019484

RESUMEN

Homocysteine is metabolized to methionine by the action of 5,10 methylenetetrahydrofolate reductase (MTHFR). Alternatively, by the transulfuration pathway, homocysteine is transformed to hydrogen sulphide (H2S), through multiple steps involving cystathionine ß-synthase and cystathionine γ-lyase. Here we have evaluated the involvement of H2S in the thrombotic events associated with hyperhomocysteinemia. To this purpose we have used platelets harvested from healthy volunteers or patients newly diagnosed with hyperhomocysteinemia with a C677T polymorphism of the MTHFR gene (MTHFR++). NaHS (0.1-100 µM) or l-cysteine (0.1-100 µM) significantly increased platelet aggregation harvested from healthy volunteers induced by thrombin receptor activator peptide-6 amide (2 µM) in a concentration-dependent manner. This increase was significantly potentiated in platelets harvested from MTHFR++ carriers, and it was reversed by the inhibition of either cystathionine ß-synthase or cystathionine γ-lyase. Similarly, in MTHFR++ carriers, the content of H2S was significantly higher in either platelets or plasma compared with healthy volunteers. Interestingly, thromboxane A2 production was markedly increased in response to both NaHS or l-cysteine in platelets of healthy volunteers. The inhibition of phospholipase A2, cyclooxygenase, or blockade of the thromboxane receptor markedly reduced the effects of H2S. Finally, phosphorylated-phospholipase A2 expression was significantly higher in MTHFR++ carriers compared with healthy volunteers. In conclusion, the H2S pathway is involved in the prothrombotic events occurring in hyperhomocysteinemic patients.


Asunto(s)
Sulfuro de Hidrógeno/farmacología , Hiperhomocisteinemia/sangre , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Ácido Araquidónico/metabolismo , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Fosfolipasas A2 Grupo IV/metabolismo , Heterocigoto , Humanos , Hiperhomocisteinemia/orina , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Receptores de Trombina/metabolismo , Tromboxano B2/análogos & derivados , Tromboxano B2/orina
20.
Circ Res ; 112(4): 595-600, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23283721

RESUMEN

RATIONALE: MicroRNA (miRNA) biomarkers are attracting considerable interest. Effects of medication, however, have not been investigated thus far. OBJECTIVE: To analyze changes in plasma miRNAs in response to antiplatelet therapy. METHODS AND RESULTS: Profiling for 377 miRNAs was performed in platelets, platelet microparticles, platelet-rich plasma, platelet-poor plasma, and serum. Platelet-rich plasma showed markedly higher levels of miRNAs than serum and platelet-poor plasma. Few abundant platelet miRNAs, such as miR-24, miR-197, miR-191, and miR-223, were also increased in serum compared with platelet-poor plasma. In contrast, antiplatelet therapy significantly reduced miRNA levels. Using custom-made quantitative real-time polymerase chain reaction plates, 92 miRNAs were assessed in a dose-escalation study in healthy volunteers at 4 different time points: at baseline without therapy, at 1 week with 10 mg prasugrel, at 2 weeks with 10 mg prasugrel plus 75 mg aspirin, and at 3 weeks with 10 mg prasugrel plus 300 mg aspirin. Findings in healthy volunteers were confirmed by individual TaqMan quantitative real-time polymerase chain reaction assays (n=9). Validation was performed in an independent cohort of patients with symptomatic atherosclerosis (n=33), who received low-dose aspirin at baseline. Plasma levels of platelet miRNAs, such as miR-223, miR-191, and others, that is, miR-126 and miR-150, decreased on further platelet inhibition. CONCLUSIONS: Our study demonstrated a substantial platelet contribution to the circulating miRNA pool and identified miRNAs responsive to antiplatelet therapy. It also highlights that antiplatelet therapy and preparation of blood samples could be confounding factors in case-control studies relating plasma miRNAs to cardiovascular disease.


Asunto(s)
Plaquetas/metabolismo , MicroARNs/sangre , Plasma/metabolismo , Activación Plaquetaria , Plasma Rico en Plaquetas/metabolismo , Suero/metabolismo , Adulto , Aspirina/administración & dosificación , Aspirina/farmacología , Aspirina/uso terapéutico , Biomarcadores , Plaquetas/efectos de los fármacos , Recolección de Muestras de Sangre/métodos , Enfermedades de las Arterias Carótidas/tratamiento farmacológico , Estudios de Casos y Controles , Ensayos Clínicos como Asunto , Factores de Confusión Epidemiológicos , Diabetes Mellitus Tipo 2/sangre , Relación Dosis-Respuesta a Droga , Monitoreo de Drogas , Sinergismo Farmacológico , Quimioterapia Combinada , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/biosíntesis , MicroARNs/genética , Piperazinas/administración & dosificación , Piperazinas/farmacología , Activación Plaquetaria/genética , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Clorhidrato de Prasugrel , Reacción en Cadena en Tiempo Real de la Polimerasa , Tiofenos/administración & dosificación , Tiofenos/farmacología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA