Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 132(2): 223-237, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36656971

RESUMEN

Driven by autonomous molecular clocks that are synchronized by a master pacemaker in the suprachiasmatic nucleus, cardiac physiology fluctuates in diurnal rhythms that can be partly or entirely circadian. Cardiac contractility, metabolism, and electrophysiology, all have diurnal rhythms, as does the neurohumoral control of cardiac and kidney function. In this review, we discuss the evidence that circadian biology regulates cardiac function, how molecular clocks may relate to the pathogenesis of heart failure, and how chronotherapeutics might be applied in heart failure. Disrupting molecular clocks can lead to heart failure in animal models, and the myocardial response to injury seems to be conditioned by the time of day. Human studies are consistent with these findings, and they implicate the clock and circadian rhythms in the pathogenesis of heart failure. Certain circadian rhythms are maintained in patients with heart failure, a factor that can guide optimal timing of therapy. Pharmacologic and nonpharmacologic manipulation of circadian rhythms and molecular clocks show promise in the prevention and treatment of heart failure.


Asunto(s)
Relojes Circadianos , Insuficiencia Cardíaca , Animales , Humanos , Ritmo Circadiano , Corazón , Insuficiencia Cardíaca/terapia , Biología , Relojes Circadianos/fisiología
2.
Arterioscler Thromb Vasc Biol ; 44(6): 1393-1406, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38660804

RESUMEN

BACKGROUND: Low-dose aspirin is widely used for the secondary prevention of cardiovascular disease. The beneficial effects of low-dose aspirin are attributable to its inhibition of platelet Cox (cyclooxygenase)-1-derived thromboxane A2. Until recently, the use of the Pf4 (platelet factor 4) Cre has been the only genetic approach to generating megakaryocyte/platelet ablation of Cox-1 in mice. However, Pf4-ΔCre displays ectopic expression outside the megakaryocyte/platelet lineage, especially during inflammation. The use of the Gp1ba (glycoprotein 1bα) Cre promises a more specific, targeted approach. METHODS: To evaluate the role of Cox-1 in platelets, we crossed Pf4-ΔCre or Gp1ba-ΔCre mice with Cox-1flox/flox mice to generate platelet Cox-1-/- mice on normolipidemic and hyperlipidemic (Ldlr-/-; low-density lipoprotein receptor) backgrounds. RESULTS: Ex vivo platelet aggregation induced by arachidonic acid or adenosine diphosphate in platelet-rich plasma was inhibited to a similar extent in Pf4-ΔCre Cox-1-/-/Ldlr-/- and Gp1ba-ΔCre Cox-1-/-/Ldlr-/- mice. In a mouse model of tail injury, Pf4-ΔCre-mediated and Gp1ba-ΔCre-mediated deletions of Cox-1 were similarly efficient in suppressing platelet prostanoid biosynthesis. Experimental thrombogenesis and attendant blood loss were similar in both models. However, the impact on atherogenesis was divergent, being accelerated in the Pf4-ΔCre mice while restrained in the Gp1ba-ΔCres. In the former, accelerated atherogenesis was associated with greater suppression of PGI2 biosynthesis, a reduction in the lipopolysaccharide-evoked capacity to produce PGE2 (prostaglandin E) and PGD2 (prostanglandin D), activation of the inflammasome, elevated plasma levels of IL-1ß (interleukin), reduced plasma levels of HDL-C (high-density lipoprotein receptor-cholesterol), and a reduction in the capacity for reverse cholesterol transport. By contrast, in the latter, plasma HDL-C and α-tocopherol were elevated, and MIP-1α (macrophage inflammatory protein-1α) and MCP-1 (monocyte chemoattractant protein 1) were reduced. CONCLUSIONS: Both approaches to Cox-1 deletion similarly restrain thrombogenesis, but a differential impact on Cox-1-dependent prostanoid formation by the vasculature may contribute to an inflammatory phenotype and accelerated atherogenesis in Pf4-ΔCre mice.


Asunto(s)
Plaquetas , Ciclooxigenasa 1 , Modelos Animales de Enfermedad , Integrasas , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Plaquetaria , Factor Plaquetario 4 , Receptores de LDL , Animales , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/enzimología , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 1/genética , Ciclooxigenasa 1/deficiencia , Agregación Plaquetaria/efectos de los fármacos , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Integrasas/genética , Receptores de LDL/genética , Receptores de LDL/deficiencia , Masculino , Ratones , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Aterosclerosis/sangre , Hiperlipidemias/sangre , Hiperlipidemias/genética , Hiperlipidemias/enzimología , Fenotipo , Proteínas de la Membrana , Complejo GPIb-IX de Glicoproteína Plaquetaria
3.
J Pharmacol Exp Ther ; 367(3): 425-432, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30305427

RESUMEN

Prostaglandin (PG) D2 is formed by two distinct PGD synthases (PGDS): lipocalin-type PGDS (L-PGDS), which acts as a PGD2-producing enzyme and as extracellular lipophilic transporter, and hematopoietic PGDS (H-PGDS), a σ glutathione-S-transferase. PGD2 plays an important role in the maintenance of vascular function; however, the relative contribution of L-PGDS- and H-PGDS-dependent formation of PGD2 in this setting is unknown. To gain insight into the function played by these distinct PGDS, we assessed systemic blood pressure (BP) and thrombogenesis in L-Pgds and H-Pgds knockout (KO) mice. Deletion of L-Pgds depresses urinary PGD2 metabolite (PGDM) by ∼35%, whereas deletion of H-Pgds does so by ∼90%. Deletion of L-Pgds, but not H-Pgds, elevates BP and accelerates the thrombogenic occlusive response to a photochemical injury to the carotid artery. HQL-79, a H-PGDS inhibitor, further depresses PGDM in L-Pgds KO mice, but has no effect on BP or on the thrombogenic response. Gene expression profiling reveals that pathways relevant to vascular function are dysregulated in the aorta of L-Pgds KOs. These results indicate that the functional impact of L-Pgds deletion on vascular homeostasis may result from an autocrine effect of L-PGDS-dependent PGD2 on the vasculature and/or the L-PGDS function as lipophilic carrier protein.


Asunto(s)
Hipertensión/genética , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Prostaglandina D2/genética , Eliminación de Secuencia/genética , Animales , Arterias Carótidas/patología , Glutatión Transferasa/genética , Masculino , Ratones , Ratones Noqueados
4.
Annu Rev Pharmacol Toxicol ; 54: 205-26, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24392694

RESUMEN

There is a need for improved treatment of acute vascular inflammation in conditions such as ischemia-reperfusion injury, acute lung injury, sepsis, and stroke. The vascular endothelium represents an important therapeutic target in these conditions. Furthermore, some anti-inflammatory agents (AIAs) (e.g., biotherapeutics) require precise delivery into subcellular compartments. In theory, optimized delivery to the desired site of action may improve the effects and enable new mechanisms of action of these AIAs. Diverse nanocarriers (NCs) and strategies for targeting them to endothelial cells have been designed and explored for this purpose. Studies in animal models suggest that delivery of AIAs using NCs may provide potent and specific molecular interventions in inflammatory pathways. However, the industrial development and clinical translation of complex NC-AIA formulations are challenging. Rigorous analysis of therapeutic/side effect and benefit/cost ratios is necessary to identify and optimize the approaches that may find clinical utility in the management of acute inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Sistemas de Liberación de Medicamentos/métodos , Nanoestructuras/química , Animales , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Enfermedades Vasculares/tratamiento farmacológico
5.
Blood ; 125(22): 3365-6, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26022048

RESUMEN

In this issue of Blood, Xiang et al identify a novel mechanism, involving activation of the polyol pathway and repression of microRNA-24 (miR-24), through which hyperglycemia augments von Willebrand factor (VWF) expression and secretion.


Asunto(s)
Células Endoteliales/metabolismo , Hiperglucemia/genética , MicroARNs/genética , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Animales , Femenino , Humanos , Masculino
6.
Platelets ; 28(5): 457-462, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28102731

RESUMEN

Platelets present a number of intracellular and transmembrane targets subject to pharmacological modulation, either for cardiovascular disease reduction or as an unintended drug response. Microfluidic devices allow human blood to clot on a defined surface under controlled hemodynamic and pharmacological conditions. The potencies of a number of antiplatelet and anticancer drugs have been tested with respect to platelet deposition on collagen under flow. Inhibitors of cyclooxygenase-1 (COX-1) reduce platelet deposition, either when added ex vivo to blood or ingested orally by patients prior to testing. Some individuals display a functional "aspirin-insensitivity" in microfluidic assay. When certain nonsteroidal anti-inflammatory drugs (NSAIDs) are taken orally, they block COX-1 acetylation by aspirin with concomitant reduction of aspirin efficacy against platelets in microfluidic assay. Both P2Y1 and P2Y12 inhibitors reduce platelet deposition under flow, as do NO donors and iloprost that target the guanylate cyclase and the prostacyclin receptor, respectively. In a microfluidic assay of 37 kinase inhibitors, dasatinib had potent antiplatelet activity, while bosutinib was less potent. Dasatinib and bosutinib have known profiles against numerous kinases, revealing overlapping and nonoverlapping activities relevant to their unique actions against platelets. Also, dasatinib caused a marked and specific inhibition of GPVI signaling induced by convulxin, consistent with a dasatinib-associated bleeding risk. Microfluidic devices facilitate drug library screening, dose-response testing, and drug-drug interaction studies. Kinase inhibitors developed as anticancer agents may present antiplatelet activities that are detectable by microfluidic assay and potentially linked to bleeding risks.


Asunto(s)
Plaquetas/metabolismo , Evaluación Preclínica de Medicamentos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Preparaciones Farmacéuticas , Animales , Evaluación Preclínica de Medicamentos/instrumentación , Evaluación Preclínica de Medicamentos/métodos , Humanos
7.
Proc Natl Acad Sci U S A ; 111(47): 16830-5, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385584

RESUMEN

The cardiovascular safety of nonsteroidal antiinflammatory drugs (NSAIDs) may be influenced by interactions with antiplatelet doses of aspirin. We sought to quantitate precisely the propensity of commonly consumed NSAIDs­ibuprofen, naproxen, and celecoxib­to cause a drug-drug interaction with aspirin in vivo by measuring the target engagement of aspirin directly by MS. We developed a novel assay of cyclooxygenase-1 (COX-1) acetylation in platelets isolated from volunteers who were administered aspirin and used conventional and microfluidic assays to evaluate platelet function. Although ibuprofen, naproxen, and celecoxib all had the potential to compete with the access of aspirin to the substrate binding channel of COX-1 in vitro, exposure of volunteers to a single therapeutic dose of each NSAID followed by 325 mg aspirin revealed a potent drug-drug interaction between ibuprofen and aspirin and between naproxen and aspirin but not between celecoxib and aspirin. The imprecision of estimates of aspirin consumption and the differential impact on the ability of aspirin to inactivate platelet COX-1 will confound head-to-head comparisons of distinct NSAIDs in ongoing clinical studies designed to measure their cardiovascular risk.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Plaquetas/efectos de los fármacos , Ciclooxigenasa 1/metabolismo , Acetilación , Plaquetas/enzimología , Humanos , Microfluídica
10.
Carcinogenesis ; 35(8): 1788-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590894

RESUMEN

Cyclooxygenase-2 (COX-2) expression is associated with poor prognosis across a range of human cancers, including breast cancer. The contribution of tumor cell-derived COX-2 to tumorigenesis has been examined in numerous studies; however, the role of stromal-derived COX-2 is ill-defined. Here, we examined how COX-2 in myeloid cells, an immune cell subset that includes macrophages, influences mammary tumor progression. In mice engineered to selectively lack myeloid cell COX-2 [myeloid-COX-2 knockout (KO) mice], spontaneous neu oncogene-induced tumor onset was delayed, tumor burden reduced, and tumor growth slowed compared with wild-type (WT). Similarly, growth of neu-transformed mammary tumor cells as orthotopic tumors in immune competent syngeneic myeloid-COX-2 KO host mice was reduced compared with WT. By flow cytometric analysis, orthotopic myeloid-COX-2 KO tumors had lower tumor-associated macrophage (TAM) infiltration consistent with impaired colony stimulating factor-1-dependent chemotaxis by COX-2 deficient macrophages in vitro. Further, in both spontaneous and orthotopic tumors, COX-2-deficient TAM displayed lower immunosuppressive M2 markers and this was coincident with less suppression of CD8(+) cytotoxic T lymphocytes (CTLs) in myeloid-COX-2 KO tumors. These studies suggest that reduced tumor growth in myeloid-COX-2 KO mice resulted from disruption of M2-like TAM function, thereby enhancing T-cell survival and immune surveillance. Antibody-mediated depletion of CD8(+), but not CD4(+) cells, restored tumor growth in myeloid-COX-2 KO to WT levels, indicating that CD8(+) CTLs are dominant antitumor effectors in myeloid-COX-2 KO mice. Our studies suggest that inhibition of myeloid cell COX-2 can potentiate CTL-mediated tumor cytotoxicity and may provide a novel therapeutic approach in breast cancer therapy.


Asunto(s)
Ciclooxigenasa 2/fisiología , Macrófagos/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/prevención & control , Células Mieloides/inmunología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología , Animales , Western Blotting , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Activación de Linfocitos , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Células Tumorales Cultivadas
11.
Circulation ; 127(3): 377-85, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23212718

RESUMEN

BACKGROUND: Low dose aspirin reduces the secondary incidence of myocardial infarction and stroke. Drug resistance to aspirin might result in treatment failure. Despite this concern, no clear definition of aspirin resistance has emerged, and estimates of its incidence have varied remarkably. We aimed to determine the commonality of a mechanistically consistent, stable, and specific phenotype of true pharmacological resistance to aspirin-such as might be explained by genetic causes. METHODS AND RESULTS: Healthy volunteers (n=400) were screened for their response to a single oral dose of 325-mg immediate release or enteric coated aspirin. Response parameters reflected the activity of the molecular target of aspirin, cyclooxygenase-1. Individuals who appeared aspirin resistant on 1 occasion underwent repeat testing, and if still resistant were exposed to low-dose enteric coated aspirin (81 mg) and clopidogrel (75 mg) for 1 week each. Variable absorption caused a high frequency of apparent resistance to a single dose of 325-mg enteric coated aspirin (up to 49%) but not to immediate release aspirin (0%). All individuals responded to aspirin on repeated exposure, extension of the postdosing interval, or addition of aspirin to their platelets ex vivo. CONCLUSIONS: Pharmacological resistance to aspirin is rare; this study failed to identify a single case of true drug resistance. Pseudoresistance, reflecting delayed and reduced drug absorption, complicates enteric coated but not immediate release aspirin administration. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00948987.


Asunto(s)
Aspirina/farmacología , Ciclooxigenasa 1/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Resistencia a Medicamentos , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Administración Oral , Adolescente , Adulto , Aspirina/administración & dosificación , Aspirina/uso terapéutico , Clopidogrel , Estudios Cruzados , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Infarto del Miocardio/prevención & control , Inhibidores de Agregación Plaquetaria/administración & dosificación , Inhibidores de Agregación Plaquetaria/uso terapéutico , Accidente Cerebrovascular/prevención & control , Comprimidos Recubiertos , Ticlopidina/administración & dosificación , Ticlopidina/análogos & derivados , Ticlopidina/farmacología , Resultado del Tratamiento , Adulto Joven
13.
medRxiv ; 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38854091

RESUMEN

Background: Non-steroidal anti-inflammatory drugs (NSAIDs) increase the risk of adverse cardiovascular events via suppression of cyclooxygenase (COX)-2-derived prostacyclin (PGI2) formation in heart, vasculature, and kidney. The Prospective Randomized Evaluation of Celecoxib Integrated Safety versus Ibuprofen Or Naproxen (PRECISION) trial and other large clinical studies compared the cardiovascular risk of traditional NSAIDs (i.e. naproxen), which inhibit both COX isozymes, with NSAIDs selective for COX-2 (i.e. celecoxib). However, whether pharmacologically equipotent doses were used - that is, whether a similar degree of COX-2 inhibition was achieved - was not considered. We compared drug target inhibition and blood pressure response to celecoxib at the dose used by most patients in PRECISION with the lowest recommended naproxen dose for osteoarthritis, which is lower than the dose used in PRECISION. Methods: Sixteen healthy participants (19-61 years) were treated with celecoxib (100 mg every 12h), naproxen (250 mg every 12h), or placebo administered twice daily for seven days in a double-blind, crossover design randomized by order. On Day 7 when drug levels had reached steady state, the degree of COX inhibition was assessed ex vivo and in vivo. Ambulatory blood pressure was measured throughout the final 12h dosing interval. Results: Both NSAIDs inhibited COX-2 activity relative to placebo, but naproxen inhibited COX-2 activity to a greater degree (62.9±21.7%) than celecoxib (35.7±25.2%; p<0.05). Similarly, naproxen treatment inhibited PGI2 formation in vivo (48.0±24.9%) to a greater degree than celecoxib (26.7±24.6%; p<0.05). Naproxen significantly increased blood pressure compared to celecoxib (differences in least-square means of mean arterial pressure: 2.5 mm Hg (95% CI: 1.5, 3.5); systolic blood pressure: 4.0 mm Hg (95% CI: 2.9, 5.1); diastolic blood pressure: 1.8 mm Hg (95% CI: 0.8, 2.8); p<0.05 for all). The difference in systolic blood pressure relative to placebo was associated with the degree of COX-2 inhibition (p<0.05). Conclusions: Celecoxib 200 mg/day inhibited COX-2 activity to a lesser degree than naproxen 500 mg/day, resulting in a less pronounced blood pressure increase. While the PRECISION trial concluded the non-inferiority of celecoxib regarding cardiovascular risk, this is based on a comparison of doses that are not equipotent.ClinicalTrials.gov identifier: NCT02502006 (https://clinicaltrials.gov/study/NCT02502006).

14.
Clin Chem ; 59(8): 1195-204, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23592503

RESUMEN

BACKGROUND: Microfluidic devices can create hemodynamic conditions for platelet assays. We validated an 8-channel device in a study of interdonor response to acetylsalicylic acid (ASA, aspirin) with whole blood from 28 healthy individuals. METHODS: Platelet deposition was assessed before treatment or 24 h after ingestion of 325 mg ASA. Whole blood (plus 100 µmol/L H-d-Phe-Pro-Arg-chloromethylketone to inhibit thrombin) was further treated ex vivo with ASA (0-500 µmol/L) and perfused over fibrillar collagen for 300 s at a venous wall shear rate (200 s(-1)). RESULTS: Ex vivo ASA addition to blood drawn before aspirin ingestion caused a reduction in platelet deposition [half-maximal inhibitory concentration (IC50) approximately 10-20 µmol/L], especially between 150 and 300 s of perfusion, when secondary aggregation mediated by thromboxane was expected. Twenty-seven of 28 individuals displayed smaller deposits (45% mean reduction; range 10%-90%; P < 0.001) from blood obtained 24 h after ASA ingestion (no ASA added ex vivo). In replicate tests, an R value to score secondary aggregation [deposition rate from 150 to 300 s normalized by rate from 60 to 150 s] showed R < 1 in only 2 of 28 individuals without ASA ingestion, with R > 1 in only 3 of 28 individuals after 500 µmol/L ASA addition ex vivo. At 24 h after ASA ingestion, 21 of 28 individuals displayed poor secondary aggregation (R < 1) without ex vivo ASA addition, whereas the 7 individuals with residual secondary aggregation (R > 1) displayed insensitivity to ex vivo ASA addition. Platelet deposition was not correlated with platelet count. Ex vivo ASA addition caused similar inhibition at venous and arterial wall shear rates. CONCLUSIONS: Microfluidic devices quantified platelet deposition after ingestion or ex vivo addition of aspirin.


Asunto(s)
Aspirina/farmacología , Plaquetas/efectos de los fármacos , Colágeno , Técnicas Analíticas Microfluídicas/instrumentación , Inhibidores de Agregación Plaquetaria/farmacología , Administración Oral , Adolescente , Adulto , Aspirina/uso terapéutico , Plaquetas/fisiología , Ciclooxigenasa 1/metabolismo , Femenino , Colorantes Fluorescentes , Humanos , Masculino , Persona de Mediana Edad , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/uso terapéutico , Pruebas de Función Plaquetaria , Valores de Referencia , Flujo Sanguíneo Regional , Transducción de Señal , Estrés Mecánico , Tromboxano A2/metabolismo , Adulto Joven
15.
Blood ; 117(1): e15-26, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20962327

RESUMEN

Activated platelets shed surface proteins, potentially modifying platelet function as well as providing a source of bioactive fragments. Previous studies have identified several constituents of the platelet sheddome, but the full extent of shedding is unknown. Here we have taken a global approach, analyzing protein fragments in the supernate of activated platelets using mass spectroscopy and looking for proteins originating from platelet membranes. After removing plasma proteins and microparticles, 1048 proteins were identified, including 69 membrane proteins. Nearly all of the membrane proteins had been detected previously, but only 10 had been shown to be shed in platelets. The remaining 59 are candidates subject to confirmation. Based on spectral counts, protein representation in the sheddome varies considerably. As proof of principle, we validated one of the less frequently detected proteins, semaphorin 7A, which had not previously been identified in platelets. Surface expression, cleavage, and shedding of semaphorin 7A were demonstrated, as was its association with α-granules. Finally, cleavage of semaphorin 7A and 12 other proteins was substantially reduced by an inhibitor of ADAM17, a known sheddase. These results define a subset of membrane proteins as sheddome candidates, forming the basis for further studies examining the impact of ectodomain shedding on platelet function.


Asunto(s)
Proteínas ADAM/metabolismo , Plaquetas/fisiología , Proteínas de la Membrana/metabolismo , Activación Plaquetaria/fisiología , Semaforinas/antagonistas & inhibidores , Proteína ADAM17 , Adulto , Western Blotting , Gránulos Citoplasmáticos/química , Gránulos Citoplasmáticos/metabolismo , Citometría de Flujo , Humanos , Quinolinas/farmacología , Semaforinas/metabolismo , Espectrometría de Masas en Tándem
16.
Prostaglandins Other Lipid Mediat ; 104-105: 58-66, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22981510

RESUMEN

Prostanoids play pivotal roles in inflammation and pain. Cyclooxygenase (COX) inhibitors, the nonsteroidal anti-inflammatory drugs (NSAIDs), depress prostanoid formation and are widely used to treat inflammatory pain. However, their therapeutic benefit is offset by serious side-effects, primarily gastrointestinal and cardiovascular complications. Pathway elements downstream of the COX enzymes, particularly the terminal synthases and receptors of prostaglandin E2, have been proposed as alternative targets for the development of novel NSAID like drugs. Here, we summarize the current knowledge on the roles of individual prostanoids in modulating inflammatory pain.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Dolor/metabolismo , Prostaglandinas/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Inhibidores de la Ciclooxigenasa/farmacología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/fisiopatología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Prostaglandina-E Sintasas , Prostaglandina-Endoperóxido Sintasas/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo
17.
Nature ; 447(7147): 1007-11, 2007 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-17581586

RESUMEN

Haematopoietic stem cell (HSC) homeostasis is tightly controlled by growth factors, signalling molecules and transcription factors. Definitive HSCs derived during embryogenesis in the aorta-gonad-mesonephros region subsequently colonize fetal and adult haematopoietic organs. To identify new modulators of HSC formation and homeostasis, a panel of biologically active compounds was screened for effects on stem cell induction in the zebrafish aorta-gonad-mesonephros region. Here, we show that chemicals that enhance prostaglandin (PG) E2 synthesis increased HSC numbers, and those that block prostaglandin synthesis decreased stem cell numbers. The cyclooxygenases responsible for PGE2 synthesis were required for HSC formation. A stable derivative of PGE2 improved kidney marrow recovery following irradiation injury in the adult zebrafish. In murine embryonic stem cell differentiation assays, PGE2 caused amplification of multipotent progenitors. Furthermore, ex vivo exposure to stabilized PGE2 enhanced spleen colony forming units at day 12 post transplant and increased the frequency of long-term repopulating HSCs present in murine bone marrow after limiting dilution competitive transplantation. The conserved role for PGE2 in the regulation of vertebrate HSC homeostasis indicates that modulation of the prostaglandin pathway may facilitate expansion of HSC number for therapeutic purposes.


Asunto(s)
Dinoprostona/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Homeostasis/efectos de los fármacos , Vertebrados , Animales , Diferenciación Celular/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Dinoprostona/agonistas , Dinoprostona/antagonistas & inhibidores , Dinoprostona/biosíntesis , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Proteínas Proto-Oncogénicas c-myb/genética , Vertebrados/embriología , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
18.
Annu Rev Med ; 61: 17-33, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20059330

RESUMEN

Nonsteroidal antinflammatory drugs (NSAIDs) inhibit prostaglandin formation by cyclooxygenases (COX) 1 and 2. NSAIDs selective for inhibition of COX-2 are less likely than traditional drugs to cause serious gastrointestinal adverse effects, but predispose to adverse cardiovascular events, such as heart failure, myocardial infarction, and stroke. Evidence from human pharmacology and genetics, genetically manipulated rodents, and other animal models and randomized trials indicates that this is consequent to suppression of COX-2-dependent cardioprotective prostagladins, particularly prostacyclin. Lessons drawn from how this saga unfolded are relevant to how we approach drug surveillance and regulation, integrate diversifed forms of information and might pursue a more personalized approach to drug efficacy and risk.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Ciclooxigenasa 1/fisiología , Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/fisiología , Animales , Inhibidores de la Ciclooxigenasa 2/efectos adversos , Humanos , Ratones , Vigilancia de Productos Comercializados , Medición de Riesgo
19.
Hypertension ; 79(9): 1922-1926, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35862146

RESUMEN

Acetaminophen is widely regarded as a safe therapy for pain and fever in patients with cardiovascular disease and those taking anticoagulants. However, recent studies report that acetaminophen, like most other nonsteroidal anti-inflammatory drugs, increases blood pressure, and a formulation containing sodium increases cardiovascular risk. Those findings call into question guidelines recommending acetaminophen for patients with cardiovascular disease and pain, and those taking anticoagulants. We review evidence that acetaminophen has effects in common with nonsteroidal anti-inflammatory drugs, and its influence on coagulation via effects on vitamin K metabolism. Possible alternatives to acetaminophen for patients with pain are discussed.


Asunto(s)
Analgésicos no Narcóticos , Enfermedades Cardiovasculares , Hipertensión , Acetaminofén/efectos adversos , Analgésicos no Narcóticos/efectos adversos , Antiinflamatorios no Esteroideos/efectos adversos , Anticoagulantes/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Humanos , Hipertensión/tratamiento farmacológico , Dolor/tratamiento farmacológico
20.
J Biol Rhythms ; 37(1): 43-52, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34724846

RESUMEN

Circadian omics analyses present investigators with large amounts of data to consider and many choices for methods of analysis. Visualization is crucial as rhythmicity can take many forms and p-values offer an incomplete picture. Yet statically viewing the entirety of high-throughput datasets is impractical, and there is often limited ability to assess the impact of choices, such as significance threshold cutoffs. Nitecap provides an intuitive and unified web-based solution to these problems. Through highly responsive visualizations, Nitecap enables investigators to see dataset-wide behavior. It supports deep analyses, including comparisons of two conditions. Moreover, it focuses upon ease-of-use and enables collaboration through dataset sharing. As an application, we investigated cross talk between peripheral clocks in adipose and liver tissues and determined that adipocyte clock disruption does not substantially modulate the transcriptional rhythmicity of liver but does advance the phase of core clock gene Bmal1 (Arntl) expression in the liver. Nitecap is available at nitecap.org and is free-to-use.


Asunto(s)
Factores de Transcripción ARNTL , Relojes Circadianos , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Proteínas CLOCK/genética , Relojes Circadianos/genética , Ritmo Circadiano/genética , Hígado/metabolismo , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA