Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cardiovasc Drugs Ther ; 37(5): 989-996, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-35920946

RESUMO

Results from DELIVER trial and publication of EMPEROR-Preserved with sodium-glucose cotransporter 2 (SGLT2) inhibitors in patients with heart failure (HF) with ejection fraction (EF) > 40% represent a significant step forward in the treatment of HF with preserved EF (HFpEF). However, detailed analysis and attenuation of effect at higher EF levels have sparked some doubts about whether empagliflozin is effective across the entire spectrum of EF. HFpEF is no longer considered as one disease entity, but has been reconceptualized as a heterogenous group of phenotypes with derangements in multiple organ systems, driven by comorbidities. This heterogeneity suggests that it should not be considered as a single group in terms of treatment goals or clinical approach. Future research at the higher range of EF should ideally tailor investigations for unequivocally preserved EF (> 50%), consider the dynamic nature of EF over time, and use low-variability imaging techniques such as CMR. Furthermore, classifications based on pathophysiology and HF phenotypes beyond the EF construct will shape the design of future trials and help narrow down groups of patients who may respond to personalized treatment.


Assuntos
Insuficiência Cardíaca , Inibidores do Transportador 2 de Sódio-Glicose , Humanos , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/tratamento farmacológico , Volume Sistólico , Inibidores do Transportador 2 de Sódio-Glicose/efeitos adversos , Transportador 2 de Glucose-Sódio
2.
Cardiovasc Drugs Ther ; 36(6): 1187-1196, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-34533692

RESUMO

Hypoxia, via the activity of hypoxia-inducible factors (HIFs), plays a crucial role in fibrosis, inflammation, and oxidative injury, processes which are associated with progression of cardiovascular and kidney diseases. HIFs are key transcription heterodimers consisting of regulatory α-subunits (HIF-1α, HIF-2α, HIF-3α) and a constitutive ß-subunit (HIF-ß). The stability of HIFs is regulated by the prolyl hydroxylases (PHDs). Specific PHD inhibitors (PHD-i) are being investigated as a therapeutic approach to modulate the cellular signaling pathways and harness the native protective adaptive responses to hypoxia. Selective inhibition of PHD leads to the stabilization of the HIFs, which is the transcriptional gatekeeper of a multitude of genes involved in angiogenesis, energy metabolism, apoptosis, inflammation, and fibrosis. PHD-i downregulate hepcidin, improve iron absorption, and increase the endogenous production of erythropoietin. Furthermore, this pharmacological group has also been proven to ameliorate ischemic injuries in several organs, opening a new and promising field in cardiovascular research.. In this review, we present the basic and clinical potential of PHD-i treatment in different scenarios, such as ischemic heart disease, cardiac hypertrophy and heart failure, and their interplay with other pharmacological agents with proven cardiovascular benefits, such as sodium-glucose cotransporter 2 (SGLT2) inhibitors.


Assuntos
Inibidores de Prolil-Hidrolase , Humanos , Inibidores de Prolil-Hidrolase/farmacologia , Inibidores de Prolil-Hidrolase/uso terapêutico , Prolil Hidroxilases/metabolismo , Fibrose , Hipóxia , Inflamação , Subunidade alfa do Fator 1 Induzível por Hipóxia
3.
Platelets ; 33(2): 312-319, 2022 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33856288

RESUMO

Stent thrombosis (ST) is a catastrophic event and efforts to reduce its incidence by altering blood-stent interactions are longstanding. A new electret coating technology that produces long-lasting negative charge on stent surface could make them intrinsically resistant to thrombosis. We assessed the thrombogenicity of stents using an annular perfusion model with confocal microscopy, and determined the efficacy of electret coating technology to confer thrombo-resistant properties to standard stents. Using an annular perfusion chamber, Bare Metal Stent (BMS), standard uncoated DES (DES), and Electret-coated DES (e-DES) were exposed to human blood under arterial flow conditions. Deposits of fibrinogen and platelets on the stent surface were analyzed using immunofluorescence staining and confocal microscopy. Surface coverage by fibrinogen and platelets and the deposit/aggregate size were quantified using computerized morphometric analysis. The experimental methodology produced consistent, quantifiable results. Area of stent surface covered by fibrinogen and platelets and the average size of the deposits/aggregates were lowest for e-DES and highest on BMS, with DES in the middle. The size of fibrinogen-deposits showed no differences between the stents. The testing methodology used in our study successfully demonstrated that electret coating confers significant antithrombotic property to DES stents. These findings warrant confirmation in a larger study.


Assuntos
Stents Farmacológicos/normas , Trombose/terapia , Adulto , Feminino , Voluntários Saudáveis , Humanos , Masculino , Estudo de Prova de Conceito , Resultado do Tratamento
4.
Cardiovasc Drugs Ther ; 35(3): 521-532, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32651897

RESUMO

PURPOSE: Chronic kidney disease (CKD) associates with inflammatory and prothrombotic phenotypes, resulting in higher cardiovascular risk. Factor Xa displays functions beyond coagulation, exhibiting proinflammatory effects. The aim of the present study was to investigate whether a direct FXa inhibitor protects from the endothelial dysfunction (ED) caused by uremia. METHODS: Macro (HUVEC) and microvascular (HMEC) endothelial cells (ECs) were exposed to serum from uremic patients or healthy donors, in absence and presence of apixaban (60 ng/ml). We evaluated changes in surface VCAM-1 and ICAM-1, intracellular eNOS, reactive oxygen species (ROS), and von Willebrand Factor (VWF) production by immunofluorescence, reactivity of the extracellular matrix (ECM) towards platelets, and intracellular signaling. RESULTS: ECs exposed to uremic serum triggered dysregulation of all the parameters. Presence of apixaban resulted in decreased expression of VCAM-1 (178 ± 14 to 89 ± 2% on HMEC and 324 ± 71 to 142 ± 25% on HUVEC) and ICAM-1 (388 ± 60 to 111 ± 10% on HMEC and 148 ± 9% to 90 ± 7% on HUVEC); increased eNOS (72 ± 8% to 95 ± 10% on HMEC); normalization of ROS levels (173 ± 21 to 114 ± 13% on HMEC and 165 ± 14 to 127 ± 7% on HUVEC); lower production of VWF (168 ± 14 to 92 ± 4% on HMEC and 151 ± 22 to 99 ± 11% on HUVEC); and decreased platelet adhesion onto ECM (134 ± 22 to 93 ± 23% on HMEC and 161 ± 14 to 117 ± 7% on HUVEC). Apixaban inhibited p38MAPK and p42/44 activation in HUVEC (139 ± 15 to 48 ± 15% and 411 ± 66 to 177 ± 57%, respectively) (p < 0.05 vs control for all parameters). CONCLUSION: Anti-FXa strategies, such as apixaban, prevented ED caused by the uremic milieu, exhibiting anti-inflammatory and antioxidant properties and modulating the reactivity of the ECM.


Assuntos
Inibidores do Fator Xa/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Pirazóis/farmacologia , Piridonas/farmacologia , Uremia/fisiopatologia , Células Endoteliais/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Humanos , Inflamação/fisiopatologia , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Fator de von Willebrand/efeitos dos fármacos
5.
Transfusion ; 59(7): 2436-2445, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30946491

RESUMO

BACKGROUND: Comparative studies on the restoration of hemostasis with different reversal agents after dabigatran therapy have not been performed. We compared the efficacy and prothrombotic potential of the specific antidote idarucizumab with that of previously recommended non-specific procoagulant concentrates. STUDY DESIGN AND METHODS: We explored the in vitro effects of dabigatran (184 ng/mL) on fibrin and platelet-aggregate formation onto a damaged vessel under flow conditions (600 s-1 ). The reversal mechanisms and efficacy of idarucizumab (0.3-3 mg/mL) were compared with that of the non-specific procoagulant concentrates aPCC (25-75 U/Kg), PCC (70 U/Kg), or rFVIIa (120 µg/Kg). Generation of thrombin and prothrombin fragment (F1 + 2), and thromboelastometry parameters of clot formation were measured. RESULTS: Dabigatran caused pronounced reductions in fibrin (87%) and platelet interactions (36%) with damaged vessels (p < 0.01) and significantly impaired thrombin generation and thromboelastometric parameters (delayed dynamics and reduced firmness). Idarucizumab completely normalized rates of fibrin and platelet coverage to baseline values in flow studies; and reversed the alterations in thrombin generation, F1 + 2 and thromboelastometry parameters produced by dabigatran. In comparison, aPCC and PCC only partially compensated for the dabigatran-induced alterations in fibrin deposition, but were unable to fully restore them to baseline values. Reversal with aPCC or PCC improved the majority of alterations in coagulation-related tests, but tended to overcompensate thrombin generation kinetics and significantly increased F1 + 2 levels. CONCLUSION: Idarucizumab antagonizes alterations of direct and indirect biomarkers of hemostasis caused by dabigatran. In our studies, idarucizumab was clearly more efficacious than strategies with non-specific procoagulant concentrates and devoid of the excessive procoagulant tendency observed with the latter.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Dabigatrana/farmacologia , Fibrina/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Animais , Feminino , Humanos , Cinética , Masculino , Coelhos , Tromboelastografia
6.
Am Heart J ; 182: 125-134, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27914492

RESUMO

BACKGROUND: Dual antiplatelet therapy (DAPT) is necessary to prevent thrombosis yet increases bleeding after percutaneous coronary intervention (PCI) with drug-eluting stents (DES). Antiplatelet monotherapy with a potent P2Y12 receptor antagonist may reduce bleeding while maintaining anti thrombotic efficacy compared with conventional DAPT. METHODS: TWILIGHT is a randomized, double-blind placebo-controlled trial evaluating the comparative efficacy and safety of antiplatelet monotherapy versus DAPT in up to 9000 high-risk patients undergoing PCI with DES. Upon enrollment after successful PCI, all patients will be treated with open label low-dose aspirin (81-100 mg daily) plus ticagrelor (90 mg twice daily) for 3 months. Event-free patients will then be randomized in a double-blind fashion to low-dose aspirin versus matching placebo with continuation of open-label ticagrelor for an additional 12 months. The primary hypothesis is that a strategy of ticagrelor monotherapy will be superior with respect to the primary endpoint of bleeding academic research consortium type 2, 3 or 5, while maintaining non-inferiority for ischemic events compared with ticagrelor plus ASA. CONCLUSIONS: TWILIGHT is the largest study to date that is specifically designed and powered to demonstrate reductions in bleeding with ticagrelor monotherapy versus ticagrelor plus ASA beyond 3 months post-procedure in a high-risk PCI population treated with DES. The trial will provide novel insights with respect to the potential role of ticagrelor monotherapy as an alternative for long-term platelet inhibition in a broad population of patients undergoing PCI with DES.


Assuntos
Síndrome Coronariana Aguda/terapia , Adenosina/análogos & derivados , Aspirina , Reestenose Coronária/prevenção & controle , Hemorragia , Intervenção Coronária Percutânea , Adenosina/administração & dosagem , Adenosina/efeitos adversos , Idoso , Aspirina/administração & dosagem , Aspirina/efeitos adversos , Reestenose Coronária/etiologia , Método Duplo-Cego , Quimioterapia Combinada/métodos , Stents Farmacológicos , Feminino , Hemorragia/induzido quimicamente , Hemorragia/prevenção & controle , Humanos , Masculino , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/métodos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/efeitos adversos , Risco Ajustado/métodos , Ticagrelor , Resultado do Tratamento
7.
Eur Heart J ; 33(2): 264-73, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21606082

RESUMO

AIMS: The aim of this study was to investigate the effects of liver X receptors (LXRs)-ß preferential activation by LXR-623 (WAY-252623), a novel LXRs agonist, on plaque progression/regression in a rabbit model of advanced atherosclerosis. METHODS AND RESULTS: Advanced atherosclerosis was induced in New Zealand White Rabbits (n= 41). At the end of atherosclerosis induction, animals underwent a baseline magnetic resonance imaging (MRI) and were randomized to receive LXR-623 (1.5, 5, or 15 mg/kg/day), simvastatin (5 mg/kg/day), or placebo. The combination of LXR-1.5/simvastatin was also tested. After a final MRI, animals were euthanized and their aortas processed for further analysis. Simvastatin significantly reduced lesion progression (-25%; P< 0.01) in comparison with the placebo group. A similar effect was observed in the LXR-1.5 and -5 groups. A significant regression (16.5%; P< 0.01) of existing atherosclerosis was observed in the LXR-1.5/simvastatin group. Histological and molecular analysis showed plaque stabilization in the animals treated with the LXR-1.5 and -5, and LXR-1.5/simvastatin. The effects of LXR-623 were observed in the presence of a non-significant effect on total-cholesterol, low-density lipoproteins-cholesterol, and triglyceride levels. CONCLUSION: The results of the present study show that LXR-623 significantly reduces the progression of atherosclerosis and induces plaque regression in combination with simvastatin. These observations could drive future development of novel anti-atherosclerotic therapeutic approaches.


Assuntos
Anticolesterolemiantes/farmacologia , Aterosclerose/tratamento farmacológico , Indazóis/farmacologia , Receptores Nucleares Órfãos/efeitos dos fármacos , Placa Aterosclerótica/tratamento farmacológico , Sinvastatina/farmacologia , Animais , Aorta Abdominal , Doenças da Aorta/tratamento farmacológico , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Quimiocina CCL2/metabolismo , Ciclo-Oxigenase 2/metabolismo , Progressão da Doença , Combinação de Medicamentos , Sinergismo Farmacológico , Receptores X do Fígado , Angiografia por Ressonância Magnética , Receptores Nucleares Órfãos/antagonistas & inibidores , Placa Aterosclerótica/metabolismo , Coelhos , Distribuição Aleatória , Tromboplastina/metabolismo , Regulação para Cima
8.
Sci Rep ; 13(1): 22969, 2023 12 27.
Artigo em Inglês | MEDLINE | ID: mdl-38151494

RESUMO

We evaluated modifications in the hemostatic balance of different concentrations of apixaban (APIX) in 25 healthy donors and 53 patients treated with aspirin (ASA, n = 21), ASA and clopidogrel (ASA + CLOPI, n = 11), or ASA and ticagrelor (ASA + TICA, n = 21). Blood samples from participants were spiked ex vivo with apixaban 0 (APIX0), 40 (APIX40), and 160 ng/mL (APIX160). We assessed the effects of APIX on (1) clot formation, by ROTEM thromboelastometry; (2) thrombin generation primed by platelets; and (3) platelet and fibrin interactions with a thrombogenic surface, in a microfluidic model with circulating blood. APIX caused dose-related prolongations of clotting time with minimal impact on other ROTEM parameters. Thrombin generation was significantly inhibited by APIX160, with ASA + TICA actions showing the strongest inhibition (p < 0.01 vs APIX0). Microfluidic studies showed that APIX160 was more potent at suppressing platelet and fibrin interactions (p < 0.001 vs. APIX0). APIX40 demonstrated a consistent antithrombotic action but with a favorable protective effect on the structural quality of fibrin. APIX potentiated the antithrombotic effects of current antiplatelet regimens. APIX at 40 ng/mL, enhanced the antithrombotic action of single or dual antiplatelet regimens but was more conservative for hemostasis than the 160 ng/mL concentration.


Assuntos
Fibrinolíticos , Trombina , Humanos , Fibrinolíticos/farmacologia , Trombina/farmacologia , Aspirina/farmacologia , Plaquetas , Fibrina/farmacologia , Inibidores da Agregação Plaquetária/farmacologia
9.
J Cardiovasc Dev Dis ; 9(12)2022 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-36547434

RESUMO

Despite major advancements in the development of safer and more effective anticoagulant agents, bleeding complications remain a significant concern in the treatment of thromboembolic diseases. Improvements in our understanding of the coagulation pathways highlights the notion that the contact pathway-specifically factor XI (FXI)-has a greater role in the etiopathogenesis of thrombosis than in physiological hemostasis. As a result, a number of drugs targeting FXI are currently in different stages of testing and development. This article aims to review the different strategies directed towards FXI-inhibition with a brief summation of the agents in clinical development, and to comment on the therapeutic areas that could be explored for potential indications. Therapeutics targeting FXI/FXIa inhibition have the potential to usher in a new era of anticoagulation therapy.

10.
Eur Heart J ; 31(13): 1573-82, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20097703

RESUMO

AIMS: Depression and anxiety are linked to coronary events but the mechanism(s) remains unclear. We investigated the associations of depression and anxiety with serotonin-mediated platelet hyperactivity in coronary artery disease (CAD) patients in a cross-sectional study. METHODS AND RESULTS: Three months after an acute coronary event, stable CAD patients (n = 83) on aspirin and clopidogrel were evaluated for depression (beck depression inventory) and anxiety (hospital anxiety and depression scale), and their platelet reactivity was measured (optical aggregometry and flow cytometric fibrinogen binding in response to adenosine diphosphate (ADP = 5 microM) and two serotonin + epinephrine doses [5HT:E (L) = 4 microM + 4 microM and 5HT:E (H) = 10 microM + 4 microM]. Platelet reactivity was significantly higher in depressed and anxious than in depressed only or non-depressed-and-non-anxious patients. Aggregation (mean +/- SE) was 41.9 +/- 2.6% vs. 32.2 +/- 2.6% vs. 30.4 +/- 3.7% with 5HT:E (L) and 46.9 +/- 2.7% vs. 35.6 +/- 2.7% vs. 31.7 +/- 3.8% with 5HT:E (H) (P < 0.05 for both). Differences in ADP aggregations were not significant, perhaps because of clopidogrel therapy. Flow cytometry findings were similar. In a multivariate linear regression model adjusted for age, body mass index, and each other, anxiety symptoms independently predicted all 5HT:E-mediated platelet reactivity measures, whereas depression predicted none. CONCLUSION: Anxiety is associated with elevated serotonin-mediated platelet reactivity in stable CAD patients and symptoms of anxiety show strong, independent correlations with platelet function.


Assuntos
Angina Instável/psicologia , Transtornos de Ansiedade/complicações , Transtorno Depressivo/complicações , Infarto do Miocárdio/psicologia , Ativação Plaquetária/fisiologia , Difosfato de Adenosina/farmacologia , Idoso , Angina Instável/sangue , Transtornos de Ansiedade/sangue , Aspirina/uso terapêutico , Clopidogrel , Estudos Transversais , Transtorno Depressivo/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Inibidores da Agregação Plaquetária/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/agonistas , Receptores Acoplados a Proteínas G/agonistas , Serotonina/farmacologia , Ticlopidina/análogos & derivados , Ticlopidina/uso terapêutico
11.
J Am Coll Cardiol ; 77(3): 243-255, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33197559

RESUMO

BACKGROUND: Large clinical trials established the benefits of sodium-glucose cotransporter 2 inhibitors in patients with diabetes and with heart failure with reduced ejection fraction (HFrEF). The early and significant improvement in clinical outcomes is likely explained by effects beyond a reduction in hyperglycemia. OBJECTIVES: The purpose of this study was to assess the effect of empagliflozin on left ventricular (LV) function and volumes, functional capacity, and quality of life (QoL) in nondiabetic HFrEF patients. METHODS: In this double-blind, placebo-controlled trial, nondiabetic HFrEF patients (n = 84) were randomized to empagliflozin 10 mg daily or placebo for 6 months. The primary endpoint was change in LV end-diastolic and -systolic volume assessed by cardiac magnetic resonance. Secondary endpoints included changes in LV mass, LV ejection fraction, peak oxygen consumption in the cardiopulmonary exercise test, 6-min walk test, and quality of life. RESULTS: Empagliflozin was associated with a significant reduction of LV end-diastolic volume (-25.1 ± 26.0 ml vs. -1.5 ± 25.4 ml for empagliflozin vs. placebo, respectively; p < 0.001) and LV end-systolic volume (-26.6 ± 20.5 ml vs. -0.5 ± 21.9 ml for empagliflozin vs. placebo; p < 0.001). Empagliflozin was associated with reductions in LV mass (-17.8 ± 31.9 g vs. 4.1 ± 13.4 g, for empagliflozin vs. placebo, respectively; p < 0.001) and LV sphericity, and improvements in LV ejection fraction (6.0 ± 4.2 vs. -0.1 ± 3.9; p < 0.001). Patients who received empagliflozin had significant improvements in peak O2 consumption (1.1 ± 2.6 ml/min/kg vs. -0.5 ± 1.9 ml/min/kg for empagliflozin vs. placebo, respectively; p = 0.017), oxygen uptake efficiency slope (111 ± 267 vs. -145 ± 318; p < 0.001), as well as in 6-min walk test (81 ± 64 m vs. -35 ± 68 m; p < 0.001) and quality of life (Kansas City Cardiomyopathy Questionnaire-12: 21 ± 18 vs. 2 ± 15; p < 0.001). CONCLUSIONS: Empagliflozin administration to nondiabetic HFrEF patients significantly improves LV volumes, LV mass, LV systolic function, functional capacity, and quality of life when compared with placebo. Our observations strongly support a role for sodium-glucose cotransporter 2 inhibitors in the treatment of HFrEF patients independently of their glycemic status. (Are the "Cardiac Benefits" of Empagliflozin Independent of Its Hypoglycemic Activity? [ATRU-4] [EMPA-TROPISM]; NCT03485222).


Assuntos
Compostos Benzidrílicos/uso terapêutico , Glucosídeos/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Volume Sistólico/efeitos dos fármacos , Idoso , Compostos Benzidrílicos/farmacologia , Técnicas de Imagem Cardíaca , Método Duplo-Cego , Teste de Esforço , Feminino , Glucosídeos/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
12.
Atherosclerosis ; 294: 17-24, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31945614

RESUMO

BACKGROUND AND AIMS: Circulating platelet microparticles (PMP) are the most abundant in bloodstream, are highly procoagulant and contribute to cross-talk with inflammatory cells. The aim of the present study was to investigate the interactions of PMP with platelets and explore the involvement of toll-like receptor 4 (TLR-4). METHODS: PMP were separated by ultracentrifugation of expired platelet concentrates and added to: i) washed platelets, to confirm uptake, by flow cytometry and confocal and transmission electron microscopy, ii) platelet rich plasma (PRP), to assess changes in platelet function due to uptake by aggregometry in response to ADP; and iii) whole blood, to evaluate heterotypic aggregate (HA) formation by flow cytometry. Moreover, whole blood previously enriched with platelets with internalized PMP was used to explore modifications in thromboelastometry parameters (ROTEM). The inhibitory action of anti-TLR-4 was investigated. RESULTS: Confocal and ultrastructural microscopy studies revealed PMP internalization by platelets. Flow cytometry showed PMP-platelet association (p < 0.01 vs controls, at different PMP dilutions). PMP, at 1/20 dilution, increased HA (p < 0.05 vs controls), the percentage of maximal platelet aggregation to ADP (p < 0.05 vs controls), and accelerated clotting and clot formation times (p < 0.05 vs controls). Incubation of platelets with anti-TLR-4 prior to exposure to PMP reduced PMP-platelet association (p < 0.05 vs absence of the antibody), prevented HA formation, reduced maximal platelet aggregation and normalized ROTEM parameters. CONCLUSIONS: Platelets exhibit internalization ability towards their own PMP, a process that potentiates their thrombogenicity and is partially mediated by the innate immunity receptor TLR-4.


Assuntos
Plaquetas/fisiologia , Micropartículas Derivadas de Células/fisiologia , Agregação Plaquetária/fisiologia , Trombose/etiologia , Receptor 4 Toll-Like/fisiologia , Plaquetas/efeitos dos fármacos , Técnicas de Cultura de Células , Micropartículas Derivadas de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Agregação Plaquetária/efeitos dos fármacos , Tromboelastografia , Receptor 4 Toll-Like/antagonistas & inibidores
13.
J Am Coll Cardiol ; 75(6): 578-586, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32057371

RESUMO

BACKGROUND: An evolving strategy in the setting of percutaneous coronary intervention (PCI) involves withdrawal of acetylsalicylic acid (ASA), or aspirin, while maintaining P2Y12 inhibition. However, the pharmacodynamic effects of this approach on blood thrombogenicity and platelet reactivity remain unknown. OBJECTIVES: This study sought to compare the antithrombotic potency of ticagrelor alone versus ticagrelor plus ASA among high-risk patients undergoing PCI with drug-eluting stents. METHODS: This was a mechanistic substudy within the TWILIGHT (Ticagrelor With Aspirin or Alone in High-Risk Patients After Coronary Intervention) trial, which randomized patients undergoing PCI to ticagrelor plus placebo versus ticagrelor plus ASA following 3 months of dual antiplatelet therapy. Substudy participants were enrolled after randomization, at which time ex vivo assays to quantify thrombus size under dynamic flow conditions and platelet reactivity were performed. Pharmacodynamic assessments were repeated 1 to 6 months thereafter. The primary endpoint was thrombus size at the post-randomization visit with platelet reactivity following stimuli to arachidonic acid, collagen, adenosine diphosphate, and thrombin as secondary endpoints. Results were analyzed using analysis of covariance. RESULTS: A total of 51 patients were enrolled, among whom 42 underwent perfusion assays at baseline and follow-up with a median time between studies of 1.5 months. The adjusted mean difference in post-randomization thrombus area was similar between groups: -218.2 µm2 (95% confidence interval [CI]: -575.9 to 139.9 µm2; p = 0.22). Markers sensitive to cyclo-oxygenase-1 blockade, including platelet reactivity in response to arachidonic acid (mean difference: 10.9 U; 95% CI: 1.9 to 19.9 U) and collagen (mean difference: 9.8 U; 95% CI: 0.8 to 18.8 U) stimuli were higher among patients receiving placebo, whereas levels of platelet reactivity were similar with adenosine diphosphate and thrombin. CONCLUSIONS: Among high-risk patients receiving drug-eluting stents, the antithrombotic potency of ticagrelor monotherapy is similar to that of ticagrelor plus ASA with respect to ex vivo blood thrombogenicity, whereas markers sensitive to cyclo-oxygenase-1 blockade are increased in the absence of ASA. (Platelet Substudy of the TWILIGHT Trial; NCT04001374).


Assuntos
Aspirina/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Complicações Pós-Operatórias/prevenção & controle , Trombose/prevenção & controle , Ticagrelor/uso terapêutico , Idoso , Aspirina/farmacologia , Sangue/efeitos dos fármacos , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Intervenção Coronária Percutânea , Inibidores da Agregação Plaquetária/farmacologia , Ticagrelor/farmacologia
14.
J Interv Cardiol ; 22(4): 385-9, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19496900

RESUMO

OBJECTIVES: To compare the absorption of 300 mg clopidogrel administered crushed via nasogastric (NG) tube versus whole tablets taken orally in healthy volunteers. BACKGROUND: Earlier antiplatelet therapy has proven benefits in treatment of myocardial infarction and in patients undergoing PCI. Aspirin can be delivered early in crushed form via NG tube after CABG surgery to prevent graft occlusion. If clopidogrel given crushed via NG tube provides faster absorption, it could allow earlier clopidogrel loading. METHODS: Nine healthy human subjects (34.7 +/- 11.1 years, 5 males) were given 300 mg clopidogrel in crushed form via NG tube with 30 mL water after 8 hours of fasting. Plasma levels of the primary circulating inactive clopidogrel metabolite SR26334 were measured after 20 minutes, 40 minutes, 1, 2, 4, 8, 12, and 24 hours of dosing. Following >or=2 week washout, same subjects swallowed 300 mg clopidogrel (four 75 mg tablets) after an 8-hour fasting and SR26334 levels were measured at the same time points. RESULTS: Plasma SR26334 concentrations peaked earlier after crushed delivery than after oral intake (44 vs. 70 minutes, P = 0.023) and the median peak was 80% higher (13,083 vs. 7,255 ng/mL, respectively, P = 0.021). At 40 minutes, area under the curve was almost twofold greater with NG administration than oral administration (geometric means ratio = 0.5299, 95% CI = 0.28-0.99, P = 0.048), but was similar over the 24-hour period with both administration methods (geometric means ratio = 1.05, 95% CI = 0.84-1.32, P = 0.646). CONCLUSIONS: A 300 mg loading dose of crushed clopidogrel administered via NG tube provides faster and greater bioavailability than an equal dose taken orally as whole tablets. The clinical benefits of this strategy need to be investigated.


Assuntos
Intubação Gastrointestinal , Inibidores da Agregação Plaquetária/uso terapêutico , Ticlopidina/análogos & derivados , Adulto , Área Sob a Curva , Disponibilidade Biológica , Clopidogrel , Intervalos de Confiança , Feminino , Humanos , Masculino , Razão de Chances , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/farmacocinética , Comprimidos , Ticlopidina/administração & dosagem , Ticlopidina/farmacocinética , Ticlopidina/uso terapêutico
15.
Platelets ; 20(8): 531-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19845440

RESUMO

Clopidogrel is a second-generation thienopyridine that irreversibly inhibits the P2Y(12) adenosine diphosphate (ADP) receptor on platelets, preventing platelet aggregation. As early as 2003, researchers have observed inter-patient variability in response to clopidogrel, leading to a multitude of studies investigating the phenomenon of "clopidogrel resistance," due to the possible link between clopidogrel resistance and in-stent thrombosis. However, due to differences in study methodology and the lack of a clear definition, there is confusion about what it means and its clinical implications. Literature searches were performed using the Web of Science Database. Keywords used to search for relevant literature included clopidogrel resistance. While several studies have shown associations between high platelet reactivity and a high incidence of adverse outcomes, the optimal level of platelet inhibition is unknown. Regardless of the term used to describe high platelet reactivity after treatment, evidence shows that this leads to adverse clinical outcomes. Future goals for research should be aimed at developing a standard method of measuring platelet function and investigating determinants of high platelet reactivity. Alternative treatment options for patients with high platelet reactivity in the face of dual antiplatelet therapy are currently being investigated.


Assuntos
Plaquetas , Resistência a Medicamentos , Inibidores da Agregação Plaquetária , Agregação Plaquetária/efeitos dos fármacos , Ticlopidina/análogos & derivados , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Ensaios Clínicos como Assunto , Clopidogrel , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Testes de Função Plaquetária , Stents , Trombose/tratamento farmacológico , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico
16.
Expert Opin Drug Metab Toxicol ; 15(4): 275-285, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30822172

RESUMO

INTRODUCTION: Cardiac metabolism represents a complex network of numerous pathways responsible for an adequate supply of ATP to the incessant contractile apparatus. Impairments of such pathways are associated with myocardial dysfunction. The newest antidiabetic drugs, the SGLT2 inhibitors, have been demonstrated to reduce cardiovascular mortality and heart failure hospitalizations. The mechanisms underlying these benefits are still uncertain; however, they may play a decisive role in restoring energy efficiency to the damaged heart. Areas covered: This article reviews normal cardiac metabolism and contribution of different substrates to fuel supply. Specific attention is devoted to alterations of these pathways and their association with myocardial dysfunction. In addition, the impact of the novel SGLT2 inhibitors on cardiac mortality and heart failure hospitalizations is discussed. Various postulated mechanisms responsible for such benefits are also discussed. Expert opinion: Metabolic alterations seem to play a crucial role in etiology and progression of heart failure. The cardiovascular benefits of the novel SGLT2 inhibitors have attracted more attention to this field. With effects beyond lowering glucose levels, these agents have been reported to induce changes in cardiac metabolism and to exert anti-inflammatory properties that may contribute to their large cardiovascular beneficial effects by improving contractile bioenergetics. Therefore, SGLT2 inhibitors may become an alternative drug to treat heart failure patients, regardless of diabetic status.


Assuntos
Doenças Cardiovasculares/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Doenças Cardiovasculares/mortalidade , Diabetes Mellitus/tratamento farmacológico , Progressão da Doença , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Hospitalização/estatística & dados numéricos , Humanos , Hipoglicemiantes/farmacologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia
17.
Sci Adv ; 5(6): eaav5463, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31206016

RESUMO

Persons with HIV infection (PWH) have increased risk for cardiovascular disease (CVD), but the underlying mechanisms remain unclear. Coronary thrombosis is known to provoke myocardial infarctions, but whether PWH have elevated thrombotic propensity is unknown. We compared thrombogenicity of PWH on antiretroviral therapy versus matched controls using the Badimon chamber. Measures of inflammation, platelet reactivity, and innate immune activation were simultaneously performed. Enrolled PWH were then randomized to placebo, aspirin (81 mg), or clopidogrel (75 mg) for 24 weeks to assess treatment effects on study parameters. Thrombogenicity was significantly higher in PWH and correlated strongly with plasma levels of D-dimer, soluble TNF receptors 1 and 2, and circulating classical and nonclassical monocytes in PWH. Clopidogrel significantly reduced thrombogenicity and sCD14. Our data suggest that higher thrombogenicity, interacting with inflammatory and immune activation markers, contributes to the increased CVD risk observed in PWH. Clopidogrel exhibits an anti-inflammatory activity in addition to its antithrombotic effect in PWH.


Assuntos
Anti-Inflamatórios/uso terapêutico , Aspirina/uso terapêutico , Plaquetas/efeitos dos fármacos , Clopidogrel/uso terapêutico , Trombose Coronária/tratamento farmacológico , Infecções por HIV/tratamento farmacológico , Inibidores da Agregação Plaquetária/uso terapêutico , Adulto , Terapia Antirretroviral de Alta Atividade/métodos , Biomarcadores/sangue , Plaquetas/imunologia , Plaquetas/virologia , Trombose Coronária/complicações , Trombose Coronária/imunologia , Trombose Coronária/virologia , Estudos Transversais , Feminino , Produtos de Degradação da Fibrina e do Fibrinogênio/genética , Produtos de Degradação da Fibrina e do Fibrinogênio/imunologia , Expressão Gênica , Infecções por HIV/complicações , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Imunidade Inata , Inflamação , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/virologia , Agregação Plaquetária/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia
18.
Methods Mol Biol ; 1816: 161-171, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29987818

RESUMO

The Badimon perfusion chamber is an ex vivo model of thrombosis that assesses the thrombogenicity of blood in humans and large animals. It works with native blood thereby excluding any interfering effects of anticoagulants unavoidable with the majority of platelet function testing methodologies. Each variable of the Virchow's triad (blood, blood flow, and endothelial wall) that modulates blood-vessel wall interaction and thrombus formation is incorporated in this perfusion model. These features make this device a valuable tool for the assessment of thrombogenic potential of various diseases and also gauging the efficacy of antithrombotic (anticoagulant and antiplatelet) treatments.


Assuntos
Aorta/patologia , Plaquetas/patologia , Perfusão/métodos , Trombose/patologia , Animais , Anticoagulantes/farmacologia , Aorta/efeitos dos fármacos , Velocidade do Fluxo Sanguíneo , Plaquetas/efeitos dos fármacos , Desenho de Equipamento , Humanos , Processamento de Imagem Assistida por Computador/métodos , Microscopia/métodos , Microtomia/métodos , Perfusão/instrumentação , Inibidores da Agregação Plaquetária/farmacologia , Coloração e Rotulagem/métodos , Suínos , Trombose/tratamento farmacológico , Inclusão do Tecido/métodos
19.
Thromb Haemost ; 97(3): 487-92, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17334518

RESUMO

It was the objective of this study to evaluate the anti-thrombotic potency of direct factor-Xa inhibition with ZK-807834 in stable coronary patients, using an ex-vivo model of arterial thrombus formation. Tissue factor pathway is important in atherothrombosis. Direct factor-Xa blockade may more potently reduce thrombosis and prevent coronary events. Badimon Perfusion Chamber 5-minute quantitative studies have shown 40-55% arterial thrombus reduction with abciximab, 23% with clopidogrel, but none with heparin. Coronary patients (n = 18, 59 +/- 9 years, 55% males) were blindly randomized to four groups receiving 24-hour infusion of a low, medium or high dose of direct factor- Xa inhibitor ZK-807834, or placebo. Arterial thrombus formation was measured in Badimon Chamber at baseline, end-of-infusion [EoI], and four hours and eight hours after EoI, and factor-X activity, prothrombin time [PT] ratio and plasma drug levels were measured simultaneously. For the low-, medium- and high-dose ZK-807834 groups, mean percent-reduction in thrombus size from baseline to EoI were 29%, 34% and 68%, respectively (p < 0.001), and at 8-h post EoI were 11%, 19% and 27%, respectively (p < 0.01). Mean PT-ratio prolongation showed a strong linear relationship (Pearson's r = 0.93) with ZK-807834 plasma concentration. Mean percent-reduction in factor-X activity from baseline was 13%, 42% and 58%, respectively. Placebo had no effect on thrombus size or factor-X activity. In conclusion, direct factor-Xa inhibition with ZK-807834 markedly reduces ex-vivo arterial thrombus formation and factor-X activity in a dose-dependent manner. Plasma levels of ZK-807834 show a strong linear correlation with PT ratio. This direct factor-Xa inhibitor may reduce the need for additional potent glycoprotein IIbIIIa inhibition.


Assuntos
Amidinas/uso terapêutico , Aorta/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Doença da Artéria Coronariana/tratamento farmacológico , Inibidores do Fator Xa , Fibrinolíticos/uso terapêutico , Piridinas/uso terapêutico , Trombose/prevenção & controle , Amidinas/administração & dosagem , Amidinas/farmacocinética , Animais , Aorta/patologia , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/complicações , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Fibrinolíticos/administração & dosagem , Fibrinolíticos/farmacocinética , Humanos , Infusões Intravenosas , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Perfusão , Tempo de Protrombina , Piridinas/administração & dosagem , Piridinas/farmacocinética , Suínos , Trombose/sangue , Trombose/etiologia , Trombose/patologia , Fatores de Tempo , Resultado do Tratamento
20.
Atherosclerosis ; 266: 81-86, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28992468

RESUMO

BACKGROUND AND AIMS: Dronedarone reduced the rate of stroke and transient ischemic attack in patients with paroxysmal atrial fibrillation (AF) in the ATHENA trial. This cannot be explained by its antiarrhythmic effect alone and may involve alternative mechanisms. This study aimed to investigate any direct effect of dronedarone on blood thrombogenicity, independent of its antiarrhythmic effects. METHODS: Blood samples from patients with cardiovascular disease (n = 30) taking no anticoagulant or antiplatelet medication except aspirin were incubated with dronedarone's active metabolite (SR35021A) at 66 ng/ml (am-L) and 119 ng/ml (am-H), i.e., minimum and maximum mean Cmax reported after repeated 400 mg BID dosing. A third aliquot of blood was incubated with vehicle (Control). Antithrombotic effects of dronedarone were assessed using Coagulation Time (CT), Clot Formation Rate (CFR) and Maximum Clot Firmness (MCF) in ThromboElastoMetry and whole blood platelet aggregation in response to ADP, collagen and TRAP. RESULTS: Compared to Control, mean CT was prolonged by am-L and am-H (164 ± 25 s vs. 180 ± 22 and 182 ± 32 s, respectively, p<0.01 for both). Small but statistically significant reductions were observed in CFR (am-L and am-H) and MCF (am-H). Platelet aggregation induced by ADP and TRAP was also reduced (p<0.05 for both) by am-H. CONCLUSIONS: Dronedarone exerts direct anticoagulant and antiplatelet effects on human blood in vitro that are independent of its antiarrhythmic actions. This suggests the reductions in ischemic events reported with dronedarone may not be due to amelioration of AF itself. Additional clinical studies are required to further improve understanding of the mechanisms involved.


Assuntos
Amiodarona/análogos & derivados , Antiarrítmicos/farmacologia , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Idoso , Amiodarona/farmacologia , Testes de Coagulação Sanguínea , Doenças Cardiovasculares/sangue , Dronedarona , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa