Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Cell Mol Med ; 25(19): 9434-9446, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34448532

RESUMEN

Naphthalimide derivatives have multiple biological activities, including antitumour and anti-inflammatory activities. We previously synthesized several naphthalimide derivatives; of them, compound 5 was found to exert the strongest inhibitory effect on human DNA topoisomerase II activity. However, the effects of naphthalimide derivatives on platelet activation have not yet been investigated. Therefore, the mechanism underlying the antiplatelet activity of compound 5 was determined in this study. The data revealed that compound 5 (5-10 µM) inhibited collagen- and convulxin- but not thrombin- or U46619-mediated platelet aggregation, suggesting that compound 5 is more sensitive to the inhibition of glycoprotein VI (GPVI) signalling. Indeed, compound 5 could inhibit the phosphorylation of signalling molecules downstream of GPVI, followed by the inhibition of calcium mobilization, granule release and GPIIb/IIIa activation. Moreover, compound 5 prevented pulmonary embolism and prolonged the occlusion time, but tended to prolong the bleeding time, indicating that it can prevent thrombus formation but may increase bleeding risk. This study is the first to demonstrate that the naphthalimide derivative compound 5 exerts antiplatelet and antithrombotic effects. Future studies should modify compound 5 to synthesize more potent and efficient antiplatelet agents while minimizing bleeding risk, which may offer a therapeutic potential for cardiovascular diseases.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Naftalimidas/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombosis/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Inmunohistoquímica , Masculino , Ratones , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Estructura Molecular , Naftalimidas/química , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal , Trombosis/tratamiento farmacológico , Trombosis/etiología , Trombosis/patología
2.
Molecules ; 26(17)2021 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-34500771

RESUMEN

Vincristine is a clinically used antimicrotubule drug for treating patients with lymphoma. Due to its property of increasing platelet counts, vincristine is also used to treat patients with immune thrombocytopenia. Moreover, antiplatelet agents were reported to be beneficial in thrombotic thrombocytopenic purpura (TTP). Therefore, we investigated the detailed mechanisms underlying the antiplatelet effect of vincristine. Our results revealed that vincristine inhibited platelet aggregation induced by collagen, but not by thrombin, arachidonic acid, and the thromboxane A2 analog U46619, suggesting that vincristine exerts higher inhibitory effects on collagen-mediated platelet aggregation. Vincristine also reduced collagen-mediated platelet granule release and calcium mobilization. In addition, vincristine inhibited glycoprotein VI (GPVI) signaling, including Syk, phospholipase Cγ2, protein kinase C, Akt, and mitogen-activated protein kinases. In addition, the in vitro PFA-100 assay revealed that vincristine did not prolong the closure time, and the in vivo study tail bleeding assay showed that vincristine did not prolong the tail bleeding time; both findings suggested that vincristine may not affect normal hemostasis. In conclusion, we demonstrated that vincristine exerts antiplatelet effects at least in part through the suppression of GPVI signaling. Moreover, this property of antiplatelet activity of vincristine may provide additional benefits in the treatment of TTP.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Plaquetas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Trombocitopenia/tratamiento farmacológico , Vincristina/farmacología , Antineoplásicos Fitogénicos/química , Plaquetas/inmunología , Colágeno/antagonistas & inhibidores , Colágeno/farmacología , Humanos , Conformación Molecular , Neoplasias/inmunología , Agregación Plaquetaria/efectos de los fármacos , Trombocitopenia/inmunología , Vincristina/química
3.
Int J Mol Sci ; 21(18)2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32971863

RESUMEN

Previously, we reported that phospholipase D1 (PLD1) and PLD2 inhibition by selective PLD1 and PLD2 inhibitors could prevent platelet aggregation in humans, but not in mice. Moreover, only the PLD1 inhibitor, but not PLD2 inhibitor, could effectively prevent thrombus formation in mice, indicating that PLD might play different roles in platelet function in humans and mice. Although PLD1 and PLD2 were reported to be implicated in thrombotic events, the role of PLD in mice remains not completely clear. Here, we investigated the role of PLD1 and PLD2 in acute pulmonary thrombosis and transient middle cerebral artery occlusion-induced brain injury in mice. The data revealed that inhibition of PLD1, but not of PLD2, could partially prevent pulmonary thrombosis-induced death. Moreover, concurrent PLD1 and PLD2 inhibition could considerably increase survival rate. Likewise, inhibition of PLD1, but not PLD2, partially improved ischemic stroke and concurrent inhibition of PLD1, and PLD2 exhibited a relatively better protection against ischemic stroke, as evidenced by the infarct size, brain edema, modified neurological severity score, rotarod test, and the open field test. In conclusion, PLD1 might play a more important role than PLD2, and both PLD1 and PLD2 could act synergistically or have partially redundant functions in regulating thrombosis-relevant events.


Asunto(s)
Trombosis Intracraneal/enzimología , Accidente Cerebrovascular Isquémico/enzimología , Fosfolipasa D/metabolismo , Animales , Trombosis Intracraneal/patología , Accidente Cerebrovascular Isquémico/patología , Masculino , Ratones , Ratones Endogámicos ICR
4.
J Cell Mol Med ; 23(3): 1723-1734, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30548082

RESUMEN

Haemorrhagic shock and resuscitation (HS/R) may cause global ischaemia-reperfusion injury, which can result in systemic inflammation, multiorgan failure (particularly liver failure) and high mortality. Hinokitiol, a bioactive tropolone-related compound, exhibits antiplatelet and anti-inflammatory activities. Targeting inflammatory responses is a potential strategy for ameliorating hepatic injury during HS/R. Whether hinokitiol prevents hepatic injury during HS/R remains unclear. In the present study, we determined the role of hinokitiol following HS/R. The in vivo assays revealed that hinokitiol markedly attenuated HS/R-induced hepatic injury. Hinokitiol could inhibited NF-κB activation and IL-6 and TNF-α upregulation in liver tissues. Moreover, hinokitiol reduced caspase-3 activation, upregulated Bax and downregulated Bcl-2. These findings suggest that hinokitiol can ameliorate liver injury following HS/R, partly through suppression of inflammation and apoptosis. Furthermore, the in vitro data revealed that hinokitiol significantly reversed hypoxia/reoxygenation (H/R)-induced cell death and apoptosis in the primary hepatocytes. Hinokitiol prevented H/R-induced caspase-3 activation, PPAR cleavage, Bax overexpression and Bcl-2 downregulation. Moreover, hinokitiol attenuated H/R-stimulated NF-κB activation and reduced the levels of IL-6 and TNF-α mRNAs, suggesting that hinokitiol can protect hepatocytes from H/R injury. Collectively, our data suggest that hinokitiol attenuates liver injury following HS/R, partly through the inhibition of NF-κB activation.


Asunto(s)
Antiinfecciosos/farmacología , Apoptosis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Hígado/efectos de los fármacos , Monoterpenos/farmacología , Choque Hemorrágico/complicaciones , Tropolona/análogos & derivados , Animales , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Hígado/lesiones , Hígado/metabolismo , Hígado/patología , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Tropolona/farmacología
5.
Int J Mol Sci ; 20(22)2019 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-31717348

RESUMEN

Auraptene is the most abundant coumarin derivative from plants. The pharmacological value of this compound has been well demonstrated, especially in the prevention of cancer and neurodegenerative diseases. Platelet activation is a major factor contributing to arterial thrombosis. Thus, this study evaluated the influence of auraptene in platelet aggregation and thrombotic formation. Auraptene inhibited platelet aggregation in human platelets stimulated with collagen only. However, auraptene was not effective in inhibiting platelet aggregation stimulated with thrombin, arachidonic acid, and U46619. Auraptene also repressed ATP release, [Ca2+]i mobilization, and P-selectin expression. Moreover, it markedly blocked PAC-1 binding to integrin αIIbß3. However, it had no influence on properties related to integrin αIIbß3-mediated outside-in signaling, such as the adhesion number, spreading area of platelets, and fibrin clot retraction. Auraptene inhibited the phosphorylation of Lyn-Fyn-Syk, phospholipase Cγ2 (PLCγ2), protein kinase C (PKC), Akt, and mitogen-activated protein kinases (MAPKs; extracellular-signal-regulated kinase (ERK1/2), and c-Jun N-terminal kinase (JNK1/2), but not p38 MAPK). Neither SQ22536, an adenylate cyclase inhibitor, nor ODQ, a guanylate cyclase inhibitor, reversed the auraptene-mediated inhibition of platelet aggregation. Auraptene reduced mortality caused by adenosine diphosphate (ADP)-induced pulmonary thromboembolism. In conclusion, this study provides definite evidence that auraptene signifies a potential therapeutic agent for preventing thromboembolic disorders.


Asunto(s)
Cumarinas/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Embolia Pulmonar/tratamiento farmacológico , Embolia Pulmonar/mortalidad , Transducción de Señal , Adenosina Trifosfato/metabolismo , Animales , Plaquetas/efectos de los fármacos , Calcio/metabolismo , Cumarinas/química , Cumarinas/farmacología , Humanos , Ratones , Nucleótidos Cíclicos/metabolismo , Selectina-P/metabolismo , Fosforilación/efectos de los fármacos , Embolia Pulmonar/sangre , Transducción de Señal/efectos de los fármacos
6.
J Cell Mol Med ; 22(4): 2142-2152, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29369482

RESUMEN

During platelet activation, fibrinogen binds to its specific platelet receptor, integrin αIIb ß3 , thus completing the final common pathway for platelet aggregation. Norcantharidin (NCTD) is a promising anticancer agent in China from medicinal insect blister beetle. In this study, we provided the evidence to demonstrate NCTD (0.1-1.0 µM) possesses very powerful antiplatelet activity in human platelets; nevertheless, it had no effects on surface P-selectin expression and only slight inhibition on ATP-release reaction in activated platelets. Moreover, NCTD markedly hindered integrin αIIb ß3 activation by interfering with the binding of FITC-labelled PAC-1. It also markedly reduced the number of adherent platelets and the single platelet spreading area on immobilized fibrinogen as well as clot retraction. Additionally, NCTD attenuated phosphorylation of proteins such as integrin ß3 , Src and FAK in platelets spreading on immobilized fibrinogen. These results indicate that NCTD restricts integrin αIIb ß3 -mediated outside-in signalling in human platelets. Besides, NCTD substantially prolonged the closure time in human whole blood and increased the occlusion time of thrombotic platelet plug formation and prolonged the bleeding time in mice. In conclusion, NCTD has dual activities, it can be a chemotherapeutic agent for cancer treatment, and the other side it possesses powerful antiplatelet activity for treating thromboembolic disorders.


Asunto(s)
Antineoplásicos/farmacología , Plaquetas/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Fibrinógeno/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Adenosina Trifosfato/metabolismo , Adulto , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Adhesión Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Selectina-P/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Sustancias Protectoras/química , Sustancias Protectoras/farmacología , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Trombosis/patología
7.
Int J Mol Sci ; 18(7)2017 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-28704925

RESUMEN

Platelet activation is involved in cardiovascular diseases, such as atherosclerosis and ischemic stroke. Licochalcone A (LA), an active ingredient of licorice, exhibits multiple biological activities such as anti-oxidation and anti-inflammation. However, its role in platelet activation remains unclear. Therefore, the study investigated the antiplatelet mechanism of LA. Our data revealed that LA (2-10 µM) concentration dependently inhibited platelet aggregation induced by collagen, but not thrombin and U46619. LA markedly attenuated collagen-stimulated ATP release, P-selectin secretion, calcium mobilization, and GPIIbIIIa activation, but did not interfere with the collagen binding to platelets. Moreover, LA significantly reduced the activation of PLCγ2, PKC, Akt and MAPKs. Thus, LA attenuates platelet activation, possibly by inhibiting collagen receptor downstream signaling but not by blocking the collagen receptors. In addition, LA prevented adenosine diphosphate (ADP)-induced acute pulmonary thrombosis, fluorescein sodium-induced platelet thrombus formation, and middle cerebral artery occlusion/reperfusion-induced brain injury in mice, but did not affect normal hemostasis. This study demonstrated that LA effectively reduced platelet activation and thrombus formation, in part, through the inhibition of PLCγ2-PKC, Akt, and MAPK pathways, without the side effect of bleeding. These findings also indicate that LA may provide a safe and alternative therapeutic approach for preventing thromboembolic disorders such as stroke.


Asunto(s)
Chalconas/uso terapéutico , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfolipasa C gamma/metabolismo , Activación Plaquetaria/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombosis/metabolismo , Trombosis/prevención & control , Animales , Calcio/metabolismo , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Selectina-P/metabolismo , Agregación Plaquetaria/efectos de los fármacos
8.
J Biomed Sci ; 23: 26, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26887315

RESUMEN

BACKGROUND: Thrombus formation, a phenomenon primarily related to increased platelet activation, plays a key role in cardiovascular and cerebrovascular diseases. Although the established antiplatelet agents, such as aspirin and clopidogrel, have been shown to be beneficial in treating thromboembolic diseases, they have considerable limitations. Hence, the development of more effective and safe antithrombotic agents is necessary to satisfy a substantial unmet clinical need. In recent years, the favorable properties of imidazole-related drugs have prompted medicinal chemists to synthesize numerous novel therapeutic agents. The chemical structure of the benzimidazole backbone has proven antiplatelet properties. Moreover, synthetic oligosaccharides have exhibited antiplatelet properties. Therefore, we developed a new aldo-benzimidazole-derived oligosaccharide compound, M3BIM, for achieving a stronger antiplatelet effect than the drugs which are being used in clinical aspects. We investigated the effects of M3BIM on platelet activation ex vivo and its antithrombotic activity in vivo. RESULTS: M3BIM (10-50 µM) exhibited a more potent activity in inhibiting platelet aggregation stimulated by collagen than it did in inhibiting that stimulated by thrombin in washed human platelets. The M3BIM treatment revealed no cytotoxicity in zebrafish embryos, even at the highest concentration of 100 µM. In addition, M3BIM inhibited the phosphorylation of phospholipase Cγ2, protein kinase C (PKC), and mitogen-activated protein kinases (MAPKs; extracellular signal-regulated kinase 2 and c-Jun N-terminal kinase 1), and markedly reduced the ATP-release reaction and intracellular calcium mobilization in collagen-activated platelets. By contrast, M3BIM showed no effects on either collagen-induced p38 MAPK and Akt phosphorylation or phorbol 12, 13-dibutyrate-induced PKC activation and platelet aggregation. Moreover, the M3BIM treatment substantially prolonged the closure time in human whole blood, and increased the occlusion time in mesenteric microvessels and attenuated cerebral infarction in mice. For the study of anticoagulant activities, M3BIM showed no significant effects in the prolongation of activated partial thromboplastin time and prothrombin time in mice. CONCLUSION: The findings of our study suggest that M3BIM is a potential therapeutic agent for preventing or treating thromboembolic disorders.


Asunto(s)
Bencimidazoles , Plaquetas/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Tromboembolia/tratamiento farmacológico , Trisacáridos , Animales , Bencimidazoles/química , Bencimidazoles/farmacología , Plaquetas/patología , Humanos , Ratones , Tromboembolia/metabolismo , Tromboembolia/patología , Trisacáridos/química , Trisacáridos/farmacología , Pez Cebra
9.
Phytother Res ; 30(2): 214-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26560814

RESUMEN

Nobiletin, a bioactive polymethoxylated flavone (5,6,7,8,3(') ,4(') -hexamethoxyflavone), is abundant in citrus fruit peel. Although nobiletin exhibits antitumor activity against various cancer cells, the effect of nobiletin on glioma cells remains unclear. The aim of this study was to determine the effects of nobiletin on the human U87 and Hs683 glioma cell lines. Treating glioma cells with nobiletin (20-100 µm) reduced cell viability and arrested the cell cycle in the G0/G1 phase, as detected using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and propidium iodide (PI) staining, respectively; however, nobiletin did not induce cell apoptosis according to PI-annexin V double staining. Data from western blotting showed that nobiletin significantly attenuated the expression of cyclin D1, cyclin-dependent kinase 2, cyclin-dependent kinase 4, and E2 promoter-binding factor 1 (E2F1) and the phosphorylation of Akt/protein kinase B and mitogen-activated protein kinases, including p38, extracellular signal-regulated kinase, and c-Jun N-terminal kinase. Our data also showed that nobiletin inhibited glioma cell migration, as detected by both functional wound healing and transwell migration assays. Altogether, the present results suggest that nobiletin inhibits mitogen-activated protein kinase and Akt/protein kinase B pathways and downregulates positive regulators of the cell cycle, leading to subsequent suppression of glioma cell proliferation and migration. Our findings evidence that nobiletin may have potential for treating glioblastoma multiforme.


Asunto(s)
Puntos de Control del Ciclo Celular/efectos de los fármacos , Flavonas/farmacología , Glioma/patología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citrus/química , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Factor de Transcripción E2F1/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
10.
Int J Mol Sci ; 17(1)2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26729100

RESUMEN

Astaxanthin, a potent antioxidant carotenoid, plays a major role in modulating the immune response. In this study, we examined the immunomodulatory effects of astaxanthin on cytokine production in primary cultured lymphocytes both in vitro and ex vivo. Direct administration of astaxanthin (70-300 nM) did not produce cytotoxicity in lipopolysaccharide (LPS, 100 µg/ mL)- or concanavalin A (Con A, 10 µg/ mL)-activated lymphocytes, whereas astaxanthin alone at 300 nM induced proliferation of splenic lymphocytes (p < 0.05) in vitro. Although astaxanthin, alone or with Con A, had no apparent effect on interferon (INF-γ) and interleukin (IL-2) production in primary cultured lymphocytes, it enhanced LPS-induced INF-γ production. In an ex vivo experiment, oral administration of astaxanthin (0.28, 1.4 and 7 mg/kg/day) for 14 days did not cause alterations in the body or spleen weights of mice and also was not toxic to lymphocyte cells derived from the mice. Moreover, treatment with astaxanthin significantly increased LPS-induced lymphocyte proliferation ex vivo but not Con A-stimulated lymphocyte proliferation ex vivo. Enzyme linked immunosorbent assay (ELISA) analysis revealed that administration of astaxanthin significantly enhanced INF-γ production in response to both LPS and Con A stimulation, whereas IL-2 production increased only in response to Con A stimulation. Also, astaxanthin treatment alone significantly increased IL-2 production in lymphocytes derived from mice, but did not significantly change production of INF-γ. These findings suggest that astaxanthin modulates lymphocytic immune responses in vitro, and that it partly exerts its ex vivo immunomodulatory effects by increasing INF-γ and IL-2 production without inducing cytotoxicity.


Asunto(s)
Interferón gamma/metabolismo , Interleucina-2/metabolismo , Linfocitos/metabolismo , Animales , Células Cultivadas , Concanavalina A , Ensayo de Inmunoadsorción Enzimática , Sistema Inmunológico/efectos de los fármacos , Lipopolisacáridos , Activación de Linfocitos , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Masculino , Ratones , Bazo , Xantófilas/inmunología , Xantófilas/farmacología
11.
Platelets ; 25(8): 595-602, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24433214

RESUMEN

Hinokitiol (4-isopropyl-tropolone) is a bioactive compound with various pharmacological activities that is found in the wood of cupressaceous plants. Platelet activation plays an important role in thrombogenesis. In our previous study, hinokitiol specifically inhibited collagen-induced platelet aggregation ex vivo and prolonged thrombogenesis in vivo. The glycoprotein (GP) VI and integrin α2ß1 are major collagen receptors that mediate platelet adhesion and aggregation. In our current study, we investigated which of these collagen receptors is involved in the hinokitiol-mediated inhibition of platelet activation. Treatment with 2-100 µM hinokitiol caused a dose-dependent right, parallel shift in the collagen concentration-response curve (0.5-10 µg/ml), with no change in the maximal responses. Furthermore, hinokitiol inhibited platelet aggregation and relative [Ca(2+)]i mobilization stimulated by convulxin, an agonist of GP VI, but not by aggretin, an agonist of integrin α2ß1, indicating that hinokitiol mediates the inhibition of platelet activation through GP VI, rather than through integrin α2ß1. Hinokitiol also specifically inhibited the convulxin-mediated activation of protein kinase C, phospholipase Cγ2, Akt, mitogen-activated protein kinases, and Lyn. Hinokitiol markedly diminished the co-immunoprecipitation of GP VI-bound Lyn after convulxin stimulation. In conclusion, hinokitiol, an antagonist of collagen GP VI may represent a novel antiplatelet drug for the prevention of thrombi associated with coronary and cerebral artery diseases.


Asunto(s)
Plaquetas/metabolismo , Glicoproteínas/antagonistas & inhibidores , Monoterpenos/antagonistas & inhibidores , Tropolona/análogos & derivados , Humanos , Monoterpenos/farmacología , Activación Plaquetaria , Tropolona/antagonistas & inhibidores , Tropolona/farmacología
12.
ScientificWorldJournal ; 2014: 745802, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25541625

RESUMEN

Antrodia camphorata is a rare Taiwanese medicinal mushroom. Antrodia camphorata extract has been reported to exhibit antioxidant, anti-inflammation, antimetastasis, and anticancer activities and plays a role in liver fibrosis, vasorelaxation, and immunomodulation. Critical vascular inflammation leads to vascular dysfunction and cardiovascular diseases, including abdominal aortic aneurysms, hypertension, and atherosclerosis. Platelet activation plays a crucial role in intravascular thrombosis, which is involved in a wide variety of cardiovascular diseases. However, the effect of Antrodia camphorata on platelet activation remains unclear. We examined the effects of Antrodia camphorata on platelet activation. In the present study, Antrodia camphorata treatment (56-224 µg/mL) inhibited platelet aggregation induced by collagen, but not U46619, an analogue of thromboxane A2, thrombin, and arachidonic acid. Antrodia camphorata inhibited collagen-induced calcium (Ca(2+)) mobilization and phosphorylation of protein kinase C (PKC) and Akt. In addition, Antrodia camphorata significantly reduced the aggregation and phosphorylation of PKC in phorbol-12, 13-dibutyrate (PDBu) activated platelets. In conclusion, Antrodia camphorata may inhibit platelet activation by inhibiting of Ca(2+) and PKC cascade and the Akt pathway. Our study suggests that Antrodia camphorata may be a potential therapeutic agent for preventing or treating thromboembolic disorders.


Asunto(s)
Antrodia/química , Plaquetas/enzimología , Señalización del Calcio/efectos de los fármacos , Mezclas Complejas/farmacología , Agregación Plaquetaria/efectos de los fármacos , Proteína Quinasa C/metabolismo , Trombosis/tratamiento farmacológico , Plaquetas/patología , Mezclas Complejas/química , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombosis/enzimología
13.
J Biomed Sci ; 20: 4, 2013 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-23350663

RESUMEN

BACKGROUND: Brazilin, isolated from the heartwood of Caesalpinia sappan L., has been shown to possess multiple pharmacological properties. METHODS: In this study, platelet aggregation, flow cytometry, immunoblotting analysis, and electron spin resonance (ESR) spectrometry were used to investigate the effects of brazilin on platelet activation ex vivo. Moreover, fluorescein sodium-induced platelet thrombi of mesenteric microvessels was also used in in vivo study. RESULTS: We demonstrated that relatively low concentrations of brazilin (1 to 10 µM) potentiated platelet aggregation induced by collagen (0.1 µg/ml) in washed human platelets. Higher concentrations of brazilin (20 to 50 µM) directly triggered platelet aggregation. Brazilin-mediated platelet aggregation was slightly inhibited by ATP (an antagonist of ADP). It was not inhibited by yohimbine (an antagonist of epinephrine), by SCH79797 (an antagonist of thrombin protease-activated receptor [PAR] 1), or by tcY-NH2 (an antagonist of PAR 4). Brazilin did not significantly affect FITC-triflavin binding to the integrin αIIbß(3) in platelet suspensions. Pretreatment of the platelets with caffeic acid phenethyl ester (an antagonist of collagen receptors) or JAQ1 and Sam.G4 monoclonal antibodies raised against collagen receptor glycoprotein VI and integrin α2ß(1), respectively, abolished platelet aggregation stimulated by collagen or brazilin. The immunoblotting analysis showed that brazilin stimulated the phosphorylation of phospholipase C (PLC)γ2 and Lyn, which were significantly attenuated in the presence of JAQ1 and Sam.G4. In addition, brazilin did not significantly trigger hydroxyl radical formation in ESR analysis. An in vivo mouse study showed that brazilin treatment (2 and 4 mg/kg) significantly shortened the occlusion time for platelet plug formation in mesenteric venules. CONCLUSION: To the best of our knowledge, this study provides the first evidence that brazilin acts a novel collagen receptor agonist. Brazilin is a plant-based natural product, may offer therapeutic potential as intended anti-thrombotic agents for targeting of collagen receptors or to be used a useful tool for the study of detailed mechanisms in collagen receptors-mediated platelet activation.


Asunto(s)
Benzopiranos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Trombosis/tratamiento farmacológico , Animales , Benzopiranos/química , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Caesalpinia/química , Colágeno/metabolismo , Citometría de Flujo , Humanos , Ratones , Fosforilación , Agregación Plaquetaria/efectos de los fármacos , Receptores de Colágeno/agonistas , Receptores de Colágeno/metabolismo
14.
Phytother Res ; 27(11): 1671-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23292890

RESUMEN

Andrographolide, a novel nuclear factor-κB (NF-κB) inhibitor, is isolated from the leaves of Andrographis paniculata. Platelet activation is relevant to a variety of coronary heart diseases. Our recent studies revealed that andrographolide possesses potent antiplatelet activity by inhibition of the p38 MAPK/(●) HO-NF-κB-ERK2 cascade. Although platelets are anucleated cells, apoptotic machinery apparatus recently has been found to regulate platelet activation and limit platelet lifespan. Therefore, we further investigated the regulatory effects of andrographolide on platelet apoptotic events. In this study, apoptotic signaling events for caspase-3, -8, and Bid were time (10-60 min)- and dose (25-100 µΜ)-dependently activated by andrographolide in human platelets. Andrographolide could also disrupt mitrochondrial membrane potential. In addition, caspase-8 inhibitor (z-IETD-fmk, 50 µΜ) was found to reverse andrographolide-induced caspase-8 activation, whereas the antagonistic anti-Fas receptor (ZB4, 500 ng/mL) and anti-tumor necrosis factor-R1 (H398, 10 µg/mL) monoclonal antibodies did not. In conclusion, this study for the first time demonstrated that andrographolide might limit platelet lifespan by initiating the caspase-8-dependent extrinsic apoptotic pathway, in spite of no direct evidence that death receptors are involved in this process proved. Overall, the various medicinal properties of andrographolide suggest its potential value in treating patients with thromboembolic disorders.


Asunto(s)
Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Diterpenos/farmacología , Andrographis/química , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Plaquetas/citología , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Potencial de la Membrana Mitocondrial , Hojas de la Planta/química , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
Pharm Biol ; 51(9): 1150-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23930775

RESUMEN

CONTEXT: Andrographolide, extracted from the leaves of Andrographis paniculata (Burm. f.) Nees (Acanthaceae), is a labdane diterpene lactone. It is widely reported to possess anti-inflammatory and antitumorigenic activities. Cerebral endothelial cells (CECs) play a crucial role in supporting the integrity and the function of the blood-brain barrier (BBB). However, no data are available concerning the effects of andrographolide in CECs. The aim of this study was to examine the detailed mechanisms of andrographolide on CECs. OBJECTIVE: This study investigated a novel bioactivity of andrographolide on cerebral ischemia/reperfusion-induced brain injury. MATERIALS AND METHODS: CECs were treated with andrographolide (20-100 µΜ) for the indicated times (0-24 h). After the reactions, cell survival rate and cytotoxicity were tested by the MTT assay and the lactate dehydrogenase (LDH) test, respectively. Western blotting was used to detect caspase-3 expression. In addition, analysis of cell cycle and apoptosis using PI staining and annexin V-FITC/PI labeling, respectively, was performed by flow cytometry. We also investigated the effect of andrographolide on middle cerebral artery occlusion (MCAO)/reperfusion-induced brain injury in a rat model. RESULTS: In the present study, we found that andrographolide (50-100 µΜ) markedly inhibited CEC growth according to an MTT assay and caused CEC damage according to a LDH test. Our data also revealed that andrographolide (50 µM) induced CEC apoptosis and caspase-3 activation as respectively detected by PI/annexin-V double staining and western blotting. Moreover, andrographolide arrested the CEC cell cycle at the G0/G1 phase by PI staining. In addition, andrographolide (5 mg/kg) caused deterioration of MCAO/reperfusion-induced brain injury in a rat model. CONCLUSIONS: These data suggest that andrographolide may disrupt BBB integrity, thereby deteriorating MCAO/reperfusion-induced brain injury, which are, in part, associated with its capacity to arrest cell-cycle and induce CEC apoptosis.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Apoptosis/efectos de los fármacos , Ventrículos Cerebrales/efectos de los fármacos , Diterpenos/efectos adversos , Endotelio Vascular/efectos de los fármacos , Infarto de la Arteria Cerebral Media/fisiopatología , Daño por Reperfusión/inducido químicamente , Andrographis/química , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/enzimología , Caspasa 3/química , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Cerebrales/irrigación sanguínea , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/enzimología , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Masculino , Ratones , Hojas de la Planta/química , Ratas , Ratas Wistar , Daño por Reperfusión/enzimología , Daño por Reperfusión/etiología , Fase de Descanso del Ciclo Celular/efectos de los fármacos
16.
Thromb Res ; 225: 63-72, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37030187

RESUMEN

INTRODUCTION: Paclitaxel is a microtubule-stabilizing drug used to treat several types of cancer, including ovarian and breast cancer. Because of its antiproliferative effect on vascular smooth muscle cells, balloons and stents are coated with paclitaxel for use in coronary revascularization and prevention of in-stent restenosis (ISR). However, mechanisms underlying ISR are complicated. Platelet activation is one of the major causes of ISR after percutaneous coronary intervention. Although the antiplatelet activity of paclitaxel was noted in rabbit platelets, the effect of paclitaxel on platelets remains unclear. This study investigated whether paclitaxel exhibits antiplatelet activity in human platelets. METHODS AND RESULTS: Paclitaxel inhibited platelet aggregation induced by collagen but not that induced by thrombin, arachidonic acid, or U46619, suggesting that paclitaxel is more sensitive to the inhibition of collagen-induced platelet activation. Moreover, paclitaxel blocked collagen receptor glycoprotein (GP) VI downstream signaling molecules, including Lyn, Fyn, PLCγ2, PKC, Akt, and MAPKs. However, paclitaxel did not directly bind to GPVI and cause GPVI shedding, as detected by surface plasmon resonance and flow cytometry, respectively, indicating that paclitaxel may interfere with GPVI downstream signaling molecules, such as Lyn and Fyn. Paclitaxel also prevented granule release and GPIIbIIIa activation induced by collagen and low convulxin doses. Moreover, paclitaxel attenuated pulmonary thrombosis and delayed platelet thrombus formation in mesenteric microvessels without significantly affecting hemostasis. CONCLUSION: Paclitaxel exerts antiplatelet and antithrombotic effects. Thus, paclitaxel may provide additional benefits beyond its antiproliferative effect when used in drug-coated balloons and drug-eluting stents for coronary revascularization and prevention of ISR.


Asunto(s)
Reestenosis Coronaria , Stents Liberadores de Fármacos , Intervención Coronaria Percutánea , Animales , Humanos , Conejos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Stents Liberadores de Fármacos/efectos adversos , Fibrinolíticos , Reestenosis Coronaria/etiología , Reestenosis Coronaria/terapia , Resultado del Tratamiento , Stents/efectos adversos , Intervención Coronaria Percutánea/efectos adversos , Colágeno , Angiografía Coronaria/efectos adversos
17.
Artículo en Inglés | MEDLINE | ID: mdl-22611436

RESUMEN

Xanthohumol is the principal prenylated flavonoid in the hop plant (Humulus lupulus L.). Xanthohumol was found to be a very potent cancer chemopreventive agent through regulation of diverse mechanisms. However, no data are available concerning the effects of xanthohumol on platelet activation. The aim of this paper was to examine the antiplatelet effect of xanthohumol in washed human platelets. In the present paper, xanthohumol exhibited more-potent activity in inhibiting platelet aggregation stimulated by collagen. Xanthohumol inhibited platelet activation accompanied by relative [Ca(2+)](i) mobilization, thromboxane A(2) formation, hydroxyl radical (OH(●)) formation, and phospholipase C (PLC)γ2, protein kinase C (PKC), mitogen-activated protein kinase (MAPK), and Akt phosphorylation. Neither SQ22536, an inhibitor of adenylate cyclase, nor ODQ, an inhibitor of guanylate cyclase, reversed the xanthohumol-mediated inhibitory effect on platelet aggregation. Furthermore, xanthohumol did not significantly increase nitrate formation in platelets. This study demonstrates for the first time that xanthohumol possesses potent antiplatelet activity which may initially inhibit the PI3-kinase/Akt, p38 MAPK, and PLCγ2-PKC cascades, followed by inhibition of the thromboxane A(2) formation, thereby leading to inhibition of [Ca(2+)](i) and finally inhibition of platelet aggregation. Therefore, this novel role of xanthohumol may represent a high therapeutic potential for treatment or prevention of cardiovascular diseases.

19.
Biomed Pharmacother ; 153: 113531, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36076516

RESUMEN

Platelets play a crucial role on hemostasis and are also involved in cardiovascular diseases, such as heart attack and stroke. Artesunate has been reported to possess multiple biological activities, including antitumor and anti-inflammatory activities. However, its effect on platelet activation remains unclear. Thus, we explored the detailed mechanisms underlying its antiplatelet effect. For the in vitro study, the data indicated that artesunate inhibited platelet aggregation induced by collagen, but not thrombin or U46619, indicating that artesunate may selectively inhibit collagen-mediated platelet activation Artesunate also blocked glycoprotein VI (GPVI) downstream signaling, including Syk, PLCγ2, PKC, Akt, and MAPKs. Moreover, artesunate could compete with collagen for binding to collagen receptor and bind to human recombinant GPVI with a high affinity (KD = 44 nM), indicating that it may directly interfere with GPVI. Artesunate also reduced collagen-induced granule release, calcium mobilization, and GPIIbIIIa activation. For the in vivo study, artesunate markedly prevented pulmonary thrombosis and delayed platelet thrombus formation in mesenteric veins and arteries but had minimal effects on hemostasis. In conclusion, we for the first time demonstrated that artesunate acts as a GPVI antagonist and effectively prevents platelet activation and thrombus formation with minimal risk of bleeding, highlighting its therapeutic potential in cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Trombosis , Artesunato/farmacología , Artesunato/uso terapéutico , Plaquetas , Enfermedades Cardiovasculares/metabolismo , Colágeno/metabolismo , Humanos , Activación Plaquetaria , Agregación Plaquetaria , Trombosis/tratamiento farmacológico , Trombosis/metabolismo , Trombosis/prevención & control
20.
J Biomed Sci ; 18: 93, 2011 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-22168157

RESUMEN

BACKGROUND: Platelet activation is relevant to a variety of coronary heart diseases. Our previous studies revealed that sesamol possesses potent antiplatelet activity through increasing cyclic AMP formation. Although platelets are anucleated cells, they also express the transcription factor, NF-κB, that may exert non-genomic functions in platelet activation. Therefore, we further investigated the inhibitory roles of sesamol in NF-κB-mediated platelet function. METHODS: Platelet aggregation, Fura 2-AM fluorescence, and immunoblotting analysis were used in this study. RESULTS: NF-κB signaling events, including IKKß phosphorylation, IκBα degradation, and p65 phosphorylation, were markedly activated by collagen (1 µg/ml) in washed human platelets, and these signaling events were attenuated by sesamol (2.5~25 µM). Furthermore, SQ22536 and ODQ, inhibitors of adenylate cyclase and guanylate cyclase, respectively, strongly reversed the sesamol (25 µM)-mediated inhibitory effects of IKKß phosphorylation, IκBα degradation, and p65 phosphorylation stimulated by collagen. The protein kinase A (PKA) inhibitor, H89, also reversed sesamol-mediated inhibition of IκBα degradation. Moreover, BAY11-7082, an NF-κB inhibitor, abolished IκBα degradation, phospholipase C (PLC)γ2 phosphorylation, protein kinase C (PKC) activation, [Ca(2+)]i mobilization, and platelet aggregation stimulated by collagen. Preincubation of platelets with the inhibitors, SQ22536 and H89, both strongly reversed sesamol-mediated inhibition of platelet aggregation and [Ca(2+)]i mobilization. CONCLUSIONS: Sesamol activates cAMP-PKA signaling, followed by inhibition of the NF-κB-PLC-PKC cascade, thereby leading to inhibition of [Ca(2+)]i mobilization and platelet aggregation. Because platelet activation is not only linked to hemostasis, but also has a relevant role in inflammation and metastasis, our data demonstrating that inhibition of NF-κB interferes with platelet function may have a great impact when these types of drugs are considered for the treatment of cancer and various inflammatory diseases.


Asunto(s)
Antioxidantes/farmacología , Benzodioxoles/farmacología , FN-kappa B/antagonistas & inhibidores , Fenoles/farmacología , Activación Plaquetaria/efectos de los fármacos , Transducción de Señal , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , FN-kappa B/metabolismo , Nucleótidos Cíclicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA