Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.175
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS One ; 19(8): e0309125, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39190751

RESUMEN

Increasing evidence is implicating roles for platelets in the development and progression of ovarian cancer, a highly lethal disease that can arise from the fallopian tubes, and has no current method of early detection or prevention. Thrombosis is a major cause of mortality of ovarian cancer patients suggesting that the cancer alters platelet behavior. The objective of this study was to develop a cell culture model of the pathological interactions of human platelets and ovarian cancer cells, using normal FT epithelial cells as a healthy control, and to test effects of the anti-platelet dihomo-gamma-linolenic acid (DGLA) in the model. Both healthy and cancer cells caused platelet aggregation, however platelets only affected spheroid formation by cancer cells and had no effect on healthy cell spheroid formation. When naturally-formed spheroids of epithelial cells were exposed to platelets in transwell inserts that did not allow direct interactions of the two cell types, platelets caused increased size of the spheroids formed by cancer cells, but not healthy cells. When cancer cell spheroids formed using magnetic nanoshuttle technology were put in direct physical contact with platelets, the platelets caused spheroid condensation. In ovarian cancer cells, DGLA promoted epithelial-to-mesenchymal (EMT) transition at doses as low as 100 µM, and inhibited metabolic viability and induced apoptosis at doses ≥150 µM. DGLA doses ≤150 µM used to avoid direct DGLA effects on cancer cells, had no effect on the pathological interactions of platelets and ovarian cancer cells in our models. These results demonstrate that the pathological interactions of platelets with ovarian cancer cells can be modeled in cell culture, and that DGLA has no effect on these interactions, suggesting that targeting platelets is a rational approach for reducing cancer aggressiveness and thrombosis risk in ovarian cancer patients, however DGLA is not an appropriate candidate for this strategy.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico , Plaquetas , Células Epiteliales , Neoplasias Ováricas , Esferoides Celulares , Humanos , Plaquetas/efectos de los fármacos , Femenino , Células Epiteliales/efectos de los fármacos , Neoplasias Ováricas/patología , Ácido 8,11,14-Eicosatrienoico/farmacología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Esferoides Celulares/efectos de los fármacos , Línea Celular Tumoral , Agregación Plaquetaria/efectos de los fármacos , Técnicas de Cultivo de Célula/métodos , Comunicación Celular/efectos de los fármacos
2.
J Chem Inf Model ; 64(16): 6506-6520, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39109515

RESUMEN

Thrombocytopenia, which is associated with thrombopoietin (TPO) deficiency, presents very limited treatment options and can lead to life-threatening complications. Discovering new therapeutic agents against thrombocytopenia has proven to be a challenging task using traditional screening approaches. Fortunately, machine learning (ML) techniques offer a rapid avenue for exploring chemical space, thereby increasing the likelihood of uncovering new drug candidates. In this study, we focused on computational modeling for drug-induced megakaryocyte differentiation and platelet production using ML methods, aiming to gain insights into the structural characteristics of hematopoietic activity. We developed 112 different classifiers by combining eight ML algorithms with 14 molecule features. The top-performing model achieved good results on both 5-fold cross-validation (with an accuracy of 81.6% and MCC value of 0.589) and external validation (with an accuracy of 83.1% and MCC value of 0.642). Additionally, by leveraging the Shapley additive explanations method, the best model provided quantitative assessments of molecular properties and structures that significantly contributed to the predictions. Furthermore, we employed an ensemble strategy to integrate predictions from multiple models and performed in silico predictions for new molecules with potential activity against thrombocytopenia, sourced from traditional Chinese medicine and the Drug Repurposing Hub. The findings of this study could offer valuable insights into the structural characteristics and computational prediction of thrombopoiesis inducers.


Asunto(s)
Aprendizaje Automático , Trombocitopenia , Trombocitopenia/tratamiento farmacológico , Humanos , Descubrimiento de Drogas/métodos , Megacariocitos/metabolismo , Megacariocitos/efectos de los fármacos , Megacariocitos/citología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Simulación por Computador , Algoritmos
3.
Respir Res ; 25(1): 311, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39154188

RESUMEN

BACKGROUND: Tea polyphenols (TPs), prominent constituents of green tea, possess remarkable antioxidant and anti-inflammatory properties. However, their therapeutic potential is limited due to low absorption and poor bioavailability. To address this limitation and enhance their efficacy, we developed a biomimetic nanoplatform by coating platelet membrane (PM) onto poly-lactic-co-glycolic acid (PLGA) nanoparticles (NPs) to create targeted delivery vehicles for TPs (PM@TP/NPs) to the inflamed tissues in asthma. METHODS: After synthesizing and characterizing PM@TP/NPs, we assessed their biocompatibility and biosafety through cell viability assays, hemolysis tests, and inflammation analysis in vivo and in vitro. The therapeutic effect of PM@TP/NPs on asthma was then evaluated using a mouse model of HDM-induced asthma. Additionally, PM@TP/NPs-mediated reactive oxygen species (ROS) scavenging capacity, as well as the activation of signaling pathways, were analyzed in HBE cells and asthmatic mice via flow cytometry, RT-qPCR, and western blotting. RESULTS: Compared with free TPs, PM@TP/NPs demonstrated excellent biocompatibility and safety profiles in both in vitro and in vivo, as well as enhanced retention in inflamed lungs. In HDM-induced mouse asthma model, inhaled PM@TP/NPs largely attenuated lung inflammation and reduced the secretion of type 2 pro-inflammatory cytokines in the lungs compared to free TPs. The therapeutic effects of PM@TP/NPs on asthma might be associated with an enhanced ROS scavenging capacity, increased activation of the Nrf2/HO-1 pathway, and decreased activation of the CCL2/MAPK and TLR4/NF-κB pathway in the lungs. CONCLUSIONS: Our findings demonstrate that inhalation of PM@TP/NPs largely attenuated lung inflammation in HDM-induced asthmatic mice. These results suggest that PM@TP/NPs might be a novel therapeutic strategy for asthma.


Asunto(s)
Asma , Plaquetas , Nanopartículas , Polifenoles , , Animales , Ratones , Polifenoles/administración & dosificación , Polifenoles/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Nanopartículas/administración & dosificación , Té/química , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Administración por Inhalación , Humanos , Ratones Endogámicos BALB C , Femenino , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología
4.
Int J Nanomedicine ; 19: 8253-8270, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39157734

RESUMEN

Background: Myocardial infarction (MI) is characterized by irreversible cardiomyocyte death resulting from an inadequate supply of oxygenated blood to the myocardium. Recent studies have indicated that ferroptosis, a form of regulated cell death, exacerbates myocardial injury during MI. Concurrently, the upregulation of CD47 on the surface of damaged myocardium following MI impairs the clearance of dead cells by macrophages, thereby hindering efferocytosis. In this context, simultaneously inhibiting ferroptosis and enhancing efferocytosis may represent a promising strategy to mitigate myocardial damage post-MI. Methods: In this study, we engineered platelet membrane-coated hollow mesoporous silicon nanoparticles (HMSN) to serve as a drug delivery system, encapsulating ferroptosis inhibitor, Ferrostatin-1, along with an anti-CD47 antibody. We aimed to assess the potential of these nanoparticles (designated as Fer-aCD47@PHMSN) to specifically target the site of MI and evaluate their efficacy in reducing cardiomyocyte death and inflammation. Results: The platelet membrane coating on the nanoparticles significantly enhanced their ability to successfully target the site of myocardial infarction (MI). Our findings demonstrate that treatment with Fer-aCD47@PHMSN resulted in a 38.5% reduction in cardiomyocyte ferroptosis under hypoxia, indicated by decreased lipid peroxidation and increased in vitro. Additionally, Fer-aCD47@PHMSN improved cardiomyocyte efferocytosis by approximately 15% in vitro. In MI mice treated with Fer-aCD47@PHMSN, we observed a substantial reduction in cardiomyocyte death (nearly 30%), decreased inflammation, and significant improvement in cardiac function. Conclusion: Our results demonstrated that the cooperation between the two agents induced anti-ferroptosis effects and enhanced dead cardiomyocyte clearance by macrophage as well as anti-inflammation effects. Thus, our nanoparticle Fer-aCD47@PHMSN provides a new therapeutic strategy for targeted therapy of MI.


Asunto(s)
Antígeno CD47 , Ferroptosis , Infarto del Miocardio , Miocitos Cardíacos , Nanopartículas , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Ferroptosis/efectos de los fármacos , Animales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Nanopartículas/química , Ratones , Antígeno CD47/metabolismo , Fagocitosis/efectos de los fármacos , Ciclohexilaminas/farmacología , Ciclohexilaminas/química , Masculino , Fenilendiaminas/farmacología , Fenilendiaminas/química , Macrófagos/efectos de los fármacos , Plaquetas/efectos de los fármacos , Ratones Endogámicos C57BL , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Portadores de Fármacos/química , Humanos , Eferocitosis
5.
Eur J Med Res ; 29(1): 420, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143607

RESUMEN

BACKGROUND: It is well-established that thrombus aspiration during primary percutaneous coronary intervention (PCI) in patients with acute myocardial infarction (AMI) indicates a higher thrombus burden and necessitates more intensive antithrombotic therapy. The bidirectional association between adverse events in AMI patients and platelet reactivity is typically observed during dual antiplatelet therapy (DAPT). OBJECTIVE: To investigate platelet reactivity after DAPT in AMI patients with thrombus aspiration performed during PCI. METHODS: In this retrospective study, we examined 269 consecutive AMI patients who underwent PCI and recorded their demographic, clinical and laboratory data. The platelet reactivity was measured with thromboelastogram (TEM). RESULTS: Ultimately, 208 patients were included in this study and divided into a Thrombus Aspiration group (N = 97) and a PCI Alone group (N = 111) based on whether thrombus aspiration was performed or not. The adenosine diphosphate (ADP)-induced platelet inhibition rate in the Thrombus Aspiration group was higher than that in the PCI Alone group (P < 0.001). Furthermore, multivariate linear regression analysis revealed that the ADP-induced platelet inhibition rate was independently associated with leukocyte count, thrombus aspiration and the combination of aspirin and ticagrelor as DAPT after adjusting for potential covariates in all AMI patients. CONCLUSION: In conclusion, clinicians should exercise heightened attention towards the bleeding risk among patients undergoing PCI concomitant with Thrombus Aspiration postoperatively.


Asunto(s)
Infarto del Miocardio , Intervención Coronaria Percutánea , Inhibidores de Agregación Plaquetaria , Humanos , Masculino , Femenino , Inhibidores de Agregación Plaquetaria/uso terapéutico , Inhibidores de Agregación Plaquetaria/efectos adversos , Persona de Mediana Edad , Infarto del Miocardio/complicaciones , Anciano , Intervención Coronaria Percutánea/métodos , Intervención Coronaria Percutánea/efectos adversos , Estudios Retrospectivos , Terapia Antiplaquetaria Doble/métodos , Aspirina/uso terapéutico , Aspirina/administración & dosificación , Trombosis/etiología , Trombosis/prevención & control , Plaquetas/efectos de los fármacos , Trombectomía/métodos
6.
ACS Appl Mater Interfaces ; 16(34): 44528-44537, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39155662

RESUMEN

The high level of accumulation of therapeutic agents in tumors is crucial for cancer treatment. Compared to the passive tumor-targeting effect, active tumor-targeting delivery systems, primarily mediated by peptides with high production costs and reduced circulation time, are highly desired. Platelet-driven technologies have opened new avenues for targeted drug delivery prevalently through a membrane coating strategy that involves intricate manufacturing procedures or the fucoidan-mediated hitchhiking method with limited platelet affinity. Here, a novel type of amphiphilic glycopolymer self-assembled micellar nanoparticle has been developed to adhere to naturally activated platelets in the blood. The simultaneous integration of fucose and sialic acid segments into glycopolymers enables closer mimicry of the structure of P-selectin glycoprotein ligand-1 (PSGL-1), thereby increasing the affinity for activated platelets. It results in the formation of glycopolymeric micelle-platelet hybrids, facilitating targeted drug delivery to tumors. The selective platelet-assisted cellular uptake of docetaxel (DTX)-loaded glycopolymeric micelles leads to lower IC50 values against 4T1 cells than that of free DTX. The directed tumor-targeting effect of activated platelets has significantly improved the tumor accumulation capacity of the glycopolymeric nanoparticles, with up to 21.0% found in tumors within the initial 0.2 h. Additionally, with acid-responsive drug release and inherent antimetastasis properties, the glycopolymeric nanoparticles ensured potent therapeutic efficacy, prolonged survival time, and reduced cardiotoxicity, presenting a new and unexplored strategy for platelet-directed drug delivery to tumors, showing promising prospects in treating localized tumors and preventing tumor metastasis.


Asunto(s)
Plaquetas , Docetaxel , Micelas , Nanopartículas , Docetaxel/química , Docetaxel/farmacología , Docetaxel/farmacocinética , Docetaxel/uso terapéutico , Animales , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Nanopartículas/química , Ratones , Línea Celular Tumoral , Antineoplásicos/química , Antineoplásicos/farmacología , Ratones Endogámicos BALB C , Humanos , Femenino , Sistemas de Liberación de Medicamentos , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Neoplasias/metabolismo
7.
Int J Mol Sci ; 25(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39201591

RESUMEN

Caffeine is the most popular and widely consumed behaviourally active substance in the world. This review describes the influence of caffeine on the cardiovascular system, with a special focus on blood platelets. For many years, caffeine was thought to have a negative effect on the cardiovascular system mainly due to increasing blood pressure. However, more recent data suggest that habitual caffeine consumption may reduce the risk of cardiovascular disease and hypertension. This could be a significant finding as cardiovascular disease is the leading cause of death worldwide. Caffeine is known to inhibit A1 adenosine receptors, through which it is believed to modulate inter alia coronary blood flow, total peripheral resistance, diuresis, and heart rate. It has been shown that coffee possesses antiplatelet activity, but depending on the dose and the term of its use, caffeine may stimulate or inhibit platelet reactivity. Also, chronic exposure to caffeine may sensitize or upregulate the adenosine receptors in platelets causing increased cAMP accumulation and anti-aggregatory effects and decrease calcium levels elicited by AR agonists. The search for new, selective, and safe AR agonists is one of the new strategies for improving antiplatelet therapy involving targeting multiple pathways of platelet activation. Therefore, this review examines the AR-dependent impact of caffeine on blood platelets in the presence of adenosine receptor agonists.


Asunto(s)
Plaquetas , Cafeína , Receptores Purinérgicos P1 , Humanos , Cafeína/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Receptores Purinérgicos P1/metabolismo , Animales , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/metabolismo , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Activación Plaquetaria/efectos de los fármacos
8.
Thromb Res ; 241: 109111, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39098189

RESUMEN

Thrombosis is the primary cause of death in patients with cancer. Resveratrol inhibits platelet activation, a crucial pathophysiological basis of thrombosis, in healthy individuals. However, its effects and mechanisms of action in patients with colon cancer remain unknown. Here, we investigated the effect of resveratrol on platelet adhesion and aggregation in patients with colon cancer. Through numerous in vitro and in vivo analyses, including flow cytometry, western blotting, ELISA, and immunofluorescence and colon cancer rat models, we demonstrated that resveratrol reduced thrombosis in patients with colon cancer by inhibiting the phosphorylation of the MAPK and activating the cyclic-GMP/vasodilator-stimulated phosphoprotein pathway. These findings demonstrate the potential of resveratrol in reducing thrombosis in patients with colon cancer and could be used to develop novel therapeutic strategies for this condition.


Asunto(s)
Moléculas de Adhesión Celular , Neoplasias del Colon , GMP Cíclico , Fosfoproteínas , Activación Plaquetaria , Resveratrol , Trombosis , Resveratrol/farmacología , Resveratrol/uso terapéutico , Trombosis/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/complicaciones , Humanos , Animales , Activación Plaquetaria/efectos de los fármacos , Masculino , Ratas , GMP Cíclico/metabolismo , Moléculas de Adhesión Celular/metabolismo , Fosfoproteínas/metabolismo , Femenino , Estilbenos/farmacología , Estilbenos/uso terapéutico , Ratas Sprague-Dawley , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Proteínas de Microfilamentos
9.
Int J Mol Sci ; 25(14)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39062849

RESUMEN

A key step in platelet production is the migration of megakaryocytes to the vascular sinusoids within the bone marrow. This homing is mediated by the chemokine CXCL12 and its receptor CXCR4. CXCR4 is also a positive regulator of platelet activation and thrombosis. Pim-1 kinase has been shown to regulate CXCR4 signalling in other cell types, and we have previously described how Pim kinase inhibitors attenuate platelet aggregation to CXCL12. However, the mechanism by which Pim-1 regulates CXCR4 signalling in platelets and megakaryocytes has yet to be elucidated. Using human platelets, murine bone marrow-derived megakaryocytes, and the megakaryocyte cell line MEG-01, we demonstrate that pharmacological Pim kinase inhibition leads to reduced megakaryocyte and platelet function responses to CXCL12, including reduced megakaryocyte migration and platelet granule secretion. Attenuation of CXCL12 signalling was found to be attributed to the reduced surface expression of CXCR4. The decrease in CXCR4 surface levels was found to be mediated by rapid receptor internalisation, in the absence of agonist stimulation. We demonstrate that pharmacological Pim kinase inhibition disrupts megakaryocyte and platelet function by reducing constitutive CXCR4 surface expression, decreasing the number of receptors available for agonist stimulation and signalling. These findings have implications for the development and use of Pim kinase inhibitors for the treatment of conditions associated with elevated circulating levels of CXCL12/SDF1α and increased thrombotic risk.


Asunto(s)
Plaquetas , Quimiocina CXCL12 , Megacariocitos , Proteínas Proto-Oncogénicas c-pim-1 , Receptores CXCR4 , Transducción de Señal , Receptores CXCR4/metabolismo , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Megacariocitos/metabolismo , Megacariocitos/efectos de los fármacos , Megacariocitos/citología , Humanos , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Movimiento Celular/efectos de los fármacos , Línea Celular
10.
Int J Mol Sci ; 25(14)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39062819

RESUMEN

Platelets play a significant role in hemostasis, forming plugs at sites of vascular injury to limit blood loss. However, if platelet activation is not controlled, it can lead to thrombotic events, such as myocardial infarction and stroke. To prevent this, antiplatelet agents are used in clinical settings to limit platelet activation in patients at risk of arterial thrombotic events. However, their use can be associated with a significant risk of bleeding. An enhanced comprehension of platelet signaling mechanisms should facilitate the identification of safer targets for antiplatelet therapy. Over the past decade, our comprehension of the breadth and intricacy of signaling pathways that orchestrate platelet activation has expanded exponentially. Several recent studies have provided further insight into the regulation of platelet signaling events and identified novel targets against which to develop novel antiplatelet agents. Antiplatelet drugs are essential in managing atherothrombotic vascular disease. The current antiplatelet therapy in clinical practice is limited in terms of safety and efficacy. Novel compounds have been developed in response to patient variability and resistance to aspirin and/or clopidogrel. Recent studies based on randomized controlled trials and systematic reviews have definitively demonstrated the role of antiplatelet therapy in reducing the risk of cardiovascular events. Antiplatelet therapy is the recommended course of action for patients with established atherosclerosis. These studies compared monotherapy with a P2Y12 inhibitor versus aspirin for secondary prevention. However, in patients undergoing percutaneous coronary intervention, it is still unclear whether the efficacy of P2Y12 inhibitor monotherapy after a short course of dual antiplatelet therapy depends on the type of P2Y12 inhibitor. This paper focuses on the advanced-stage evaluation of several promising antiplatelet drugs.


Asunto(s)
Inhibidores de Agregación Plaquetaria , Antagonistas del Receptor Purinérgico P2Y , Humanos , Antagonistas del Receptor Purinérgico P2Y/uso terapéutico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Inhibidores de Agregación Plaquetaria/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Animales
11.
Biomed Pharmacother ; 177: 117131, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39013224

RESUMEN

Drug delivery systems are becoming increasingly utilized; however, a major challenge in this field is the insufficient target of tissues or cells. Although efforts with engineered nanoparticles have shown some success, issues with targeting, toxicity and immunogenicity persist. Conversely, living cells can be used as drug-delivery vehicles because they typically have innate targeting mechanisms and minimal adverse effects. As active participants in hemostasis, inflammation, and tumors, platelets have shown great potential in drug delivery. This review highlights platelet-based drug delivery systems, including platelet membrane engineering, platelet membrane coating, platelet cytoplasmic drug loading, genetic engineering, and synthetic/artificial platelets for different applications.


Asunto(s)
Plaquetas , Sistemas de Liberación de Medicamentos , Humanos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Animales , Nanopartículas , Ingeniería Genética/métodos , Portadores de Fármacos/química
12.
Cells ; 13(13)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38994957

RESUMEN

Platelet activation is critical for haemostasis, but if unregulated can lead to pathological thrombosis. Endogenous platelet inhibitory mechanisms are mediated by prostacyclin (PGI2)-stimulated cAMP signalling, which is regulated by phosphodiesterase 3A (PDE3A). However, spatiotemporal regulation of PDE3A activity in platelets is unknown. Here, we report that platelets possess multiple PDE3A isoforms with seemingly identical molecular weights (100 kDa). One isoform contained a unique N-terminal sequence that corresponded to PDE3A1 in nucleated cells but with negligible contribution to overall PDE3A activity. The predominant cytosolic PDE3A isoform did not possess the unique N-terminal sequence and accounted for >99% of basal PDE3A activity. PGI2 treatment induced a dose and time-dependent increase in PDE3A phosphorylation which was PKA-dependent and associated with an increase in phosphodiesterase enzymatic activity. The effects of PGI2 on PDE3A were modulated by A-kinase anchoring protein (AKAP) disruptor peptides, suggesting an AKAP-mediated PDE3A signalosome. We identified AKAP7, AKAP9, AKAP12, AKAP13, and moesin expressed in platelets but focussed on AKAP7 as a potential PDE3A binding partner. Using a combination of immunoprecipitation, proximity ligation techniques, and activity assays, we identified a novel PDE3A/PKA RII/AKAP7 signalosome in platelets that integrates propagation and termination of cAMP signalling through coupling of PKA and PDE3A.


Asunto(s)
Proteínas de Anclaje a la Quinasa A , Plaquetas , Proteínas Quinasas Dependientes de AMP Cíclico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3 , Epoprostenol , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Humanos , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Epoprostenol/metabolismo , Epoprostenol/farmacología , Fosforilación , AMP Cíclico/metabolismo , Transducción de Señal
13.
Nutrients ; 16(13)2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38999771

RESUMEN

The study aimed to evaluate the antithrombotic action of Acrocomia aculeata pulp oil (AAPO) in natura, in an in vitro experimental model. AAPO was obtained by solvent extraction, and its chemical characterization was performed by gas chromatography coupled to a mass spectrometer (GC-MS). In vitro toxicity was evaluated with the Trypan Blue exclusion test and in vivo by the Galleria mellonella model. ADP/epinephrine-induced platelet aggregation after treatment with AAPO (50, 100, 200, 400, and 800 µg/mL) was evaluated by turbidimetry, and coagulation was determined by prothrombin activity time (PT) and activated partial thromboplastin time (aPTT). Platelet activation was measured by expression of P-selectin on the platelet surface by flow cytometry and intraplatelet content of reactive oxygen species (ROS) by fluorimetry. The results showed that AAPO has as major components such as oleic acid, palmitic acid, lauric acid, caprylic acid, and squalene. AAPO showed no toxicity in vitro or in vivo. Platelet aggregation decreased against agonists using treatment with different concentrations of AAPO. Oil did not interfere in PT and aPTT. Moreover, it expressively decreased ROS-induced platelet activation and P-selectin expression. Therefore, AAPO showed antiplatelet action since it decreased platelet activation verified by the decrease in P-selectin expression as well as in ROS production.


Asunto(s)
Fibrinolíticos , Selectina-P , Aceites de Plantas , Agregación Plaquetaria , Especies Reactivas de Oxígeno , Animales , Agregación Plaquetaria/efectos de los fármacos , Selectina-P/metabolismo , Humanos , Aceites de Plantas/farmacología , Aceites de Plantas/química , Especies Reactivas de Oxígeno/metabolismo , Fibrinolíticos/farmacología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Coagulación Sanguínea/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos
14.
Thromb Res ; 241: 109092, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39024901

RESUMEN

BACKGROUND: Leukocyte-platelet aggregates comprise a pathogenic link between hemostasis and immunity, but the prerequisites and mechanisms of their formation remain not understood. AIMS: To quantify the formation, composition, and morphology of leukocyte-platelet aggregates in vitro under the influence of various cellular activators. METHODS: Phorbol-12-myristate-13-acetate (PMA), lipopolysaccharide (LPS), thrombin receptor-activating peptide (TRAP-6), and adenosine diphosphate (ADP) were used as cellular activators. Flow cytometry was utilized to identify and quantify aggregates in whole human blood and platelet-rich plasma. Cell types and cellular aggregates were identified using fluorescently labeled antibodies against the appropriate cellular markers, and cell activation was assessed by the expression of appropriate surface markers. For confocal fluorescent microscopy, cell membranes and nuclei were labeled. Neutrophil-platelet aggregates were studied using scanning electron microscopy. RESULTS: In the presence of PMA, ADP or TRAP-6, about 17-38 % of neutrophils and 61-77 % of monocytes formed aggregates with platelets in whole blood, whereas LPS did not induce platelet aggregation with either neutrophils or monocytes due the inability to activate platelets. Similar results were obtained when isolated neutrophils were added to platelet-rich plasma. All the cell types involved in the heterotypic aggregation expressed molecular markers of activation. Fluorescent and electron microscopy of the aggregates showed that the predominant platelet/leukocyte ratios were 1:1 and 2:1. CONCLUSIONS: Formation of leukocyte-platelet aggregates depends on the nature of the cellular activator and the spectrum of its cell-activating ability. An indispensable condition for formation of leukocyte-platelet aggregates is activation of all cell types including platelets, which is the restrictive step.


Asunto(s)
Plaquetas , Leucocitos , Lipopolisacáridos , Agregación Plaquetaria , Acetato de Tetradecanoilforbol , Humanos , Plaquetas/metabolismo , Plaquetas/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Agregación Plaquetaria/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Lipopolisacáridos/farmacología , Adenosina Difosfato/farmacología , Adenosina Difosfato/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fragmentos de Péptidos/farmacología , Citometría de Flujo , Monocitos/efectos de los fármacos , Monocitos/metabolismo
15.
Int Immunopharmacol ; 139: 112654, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-38996777

RESUMEN

Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Ligando de CD40 , Diminazeno , Modelos Animales de Enfermedad , Hipertensión , Peptidil-Dipeptidasa A , Transducción de Señal , Animales , Diminazeno/análogos & derivados , Diminazeno/farmacología , Diminazeno/uso terapéutico , Enzima Convertidora de Angiotensina 2/metabolismo , Masculino , Transducción de Señal/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Ligando de CD40/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Ratas , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proto-Oncogenes Mas , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Fragmentos de Péptidos , Angiotensina I , Células Cultivadas , Microglía/efectos de los fármacos , Microglía/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ratas Wistar , Sistema Renina-Angiotensina/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Antígenos CD40/metabolismo , Humanos , Activación Plaquetaria/efectos de los fármacos , Antihipertensivos/farmacología , Antihipertensivos/uso terapéutico
16.
Am J Vet Res ; 85(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38889754

RESUMEN

OBJECTIVE: To evaluate the effects of darbepoetin on platelet population and reactivity in healthy cats (HCs) and azotemic cats with remnant kidney (RK) model-induced chronic kidney disease. ANIMALS: 12 purpose-bred domestic shorthair cats (n = 6 HCs and n = 6 RK). METHODS: In this pilot study, all cats received darbepoetin (1 µg/kg, SC) on days 0, 7, and 14. Blood was sampled at baseline and on days 3, 10, 15, 17, 20, and 21. At each time point, a CBC was performed, platelet aggregometry was assessed by impedance and optical methods, and platelet P-selectin (CD62P) was quantified before and after thrombin stimulation. Additionally, reticulated platelets were quantified using both thiazole orange staining and proprietary analysis by the CBC analyzer. For RK cats, systemic blood pressure (BP) was serially measured. RESULTS: No adverse effects of darbepoetin were seen. There was no statistically significant change in platelet count between or within groups at any time point. Hematocrit increased significantly over time in the RK but not the HC group. RBC reticulocyte numbers in both groups increased over time. Reticulated platelet percentage did not increase in either group. Differences in platelet reactivity within or between groups were not seen in the aggregometry or flow cytometric assessments. In RK cats, indirect BP did not significantly change during the study. CLINICAL RELEVANCE: This preliminary investigation did not find evidence that darbepoetin administration impacted platelet number, reactivity, nor reticulated platelet count. Anemic RK cats experienced increased hematocrit and RBC reticulocytes as expected with darbepoetin therapy.


Asunto(s)
Enfermedades de los Gatos , Darbepoetina alfa , Hematínicos , Insuficiencia Renal Crónica , Animales , Gatos , Darbepoetina alfa/farmacología , Darbepoetina alfa/uso terapéutico , Enfermedades de los Gatos/tratamiento farmacológico , Insuficiencia Renal Crónica/veterinaria , Insuficiencia Renal Crónica/sangre , Recuento de Plaquetas/veterinaria , Proyectos Piloto , Masculino , Hematínicos/farmacología , Hematínicos/uso terapéutico , Femenino , Plaquetas/efectos de los fármacos , Hematócrito/veterinaria
17.
J Ethnopharmacol ; 333: 118485, 2024 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-38908490

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Xuefu Zhuyu Decoction (XZD), a renowned traditional Chinese medicine prescription, is widely employed for the management of conditions characterized by qi-stagnation and blood stasis. Although its anti-thrombotic effect on deep vein thrombosis (DVT) patients has been clinically observed, the underlying mechanism remains largely unexplored. AIM OF THE STUDY: Our aim was to investigate the mechanisms by which XZD exerted its effect on DVT. MATERIALS AND METHODS: The ultra performance liquid chromatography (UPLC) technique was employed to evaluate quality of XZD. To examine the effect of XZD on DVT, a DVT rat model with inferior vena cava (IVC) stenosis was established. The 4D-label-free proteomics approach was then utilized to uncover the possible mechanisms of XZD against DVT. Based on proteomics, citrullinated histone H3 (CitH3), along with serum levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1ß) were observed the inhibitory activity of XZD on neutrophil activation. Subsequently, the marker of platelet activation, specifically glycoprotein IIb (CD41) and glycoprotein IIIa (CD61), were assessed along with the secretion of von Willebrand factor (vWF) to investigate the inhibitory activity of XZD on platelet activation. Finally, we explored the impact of XZD on the sirtuin 1 (SIRT1)/nuclear factor kappa-B (NF-κB) pathway, which was associated with the activation of platelets and neutrophils. RESULTS: Eight distinct components were identified for the quality control of XZD. XZD effectively reduced thrombus weight and length in DVT rats, without affecting the coagulation function or hematological parameters in the systemic circulation. Proteomics analysis revealed that XZD alleviated DVT by inhibiting the activation of platelets and neutrophils. The protein expression of CitH3, along with serum levels of TNF-α and IL-1ß, were reduced in XZD-treated DVT rats. Similarly, protein expressions of CD41 and CD61, along with the release of vWF, were markedly down-regulated in XZD-treated DVT rats. Finally, treatment with XZD resulted in an up-regulation of SIRT1 protein expression and a down-regulation of both acetylated NF-κB/p65 and phosphorylated NF-κB/p65 protein expressions in endothelium. CONCLUSIONS: XZD alleviates DVT by inhibiting the activation of platelets and neutrophils at the injured endothelium via the regulation of SIRT1/NF-κB pathway.


Asunto(s)
Plaquetas , Medicamentos Herbarios Chinos , Neutrófilos , Activación Plaquetaria , Transducción de Señal , Trombosis de la Vena , Animales , Masculino , Ratas , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , FN-kappa B/metabolismo , Activación Plaquetaria/efectos de los fármacos , Proteómica , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Trombosis de la Vena/tratamiento farmacológico
18.
Biomolecules ; 14(6)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38927016

RESUMEN

Platelets play essential roles in the formation of blood clots by clumping with coagulation factors at the site of vascular injury to stop bleeding; therefore, a reduction in the platelet number or disorder in their function causes bleeding risk. In our research, we developed a method to assess platelet aggregation using an optical approach within a microfluidic chip's channel by evaluating the size of laser speckles. These speckles, associated with slowed blood flow in the microfluidic channel, had a baseline size of 28.54 ± 0.72 µm in whole blood. Removing platelets from the sample led to a notable decrease in speckle size to 27.04 ± 1.23 µm. Moreover, the addition of an ADP-containing agonist, which activates platelets, resulted in an increased speckle size of 32.89 ± 1.69 µm. This finding may provide a simple optical method via microfluidics that could be utilized to assess platelet functionality in diagnosing bleeding disorders and potentially in monitoring therapies that target platelets.


Asunto(s)
Plaquetas , Agregación Plaquetaria , Plaquetas/efectos de los fármacos , Humanos , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria/métodos , Pruebas de Función Plaquetaria/instrumentación , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Microfluídica/métodos , Adenosina Difosfato/farmacología
20.
Mol Pharm ; 21(7): 3577-3590, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38857525

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by high malignancy and limited treatment options. Given the pressing need for more effective treatments for TNBC, this study aimed to develop platelet membrane (PM)-camouflaged silver metal-organic framework nanoparticles (PM@MOF-Ag NPs), a biomimetic nanodrug. PM@MOF-Ag NP construction involved the utilization of 2-methylimidazole and silver nitrate to prepare silver metal-organic framework (MOF-Ag) NPs. The PM@MOF-Ag NPs, due to their camouflage, possess excellent blood compatibility, immune escape ability, and a strong affinity for 4T1 tumor cells. This enhances their circulation time in vivo and promotes the aggregation of PM@MOF-Ag NPs at the 4T1 tumor site. Importantly, PM@MOF-Ag NPs demonstrated promising antitumor activity in vitro and in vivo. We further revealed that PM@MOF-Ag NPs induced tumor cell death by overproducing reactive oxygen species and promoting cell apoptosis. Moreover, PM@MOF-Ag NPs enhanced apoptosis by upregulating the ratios of Bax/Bcl-2 and cleaved caspase3/pro-caspase3. Notably, PM@MOF-Ag NPs exhibited no significant organ toxicity, whereas the administration of MOF-Ag NPs resulted in liver inflammation compared to the control group.


Asunto(s)
Apoptosis , Nanopartículas del Metal , Estructuras Metalorgánicas , Especies Reactivas de Oxígeno , Plata , Neoplasias de la Mama Triple Negativas , Estructuras Metalorgánicas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Animales , Femenino , Plata/química , Ratones , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Nanopartículas del Metal/química , Especies Reactivas de Oxígeno/metabolismo , Humanos , Ratones Endogámicos BALB C , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Biomimética/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Nanopartículas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA