Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Life Sci ; 334: 122225, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38084675

ABSTRACT

AIMS: Prostate cancer is among the highest incidence malignancies in men with a prevalence rate increasing in parallel to the rising global trends in metabolic disorders. Whereas a sizeable body of evidence links metabolic impairment to negative prognosis of prostate cancer, the molecular mechanism underlying this connection has not been thoroughly examined. Our previous work showed that localized adipose tissue inflammation occurring in select adipose depots in early metabolic derangement instigated significant molecular, structural, and functional alterations in neighboring tissues underlying the complications observed at this stage. In this context, the periprostatic adipose tissue (PPAT) constitutes an understudied microenvironment with potential influence on the prostatic milieu. MAIN METHODS AND RESULTS: We show that PPAT inflammation occurs in early prediabetes with signs of increased thrombogenic activity including enhanced expression and function of Factor X. This was mirrored by early neoplastic alterations in the prostate with fibrosis, increased epithelial thickness with marked luminal cellular proliferation and enhanced formation of intraepithelial neoplasia. Significantly, interruption of the procoagulant state in PPAT by a 10-day anticoagulant rivaroxaban treatment not only mitigated PPAT inflammation, but also reduced signs of prostatic neoplastic changes. Moreover, rivaroxaban decreased the murine PLum-AD epithelial prostatic cell viability, proliferation, migration, and colony forming capacity, while increasing oxidative stress. A protease-activated receptor-2 agonist reversed some of these effects. SIGNIFICANCE: We provide some evidence of a molecular framework for the crosstalk between PPAT and prostatic tissue leading to early neoplastic changes in metabolic impairment mediated by upregulation of PPAT thromboinflammation.


Subject(s)
Prostatic Neoplasms , Thrombosis , Male , Humans , Animals , Mice , Rivaroxaban/pharmacology , Rivaroxaban/metabolism , Thromboinflammation , Inflammation/pathology , Thrombosis/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Adipose Tissue/metabolism , Tumor Microenvironment
2.
BMC Cardiovasc Disord ; 23(1): 282, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37268884

ABSTRACT

BACKGROUND: We evaluated the efficacy of the factor Xa inhibitor rivaroxaban on the differentiation ability of vascular endothelial progenitor cells (EPCs), which play roles in vascular injury repair and atherogenesis. Antithrombotic treatment in patients with atrial fibrillation undergoing percutaneous coronary intervention (PCI) is challenging, and current guidelines recommend oral anticoagulant monotherapy 1 year or more after PCI. However, biological evidence of the pharmacological effects of anticoagulants is insufficient. METHODS: EPC colony-forming assays were performed using peripheral blood-derived CD34-positive cells from healthy volunteers. Adhesion and tube formation of cultured EPCs were assessed in human umbilical cord-derived CD34-positive cells. Endothelial cell surface markers were assessed using flow cytometry, and Akt and endothelial nitric oxide synthase (eNOS) phosphorylation were examined using western blot analysis of EPCs. Adhesion, tube formation and endothelial cell surface marker expression was observed in EPCs transfected with small interfering RNA (siRNA) against protease-activated receptor (PAR)-2. Finally, EPC behaviors were assessed in patients with atrial fibrillation undergoing PCI in whom warfarin was changed to rivaroxaban. RESULTS: Rivaroxaban increased the number of large EPC colonies and increased the bioactivities of EPCs, including adhesion and tube formation. Rivaroxaban also increased vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, Tie-2, and E-selectin expression as well as Akt and eNOS phosphorylation. PAR-2 knockdown increased the bioactivities of EPCs and endothelial cell surface marker expression. Patients in whom the number of large colonies increased after switching to rivaroxaban showed better vascular repair. CONCLUSIONS: Rivaroxaban increased the differentiation ability of EPCs, leading to potential advantages in the treatment of coronary artery disease.


Subject(s)
Atrial Fibrillation , Endothelial Progenitor Cells , Percutaneous Coronary Intervention , Humans , Endothelial Progenitor Cells/metabolism , Rivaroxaban/pharmacology , Rivaroxaban/metabolism , Factor Xa Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Atrial Fibrillation/diagnosis , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Fibrinolytic Agents/adverse effects , Percutaneous Coronary Intervention/adverse effects , Cell Differentiation/genetics , Cells, Cultured , Cell Movement
3.
Dis Markers ; 2022: 1118195, 2022.
Article in English | MEDLINE | ID: mdl-36438904

ABSTRACT

Background: Mitochondria have been involved in host defense upon viral infections. Factor Xa (FXa), a coagulating factor, may also have influence on mitochondrial functionalities. The aim was to analyze if in human pulmonary microvascular endothelial cells (HPMEC), the SARS-CoV-2 (COVID-19) spike protein subunits, S1 and S2 (S1+S2), could alter mitochondrial metabolism and what is the role of FXA. Methods: HPMEC were incubated with and without recombinants S1+S2 (10 nmol/L each). Results: In control conditions, S1+S2 failed to modify FXa expression. However, in LPS (1 µg/mL)-incubated HPMEC, S1+S2 significantly increased FXa production. LPS tended to reduce mitochondrial membrane potential with respect to control, but in higher and significant degree, it was reduced when S1+S2 were present. LPS did not significantly modify cytochrome c oxidase activity as compared with control. Addition of S1+S2 spike subunits to LPS-incubated HPMEC significantly increased cytochrome c oxidase activity with respect to control. Lactate dehydrogenase activity was also increased by S1+S2 with respect to control and LPS alone. Protein expression level of uncoupled protein-2 (UCP-2) was markedly expressed when S1+S2 were added together to LPS. Rivaroxaban (50 nmol/L), a specific FXa inhibitor, significantly reduced all the above-mentioned alterations induced by S1+S2 including UCP-2 expression. Conclusions: In HPMEC undergoing to preinflammatory condition, COVID-19 S1+S2 spike subunits promoted alterations in mitochondria metabolism suggesting a shift from aerobic towards anaerobic metabolism that was accompanied of high FXa production. Rivaroxaban prevented all the mitochondrial metabolic changes mediated by the present COVID-19 S1 and S2 spike subunits suggesting the involvement of endogenous FXa.


Subject(s)
COVID-19 , Factor Xa Inhibitors , Factor Xa , Mitochondria , Rivaroxaban , Spike Glycoprotein, Coronavirus , Humans , COVID-19/genetics , COVID-19/metabolism , Electron Transport Complex IV/metabolism , Endothelial Cells/metabolism , Factor Xa/genetics , Factor Xa/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Protein Subunits/metabolism , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment , Factor Xa Inhibitors/metabolism , Factor Xa Inhibitors/pharmacology , Factor Xa Inhibitors/therapeutic use , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
4.
Diab Vasc Dis Res ; 19(5): 14791641221129877, 2022.
Article in English | MEDLINE | ID: mdl-36250331

ABSTRACT

PURPOSE: Combination of Rivaroxaban plus Aspirin improved cardiovascular outcome in patients with stable cardiovascular disease. The aim was to determine if Rivaroxaban and acetylsalicylic acid alone or in combination may protect mitochondrial mitophagy in human coronary artery endothelial cells (HCAEC) exposed to D-glucose. METHODS: HCAEC were incubated under different conditions: 5 mmol/L glucose D-glucose (control), 30 mmol/L D-Glucose with and without 50 nmol/L Rivaroxaban (Rivaroxaban), 0.33 mmol/L ASA (ASA) or Rivaroxaban (12.5 nmol/L)+ASA (0.33 mmol/L; (Riva+ASA). RESULTS: HCAEC incubated with D-glucose showed an increased Factor Xa expression. The mitochondrial content of Pink-1 and Parkin were significantly reduced in high glucose-incubated HCAEC compared to control. Rivaroxaban+ASA significantly increased the mitochondrial content of Pink-1 and Parkin, and the mitochondrial membrane potential compared to D-Glucose group. Both ASA alone and Riva+ASA reduced reactive oxygen species (ROS) and tissue factor production induced by high glucose exposure. CONCLUSION: Under high glucose condition combining Rivaroxaban+ASA increased the mitochondrial content of Pink-1 and Parkin, restored mitochondria membrane potential and reduced ROS and tissue factor expression in HCAEC. It suggests potential effects induced by dual use of Rivaroxaban and ASA on the coronary endothelium subjected to high glucose condition.


Subject(s)
Aspirin , Rivaroxaban , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelium , Factor Xa/metabolism , Factor Xa/pharmacology , Glucose/metabolism , Humans , Mitochondria , Mitophagy , Reactive Oxygen Species/metabolism , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Thromboplastin/metabolism , Thromboplastin/pharmacology , Ubiquitin-Protein Ligases/metabolism
5.
Atherosclerosis ; 345: 7-14, 2022 03.
Article in English | MEDLINE | ID: mdl-35183904

ABSTRACT

BACKGROUND AND AIMS: The platelet inhibitor aspirin reduces inflammation and atherosclerosis in both apolipoprotein E deficient (apoE-/-) mice and low-density lipoprotein receptor deficient (Ldlr-/-) mice. Similarly, the factor Xa inhibitor rivaroxaban reduces atherosclerosis in both apoE-/- and Ldlr-/- mice. We tested the hypothesis that the combination of aspirin and rivaroxaban reduces atherosclerosis in mice to a greater extent than either agent alone. METHODS: Male Ldlr-/- mice were fed a western-type diet for 12 weeks to induce atherosclerosis. Cohorts of mice received aspirin in the water and/or rivaroxaban in the diet. Atherosclerosis and lesion composition were measured in the aortic sinus and the aorta. Expression of 55 proteins in the aorta and plasma was determined using multiplex ELISA assays. RESULTS: Aspirin alone, rivaroxaban alone, and the combination of both agents significantly reduced atherosclerosis in the Ldlr-/- mice compared with control Ldlr-/- mice fed a western-type diet. However, there were no significant differences in atherosclerosis in the group receiving aspirin and rivaroxaban compared with the groups that received aspirin or rivaroxaban alone. Aspirin, rivaroxaban and the combination reduced macrophage content and apoptosis in the lesions compared with controls but there was no difference between the three treatment groups. We observed statistically significant changes in the expression of a small number of proteins in the aorta and plasma in mice treated with aspirin and/or rivaroxaban. CONCLUSIONS: Contrary to our expectation, the combination of aspirin and rivaroxaban did not further reduce atherosclerosis in Ldlr-/- mice beyond the level observed with each agent alone.


Subject(s)
Atherosclerosis , Rivaroxaban , Animals , Aorta/pathology , Apolipoproteins E , Aspirin/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL , Rivaroxaban/metabolism , Rivaroxaban/pharmacology
6.
PLoS One ; 17(1): e0262482, 2022.
Article in English | MEDLINE | ID: mdl-35015795

ABSTRACT

Based on previous large-scale in silico screening several factor Xa inhibitors were proposed to potentially inhibit SARS-CoV-2 Mpro. In addition to their known anticoagulants activity this potential inhibition could have an additional therapeutic effect on patients with COVID-19 disease. In this study we examined the binding of the Apixaban, Betrixaban and Rivaroxaban to the SARS-CoV-2 Mpro with the use of the MicroScale Thermophoresis technique. Our results indicate that the experimentally measured binding affinity is weak and the therapeutic effect due to the SARS-CoV-2 Mpro inhibition is rather negligible.


Subject(s)
Coronavirus M Proteins/antagonists & inhibitors , Factor Xa Inhibitors/chemistry , SARS-CoV-2/metabolism , Benzamides/chemistry , Benzamides/metabolism , Binding Sites , COVID-19/virology , Coronavirus M Proteins/metabolism , Factor Xa Inhibitors/metabolism , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Stability , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyridines/chemistry , Pyridines/metabolism , Pyridones/chemistry , Pyridones/metabolism , Rivaroxaban/chemistry , Rivaroxaban/metabolism , SARS-CoV-2/isolation & purification , COVID-19 Drug Treatment
7.
Mol Pharmacol ; 100(3): 224-236, 2021 09.
Article in English | MEDLINE | ID: mdl-34210765

ABSTRACT

Mounting evidence has revealed that despite the high degree of sequence homology between cytochrome P450 3A isoforms (i.e., CYP3A4 and CYP3A5), they have the propensities to exhibit vastly different irreversible and reversible interactions with a single substrate. We have previously established that benzbromarone (BBR), a potent uricosuric agent used in the management of gout, irreversibly inhibits CYP3A4 via mechanism-based inactivation (MBI). However, it remains unelucidated if CYP3A5-its highly homologous counterpart-is susceptible to inactivation by BBR. Using three structurally distinct probe substrates, we consistently demonstrated that MBI was not elicited in CYP3A5 by BBR. Our in silico covalent docking models and molecular dynamics simulations suggested that disparities in the susceptibilities toward MBI could be attributed to the specific effects of BBR covalent adducts on the F-F' loop. Serendipitously, we also discovered that BBR reversibly activated CYP3A5-mediated rivaroxaban hydroxylation wherein apparent V max increased and K m decreased with increasing BBR concentration. Fitting data to the two-site model yielded interaction factors α and ß of 0.44 and 5.88, respectively, thereby confirming heterotropic activation of CYP3A5 by BBR. Furthermore, heteroactivation was suppressed by the CYP3A inhibitor ketoconazole in a concentration-dependent manner and decreased with increasing preincubation time, implying that activation was incited via binding of parent BBR molecule within the enzymatic active site. Finally, noncovalent docking revealed that CYP3A5 can more favorably accommodate both BBR and rivaroxaban in concert as compared with CYP3A4, which further substantiated our experimental observations. SIGNIFICANCE STATEMENT: Although it has been previously demonstrated that benzbromarone (BBR) inactivates CYP3A4, it remains uninterrogated whether it also elicits mechanism-based inactivation in CYP3A5, which shares ∼85% sequence similarity with CYP3A4. This study reported that BBR exhibited differential irreversible and reversible interactions with both CYP3A isoforms and further unraveled the molecular determinants underpinning their diverging interactions. These data offer important insight into differential kinetic behavior of CYP3A4 and CYP3A5, which potentially contributes to interindividual variabilities in drug disposition.


Subject(s)
Benzbromarone/chemistry , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A/chemistry , Benzbromarone/metabolism , Benzbromarone/pharmacology , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Humans , Hydroxylation/drug effects , Hydroxylation/physiology , Inhibitory Concentration 50 , Midazolam/metabolism , Midazolam/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Testosterone/metabolism , Testosterone/pharmacology
8.
Tissue Barriers ; 9(4): 1956284, 2021 Oct 02.
Article in English | MEDLINE | ID: mdl-34323663

ABSTRACT

BACKGROUND: Rivaroxaban is one of the direct factor Xa inhibitors. Its function in the inactivated coagulation cascade is unclear. The aim of the study was to assess the effect of rivaroxaban on the endothelial integrity and inflammatory properties of endothelial cells stimulated by 25-hydroxycholesterol (25-OHC). METHODS: HUVECs were stimulated with 25-OHC, rivaroxaban and 25-OHC+ rivaroxaban. HUVEC integrity and permeability were measured using the xCELLigence system and paracellular flux assay. The mRNA expression of tissue factor, ICAM-1, VEGF, IL-33, MCP-1, TNF-α was analyzed in the real-time PCR. Apoptosis and viability were measured by flow cytometry. The VEGF protein concentration was assessed by ELISA. The confocal microscope was used to evaluate the expression of VE-cadherin in endothelial cells. RESULTS: 25-OHC decreased endothelial cell integrity and increased the mRNA expression of IL-33, tissue factor, ICAM-1, MCP-1, VEGF, TNF-α as compared to unstimulated controls. Following the stimulation with rivaroxaban, HUVEC restored integrity disrupted by 25-OHC (p < .01). In HUVECs pre-stimulated with oxysterol, rivaroxaban decreased mRNA expression of IL-33, TNF-α, chemokines MCP-1, ICAM-1, VEGF and tissue factor (p < .01). Rivaroxaban 100 mg/ml+25-OHC increased the VE-cadherin expression in endothelium as compared to 25-OHC (p < .05). CONCLUSION: Our finding suggests that rivaroxaban may restore the endothelial barrier and inhibit the inflammatory activation caused by oxysterol in vitro.


Subject(s)
Oxysterols , Rivaroxaban , Endothelium, Vascular , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Oxysterols/metabolism , Oxysterols/pharmacology , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use
9.
Ther Drug Monit ; 42(5): 737-743, 2020 10.
Article in English | MEDLINE | ID: mdl-32433187

ABSTRACT

BACKGROUND: Oral factor Xa inhibitors (OFXais) may interfere with the heparin antifactor Xa (antiXa) assay. The best method to measure heparin activity during the transition from an OFXai to intravenous (IV) unfractionated heparin (UFH) remains unknown. This study aimed to assess the safety and effectiveness of transitioning from an OFXai to UFH. METHODS: A retrospective analysis was conducted of patients with supratherapeutic antiXa levels on UFH who received either apixaban or rivaroxaban within 72 hours before UFH initiation at NYU Langone Health. The primary objective was to identify the incidence of interference on the heparin antiXa assay due to OFXai exposure in the previous 72 hours. The secondary outcomes included the indication for transition to UFH and the rate of thromboembolic and bleeding events. RESULTS: A total of 93 patients with supratherapeutic antiXa activity levels with prior OFXai use were reviewed. Moderate renal impairment, defined as creatinine clearance less than 49 mL/min, was present in 67 (72%) patients. The primary indication for transition from OFXai to UFH was in anticipation for a procedure, and it occurred in 37 (40%) patients. There were 3 major bleeding events and 3 clinically relevant nonmajor bleeding events. No thromboembolic events occurred. CONCLUSIONS: This study assessed the prevalence of supratherapeutic antiXa levels and clinical outcomes during the transition from OFXais to UFH. Health care systems should develop guidelines to assist clinicians in monitoring antiXa activity in patients undergoing a transition from an OFXai to UFH. It is also important to assess the patient's underlying thromboembolic and bleeding risks.


Subject(s)
Factor Xa Inhibitors/metabolism , Fibrinolytic Agents/metabolism , Heparin/metabolism , Administration, Oral , Aged , Aged, 80 and over , Anticoagulants/metabolism , Anticoagulants/therapeutic use , Blood Coagulation Tests/methods , Drug Monitoring/methods , Factor Xa Inhibitors/therapeutic use , Female , Fibrinolytic Agents/therapeutic use , Hemorrhage/drug therapy , Hemorrhage/metabolism , Heparin/therapeutic use , Humans , Male , Pyrazoles/metabolism , Pyrazoles/therapeutic use , Pyridones/metabolism , Pyridones/therapeutic use , Retrospective Studies , Rivaroxaban/metabolism , Rivaroxaban/therapeutic use , Thromboembolism/drug therapy , Thromboembolism/metabolism
10.
Bull Cancer ; 107(5): 574-585, 2020 May.
Article in French | MEDLINE | ID: mdl-32252973

ABSTRACT

Direct oral anticoagulants, anti-IIa or anti-Xa, are widely used in the treatment and prevention of venous thromboembolic disease as well as in nonvalvular atrial fibrillation. Direct oral anticoagulants are characterized by a rapid onset of activity, a predictable response and a relatively wide therapeutic window. Nevertheless, theoretical drug interactions exist since direct oral anticoagulants are substrates of the transport protein P-glycoprotein and/or of isoforms of cytochromes P450 pathway. Direct oral anticoagulants do not have a marketing authorization for the treatment of cancer-associated thrombosis unlike low-molecular-weight heparins which remain the gold standard treatment today. However, recent studies have compared low-molecular-weight heparins to direct oral anticoagulants in the treatment of cancer-associated thrombosis. Results of these studies showed a non-inferiority of direct oral anticoagulants in the prevention of recurrent thromboembolic events but at the cost of an increased hemorrhagic risk, in particular for patients with gastrointestinal and urogenital cancers. Thus, international guidelines, unlike French guidelines, integrate them in first line of the therapeutic strategy of cancer patients. We are certainly entering an era of personalized therapy for cancer-associated thrombosis, considering cancer type and also the theoretical risk of drug interactions with anti-cancer treatments or supportive care.


Subject(s)
Factor Xa Inhibitors/therapeutic use , Neoplasms/complications , Thrombosis/drug therapy , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Anticoagulants/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cytochrome P-450 Enzyme System/metabolism , Dabigatran/metabolism , Dabigatran/therapeutic use , Drug Interactions , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/metabolism , Hemorrhage/chemically induced , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Neoplasm Proteins/metabolism , Neoplasms/blood , Neoplasms/drug therapy , Pyrazoles/metabolism , Pyrazoles/therapeutic use , Pyridines/metabolism , Pyridines/therapeutic use , Pyridones/metabolism , Pyridones/therapeutic use , Recurrence , Rivaroxaban/metabolism , Rivaroxaban/therapeutic use , Secondary Prevention , Thiazoles/metabolism , Thiazoles/therapeutic use , Thrombosis/etiology , Venous Thromboembolism/prevention & control
11.
Reumatol. clín. (Barc.) ; 16(2,pt.1): 120-121, mar.-abr. 2020. ilus
Article in Spanish | IBECS | ID: ibc-194331

ABSTRACT

Se presenta el caso de un varón de 83 años en tratamiento con rivaroxabán, con dolor en hombro, cara posterior de brazo, antebrazo y muñeca derechos e incapacidad para extender muñeca y dedos, tras un traumatismo. Las radiografías son normales, pero en la TC torácica se objetiva seudoaneurisma de arteria axilar y un hematoma secundario que comprime el plexo braquial. Esta entidad ha de tenerse en cuenta en pacientes con hombro doloroso, anticoagulados y sin alteraciones radiológicas asociadas


We report the case of an 83-year-old man on rivaroxaban treatment, with pain in right shoulder, back of the upper arm, forearm and wrist, and inability to extend the wrist and fingers, as the result of an injury. The radiographs were normal, but thoracic CT showed an axillary artery pseudoaneurysm and a secondary hematoma that compressed the brachial plexus. This should be borne in mind in patients with painful shoulder, treated with anticoagulant therapy and without associated radiographic changes


Subject(s)
Humans , Male , Aged, 80 and over , Shoulder Pain/drug therapy , Rivaroxaban/administration & dosage , Shoulder Pain/diagnostic imaging , Atrial Fibrillation/drug therapy , Rivaroxaban/metabolism , Tomography, X-Ray Computed , Aneurysm, False/diagnostic imaging , Axillary Artery/diagnostic imaging , Elbow/diagnostic imaging , Hand/diagnostic imaging
12.
Brain Res ; 1718: 201-211, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31103522

ABSTRACT

The timing of anti-coagulation therapy initiation after acute cardioembolic stroke remains controversial. We investigated the effects of post-stroke administration of a factor Xa inhibitor in mice, focusing on tissue repair and functional restoration outcomes. We initiated administration of rivaroxaban, a Xa inhibitor, immediately after permanent distal middle cerebral artery occlusion (pMCAO) in CB-17 mice harboring few leptomeningeal anastomoses at baseline. Rivaroxaban initiated immediately after pMCAO hindered the recovery of blood flow in ischemic areas by inhibiting leptomeningeal anastomosis development, and led to impaired restoration of neurologic functions with less extensive peri-infarct astrogliosis. Within infarct areas, angiogenesis and fibrotic responses were attenuated in rivaroxaban-fed mice. Furthermore, inflammatory responses, including the accumulation of neutrophils and monocytes/macrophages, local secretion of pro-inflammatory cytokines, and breakdown of the blood-brain barrier, were enhanced in infarct areas in mice treated immediately with rivaroxaban following pMCAO. The detrimental effects were not found when rivaroxaban was initiated after transient MCAO or on day 7 after pMCAO. Collectively, early post-stroke initiation of a factor Xa inhibitor may suppress leptomeningeal anastomosis development and blood flow recovery in ischemic areas, thereby resulting in attenuated tissue repair and functional restoration unless occluded large arteries are successfully recanalized.


Subject(s)
Rivaroxaban/metabolism , Stroke/drug therapy , Animals , Blood-Brain Barrier/drug effects , Brain Ischemia/physiopathology , Disease Models, Animal , Factor Xa/metabolism , Factor Xa Inhibitors/metabolism , Factor Xa Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Rivaroxaban/pharmacology , Stroke/physiopathology , Time Factors
13.
AAPS PharmSciTech ; 19(4): 1847-1859, 2018 May.
Article in English | MEDLINE | ID: mdl-29637496

ABSTRACT

In this paper, a novel self-nanoemulsifying drug delivery system (SNEDDS) was used to improve the oral bioavailability in fasted state and diminish the food effect for rivaroxaban. Oil, surfactant, and co-surfactant were selected by saturated solubility study. IPM, Tween80, and 1,2-propanediol were finally selected as oil, surfactant, and co-surfactant, respectively. The pseudo-ternary-phase diagram was utilized to optimize the preliminary composition of SNEDDS formulation. The optimized rivaroxaban-SNEDDS formulation was selected by central composite design (CCD) of response surface methodology. Optimized SNEDDS formulation was evaluated for drug content, self-emulsifying time, droplet size, zeta potential, polydispersity index, Fourier transform-infrared (FTIR) spectroscopy, and transmission electron microscope (TEM). The drug dissolution profile compared to the commercial formulation Xarelto® (20 mg rivaroxaban) was determined in four different media (pH 1.2HCl, pH 4.5NaAc-HAc, pH 6.8PBS, and water). The result indicated that the SNEDDS formulation had successfully increased the drug solubility in four different media. A HPLC-MS method that indicated a high sensitivity, strong attribute, and high accuracy characteristic was built to measure the drug concentration in plasma. The fast/fed in vivo pharmacokinetics studies of SNEDDS formulation and Xarelto® were carried out in adult beagle dog, rivaroxaban with no food effect was achieved in SNEDDS formulation compared with Xarelto® in fed state. The result suggested that SNEDDS formulation in this study is useful to increase the oral bioavailability and diminish the food effect in fasted state.


Subject(s)
Drug Delivery Systems/methods , Emulsifying Agents/administration & dosage , Food-Drug Interactions/physiology , Nanoparticles/administration & dosage , Rivaroxaban/administration & dosage , Administration, Oral , Animals , Biological Availability , Dogs , Drug Liberation , Emulsifying Agents/chemical synthesis , Emulsions , Fasting/metabolism , Female , Male , Nanoparticles/chemistry , Nanoparticles/metabolism , Particle Size , Rivaroxaban/chemical synthesis , Rivaroxaban/metabolism , Solubility , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemical synthesis , Surface-Active Agents/metabolism
14.
J Pharmacol Exp Ther ; 365(3): 519-525, 2018 06.
Article in English | MEDLINE | ID: mdl-29572341

ABSTRACT

Because of their lower bleeding risk and simplicity of use, direct oral anticoagulants (DOACs) could represent an interesting alternative to conventional anticoagulant treatment with vitamin K antagonists for patients with pulmonary arterial hypertension (PAH). P-glycoprotein (P-gp) plays a key role in DOAC pharmacokinetics. Type 5-phosphodiesterase inhibitors (PDE5is), a drug class commonly used in the treatment of PAH, have been shown to strongly inhibit P-gp. This work aimed to assess potential P-gp-mediated drug-drug interactions between PDE5is and DOACs using in vitro methods. A cellular model of drug transport assay, using P-gp-overexpressing Madin-Darby canine kidney cells (transfected with the human P-gp gene), was used to determine the bidirectional permeabilities of two DOACs (rivaroxaban and apixaban) in the absence and presence of increasing concentrations (0.5-100 µM) of three PDE5is (sildenafil, tadalafil, and vardenafil). Permeabilities and efflux ratios were calculated from DOAC concentrations, were measured with liquid chromatography coupled with mass spectrometry, and were subsequently used to determine the PDE5i percentage of inhibition and half maximal inhibitory concentration (IC50 ). Rivaroxaban efflux was inhibited by 99%, 66%, and 100% with 100 µM sildenafil, tadalafil, and vardenafil, respectively. Similarly, apixaban efflux was inhibited by 97%, 74%, and 100%, respectively. The IC50 values of the three PDE5is were 8, 28, and 5 µM for rivaroxaban and 23, 15, and 3 µM for apixaban, respectively. This study showed strong in vitro inhibition of DOAC efflux by PDE5is. In vivo studies are required to determine the clinical relevance of these interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anticoagulants/pharmacokinetics , Phosphodiesterase 5 Inhibitors/pharmacology , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Rivaroxaban/pharmacokinetics , Administration, Oral , Animals , Anticoagulants/administration & dosage , Anticoagulants/metabolism , Biological Transport/drug effects , Dogs , Drug Interactions , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Pyrazoles/administration & dosage , Pyrazoles/metabolism , Pyridones/administration & dosage , Pyridones/metabolism , Rivaroxaban/administration & dosage , Rivaroxaban/metabolism , Tissue Distribution/drug effects
15.
BMC Cardiovasc Disord ; 17(1): 260, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-29029621

ABSTRACT

BACKGROUND: Novel anticoagulations (NOACs) are increasingly prescribed for the prevention of stroke in premenopausal women with atrial fibrillation. Small studies suggest NOACs are associated with a higher risk of abnormal uterine bleeds than vitamin K antagonists (VKAs). Because there is no direct empirical evidence on the benefit/risk profile of rivaroxaban compared to VKAs in this subgroup, we synthesize available indirect evidence, estimate decision uncertainty on the treatments, and assess whether further research in premenopausal women is warranted. METHODS: A Markov model with annual cycles and a lifetime horizon was developed comparing rivaroxaban (the most frequently prescribed NOAC in this population) and VKAs. Clinical event rates, associated quality adjusted life years, and health care costs were obtained from different sources and adjusted for gender, age, and history of stroke. A Monte Carlo simulation with 10,000 iterations was then performed for a hypothetical cohort of premenopausal women, estimated to be reflective of the population of premenopausal women with AF in The Netherlands. RESULTS: In the simulation, rivaroxaban is the better treatment option for the prevention of ischemic strokes in premenopausal women in 61% of the iterations. Similarly, this is 98% for intracranial hemorrhages, 24% for major abnormal uterine bleeds, 1% for minor abnormal uterine bleeds, 9% for other major extracranial hemorrhages, and 23% for other minor extracranial hemorrhages. There is a 78% chance that rivaroxaban offers the most quality-adjusted life years. The expected value of perfect information in The Netherlands equals 122 quality-adjusted life years and 22 million Euros. CONCLUSIONS: There is a 22% risk that rivaroxaban offers a worse rather than a better benefit/risk profile than vitamin K antagonists in premenopausal women. Although rivaroxaban is preferred over VKAs in this population, further research is warranted, and should preferably take the shape of an internationally coordinated registry study including other NOACs.


Subject(s)
Atrial Fibrillation/drug therapy , Empirical Research , Factor Xa Inhibitors/therapeutic use , Markov Chains , Premenopause/drug effects , Rivaroxaban/therapeutic use , Adult , Atrial Fibrillation/epidemiology , Atrial Fibrillation/metabolism , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/metabolism , Female , Humans , Premenopause/metabolism , Risk Factors , Rivaroxaban/adverse effects , Rivaroxaban/metabolism , Treatment Outcome , Young Adult
16.
J Pharm Sci ; 106(9): 2524-2534, 2017 09.
Article in English | MEDLINE | ID: mdl-28456731

ABSTRACT

This study aimed to investigate the interactions of 3 anticoagulants, rivaroxaban, apixaban, and dabigatran, with 5 human solute carrier transporters, hOAT1, hOAT3, hOCT2, hOATP1B1, and hOATP1B3. Apixaban inhibited hOAT3, hOATP1B1, and hOATP1B3, and rivaroxaban inhibited hOAT3 and hOATP1B3, with IC50 values of >20 and >5 µM, respectively. The effect of dabigatran was negligible or very weak, so significant drug interactions at therapeutic doses are unlikely. Specific uptake of rivaroxaban was observed only in human and mouse OAT3-expressing cells. The Km for mouse Oat3 (mOat3) was 1.01 ± 0.70 µM. A defect in mOat3 reduced the kidney-to-plasma concentration ratio of rivaroxaban by 38% in mice. Probenecid treatment also reduced the kidney-to-plasma concentration ratio of rivaroxaban in rats by 73%. Neither mOat3 defect nor probenecid administration in rats reduced the renal clearance of rivaroxaban. The uptake of rivaroxaban by monkey kidney slices was temperature dependent and inhibited by probenecid but not by tetraethylammonium. Taken together, organic anion transporters, mainly OAT3, may mediate basolateral uptake of rivaroxaban in kidneys. hOAT3 could be an additional factor that differentiates the potential drug-drug interactions of the 3 anticoagulants in the urinary excretion process in clinical settings.


Subject(s)
Anticoagulants/pharmacokinetics , Dabigatran/pharmacokinetics , Kidney/metabolism , Organic Anion Transporters/metabolism , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Rivaroxaban/pharmacokinetics , Animals , Anticoagulants/metabolism , Anticoagulants/pharmacology , Biological Transport/drug effects , Dabigatran/metabolism , Dabigatran/pharmacology , Drug Interactions , Female , HEK293 Cells , Haplorhini , Humans , Kidney/drug effects , Mice , Mice, Inbred C57BL , Organic Anion Transporters/antagonists & inhibitors , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyridones/metabolism , Pyridones/pharmacology , Rats , Rats, Sprague-Dawley , Rivaroxaban/metabolism , Rivaroxaban/pharmacology
17.
J Plast Reconstr Aesthet Surg ; 69(5): 585-93, 2016 May.
Article in English | MEDLINE | ID: mdl-27013144

ABSTRACT

Novel oral anticoagulants (NOACs) have emerged as a good alternative to warfarin in the prevention of stroke for patients with atrial fibrillation. NOAC use is increasing rapidly; therefore, greater understanding of their use in the perioperative period is important for optimal care. Studies and reviews that reported on the use of NOACs were identified, with particular focus on the perioperative period. PubMed was searched for relevant articles published between January 2000 and August 2015. The inevitable rise in the use of NOACs such as rivaroxaban (Xarelto™), apixaban (Eliquis™), edoxaban (Lixiana™) and dabigatran (Pradaxa™) may present a simplified approach to perioperative anticoagulant management due to fewer drug interactions, rapidity of onset of action and relatively short half-lives. Coagulation status, however, cannot reliably be monitored and no antidotes are currently available. When planning for discontinuation of NOACs, special consideration of renal function is required. Advice regarding the management of bleeding complications is provided for consideration in emergency surgery. In extreme circumstances, haemodialysis may be considered for bleeding with the use of dabigatran. NOACs will increasingly affect operative planning in plastic surgery. In order to reduce the incidence of complications associated with anticoagulation, the management of NOACs in the perioperative period requires knowledge of the time of last dose, renal function and the bleeding risk of the planned procedure. Consideration of these factors will allow appropriate interpretation of the current guidelines.


Subject(s)
Algorithms , Anticoagulants , Dabigatran , Plastic Surgery Procedures , Pyrazoles , Pyridines , Pyridones , Rivaroxaban , Thiazoles , Administration, Oral , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Anticoagulants/metabolism , Dabigatran/administration & dosage , Dabigatran/adverse effects , Dabigatran/antagonists & inhibitors , Dabigatran/metabolism , Elective Surgical Procedures , Emergencies , Humans , Kidney/metabolism , Liver/metabolism , Perioperative Care , Postoperative Hemorrhage/chemically induced , Practice Guidelines as Topic , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/antagonists & inhibitors , Pyrazoles/metabolism , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/antagonists & inhibitors , Pyridines/metabolism , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/antagonists & inhibitors , Pyridones/metabolism , Rivaroxaban/administration & dosage , Rivaroxaban/adverse effects , Rivaroxaban/antagonists & inhibitors , Rivaroxaban/metabolism , Thiazoles/administration & dosage , Thiazoles/adverse effects , Thiazoles/antagonists & inhibitors , Thiazoles/metabolism
18.
Int Heart J ; 56(5): 544-50, 2015.
Article in English | MEDLINE | ID: mdl-26370374

ABSTRACT

Cell migration, proliferation, and differentiation of cardiac fibroblasts (CFs) play a central role in cardiac fibrosis. Factor Xa (FXa)-dependent protease-activated receptor (PAR)-1 and PAR-2 have been reported as important targets in proinflammatory and fibroproliferative diseases. From this viewpoint, we aimed to investigate whether treatment of rivaroxaban, an approved oral direct FXa inhibitor, attenuates functional changes in angiotensin (Ang) II-induced mouse CFs.Confluent cultured mouse CFs were pretreated with or without rivaroxaban. Ang II-induced cell migration was decreased by 73% in rivaroxaban induced cells. Rivaroxaban inhibited Ang II-induced cell proliferation by 27% at 0.01 µg/ mL, 69% at 0.1 µg/mL, 71% at 1 µg/mL, and 69% at 5 µg/mL. In mouse cytokine array measuring 40 cytokines, the productions of interleukin-16, TIMP-1, and tumor necrosis factor-α (TNF-α) were significantly reduced with 0.1 µg/mL of rivaroxaban pretreatment (all P < 0.05). TIMP-1 levels in the culture supernatant measured by ELISA were also decreased by rivaroxaban pretreatment in Ang II-induced CFs (35% decrease at 0.01 µg/mL, 47% at 0.1 µg/mL, 47% at 1 µg/mL, and 57% at 5 µg/mL). In the dual reporter assay analysis, rivaroxaban inhibited various inflammatory signal pathways, including the nuclear factor-kappa B (NF-κB), active protein-1 (AP-1), and mitogen-activated protein kinase (MAPK) pathways (decreases of 82%, 78%, and 75%, respectively).These data suggest that rivaroxaban inhibits Ang II-induced functional activation in cultured mouse CFs via inhibiting NF-κB and MAPK/AP-1 signaling pathways, which may be a possible target of heart failure, through the antifibrotic and anti-inflammatory efficacy of rivaroxaban in Ang II-stimulated cardiac fibroblasts.


Subject(s)
Cell Movement/drug effects , Fibroblasts , Fibrosis , Myocardium/metabolism , NF-kappa B/metabolism , Rivaroxaban , Angiotensin II/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Factor Xa Inhibitors/metabolism , Factor Xa Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis/etiology , Fibrosis/metabolism , Mice , Receptor, PAR-2 , Rivaroxaban/metabolism , Rivaroxaban/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...