Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
FEBS Lett ; 595(11): 1604-1612, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33792027

RESUMEN

Streptococcus pneumoniae causes pneumonia by infecting the alveolar epithelium via binding to host receptors, such as the platelet-activating factor receptor (PAFR). Although chronic periodontitis has been identified as a pneumonia risk factor, how periodontopathic bacteria cause pneumonia is not known. We found that S. pneumoniae adhered to PAFR expressed on A549 human alveolar epithelial cells stimulated by Porphyromonas gingivalis culture supernatant, and this was abrogated by a PAFR-specific inhibitor. Among the major virulence factors of P. gingivalis [lipopolysaccharide (LPS), fimbriae and gingipains (Rgps and Kgp)], PAFR expression and pneumococcal adhesion were executed in an Rgp-dependent manner. LPS and fimbriae did not induce PAFR expression. Hence, our findings suggest that P. gingivalis enhances pneumococcal adhesion to human alveoli by inducing PAFR expression and that gingipains are responsible for this.


Asunto(s)
Cisteína-Endopeptidasas Gingipaínas/farmacología , Glicoproteínas de Membrana Plaquetaria/genética , Porphyromonas gingivalis/metabolismo , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/genética , Factores de Virulencia/farmacología , Células A549 , Adhesión Bacteriana/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Fimbrias Bacterianas/química , Regulación de la Expresión Génica , Cisteína-Endopeptidasas Gingipaínas/deficiencia , Cisteína-Endopeptidasas Gingipaínas/genética , Interacciones Huésped-Patógeno/genética , Humanos , Lipopolisacáridos/farmacología , Modelos Biológicos , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidad , Alveolos Pulmonares/microbiología , ARN Mensajero/agonistas , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Factores de Virulencia/deficiencia , Factores de Virulencia/genética
2.
Thromb Haemost ; 121(10): 1337-1344, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33690868

RESUMEN

Platelet activation is characterized by shape change, granule secretion, activation of fibrinogen receptor (glycoprotein IIb/IIIa) sustaining platelet aggregation, and externalization of negatively charged aminophospholipids contributing to platelet procoagulant activity. Epinephrine (EPI) alone is a weak platelet activator. However, it is able to potentiate platelet activation initiated by other agonists. In this work, we investigated the role of EPI in the generation of procoagulant platelets. Human platelets were activated with convulxin (CVX), thrombin (THR) or protease-activated receptor (PAR) agonists, EPI, and combination thereof. Platelet aggregation was assessed by light transmission aggregometry or with PAC-1 binding by flow cytometry. Procoagulant collagen-and-THR (COAT) platelets, induced by combined activation with CVX-and-THR, were visualized by flow cytometry as Annexin-V-positive and PAC-1-negative platelets. Cytosolic calcium fluxes were monitored by flow cytometry using Fluo-3 indicator. EPI increased platelet aggregation induced by all agonist combinations tested. On the other hand, EPI dose-dependently reduced the formation of procoagulant COAT platelets generated by combined CVX-and-THR activation. We observed a decreased Annexin-V-positivity and increased binding of PAC-1 with the triple activation (CVX + THR + EPI) compared with CVX + THR. Calcium mobilization with triple activation was decreased with the higher EPI dose (1,000 µM) compared with CVX + THR calcium kinetics. In conclusion, when platelets are activated with CVX-and-THR, the addition of increasing concentrations of EPI (triple stimulation) modulates platelet response reducing cytosolic calcium mobilization, decreasing procoagulant activity, and enhancing platelet aggregation.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Coagulantes/farmacología , Epinefrina/farmacología , Agregación Plaquetaria/efectos de los fármacos , Adolescente , Adulto , Anciano , Plaquetas/metabolismo , Señalización del Calcio , Venenos de Crotálidos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Cinética , Lectinas Tipo C , Masculino , Persona de Mediana Edad , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Trombina/farmacología , Adulto Joven
3.
Biosci Biotechnol Biochem ; 84(1): 85-94, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31794329

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) regulates collagen-mediated platelet activation through its cytoplasmic immunoreceptor tyrosine-based inhibition motifs (ITIMs). However, the function of CEACAM1's extracellular cleavage fragments is currently unknown. In the present study, we used mass spectrometry (MS) to identify 9 cleavage fragments shed by matrix metallopeptidase 12 (MMP-12), and then we synthesized peptides with sequences corresponding to the fragments. QLSNGNRTLT (QLSN), a peptide from the A1-domain of CEACAM1, significantly attenuated collagen-induced platelet aggregation. QLSN also attenuated platelet static adhesion to collagen. Additionally, QLSN reduced human platelet secretion and integrin αIIbß3 activation in response to glycoprotein VI (GPVI)-selective agonist, convulxin. Correspondingly, QLSN treatment significantly decreased convulxin-mediated phosphorylation of Src, protein kinase B (Akt), spleen tyrosine kinase (Syk) and phospholipase Cγ2 (PLCγ2) in human platelets. These data indicate that the CEACAM1-derived peptide QLSN inhibits GPVI-mediated human platelet activation. QLSN could potentially be developed as a novel antiplatelet agent.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Colágeno/metabolismo , Oligopéptidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Plaquetas/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Adhesión Celular/efectos de los fármacos , Venenos de Crotálidos/farmacología , Humanos , Motivo de Inhibición del Inmunorreceptor Basado en Tirosina/fisiología , Lectinas Tipo C , Metaloproteinasa 12 de la Matriz/metabolismo , Oligopéptidos/síntesis química , Fosfolipasa C gamma/metabolismo , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/agonistas , Dominios Proteicos/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinasa Syk/metabolismo
4.
Thromb Haemost ; 119(12): 2005-2013, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31634956

RESUMEN

Synthetic cross-linked collagen-related peptide (CRP-XL) is a glycoprotein VI (GPVI) receptor activator for platelet activation. This triple helical peptide, widely used in platelet function tests, is synthesized and cross-linked through cysteine residues at its N-terminus and C-terminus. Currently, there is only one laboratory, which is capable to produce this valuable peptide for clinical applications. In an attempt to provide a standardized alternative for CRP-XL, we developed a synthetic triple helical collagen peptide (STH-CP) with the same primary sequence as CRP-XL (GPC-(GPO)10-GPCG-amide)3, which was both on the C-terminus and on the N-terminus fixed on a scaffold with a binding side for each of the three peptides. The performance of STH-CP on platelet function was studied using flow cytometry and compared with CRP-XL. We found that platelet activation pattern in response to STH-CP and CRP-XL is similar, although the STH-CP requires sixfold higher concentrations to activate platelets to the same state. The intra-assay percent coefficient of variation of STH-CP and CRP-XL were both < 5% and the interindividual variation measured in 118 individuals for both peptides was around 23 and 21% for αIIbß3 activation and P-selectin expression, respectively. The STH-CP in ready-to-use reaction mix has lower variation than CRP-XL over 1-year storage. In reference values and seasonal variation study, the platelet activation response showed a strong correlation between STH-CP and CRP-XL.Our findings show that this new STH-CP is a stable and potent platelet GPVI agonist which can induce the same reproducible platelet activation as CRP-XL and that STH-CP can be considered as a good alternative for CRP-XL.


Asunto(s)
Colágeno/química , Colágeno/farmacología , Activación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/agonistas , Adulto , Anciano , Sitios de Unión , Plaquetas/metabolismo , Proteínas Portadoras/farmacología , Cisteína/química , Femenino , Citometría de Flujo , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Selectina-P/metabolismo , Péptidos/química , Péptidos/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Dominios Proteicos , Ingeniería de Proteínas/métodos , Adulto Joven
5.
Thromb Haemost ; 119(7): 1124-1137, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31129912

RESUMEN

BACKGROUND: The G protein-coupled receptor, adenosine A2A, signals through the stimulatory G protein, Gs, in platelets leading to activation of adenylyl cyclase and elevation of cyclic adenosine monophosphate (cAMP) and inhibition of platelet activation. OBJECTIVE: This article investigates the effect of A2A receptor activation on signalling by the collagen receptor glycoprotein (GP) VI in platelets. METHODS: Washed human platelets were stimulated by collagen or the GPVI-specific agonist collagen-related peptide (CRP) in the presence of the adenosine receptor agonist, 5'-N-ethylcarboxamidoadenosine (NECA) or the adenylyl cyclase activator, forskolin and analysed for aggregation, adenosine triphosphate secretion, protein phosphorylation, spreading, Ca2+ mobilisation, GPVI receptor clustering, cAMP, thromboxane B2 (TxB2) and P-selectin exposure. RESULTS: NECA, a bioactive adenosine analogue, partially inhibits aggregation and secretion to collagen or CRP in the absence or presence of the P2Y12 receptor antagonist, cangrelor and the cyclooxygenase inhibitor, indomethacin. The inhibitory effect in the presence of the three inhibitors is largely overcome at higher concentrations of collagen but not CRP. Neither NECA nor forskolin altered clustering of GPVI, elevation of Ca2+ or spreading of platelets on a collagen surface. Further, neither NECA nor forskolin, altered collagen-induced tyrosine phosphorylation of Syk, LAT nor PLCγ2. However, NECA and forskolin inhibited platelet activation by the TxA2 mimetic, U46619, but not the combination of adenosine diphosphate and collagen. CONCLUSION: NECA and forskolin have no effect on the proximal signalling events by collagen. They inhibit platelet activation in a response-specific manner in part through inhibition of the feedback action of TxA2.


Asunto(s)
Adenosina/metabolismo , Plaquetas/fisiología , Colforsina/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptor de Adenosina A2A/metabolismo , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Adenilil Ciclasas/metabolismo , Proteínas Portadoras/farmacología , Células Cultivadas , Colágeno/metabolismo , AMP Cíclico/metabolismo , Retroalimentación Fisiológica , Humanos , Indometacina/farmacología , Péptidos/farmacología , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/agonistas , Transducción de Señal , Tromboxano B2/metabolismo
7.
Int J Mol Sci ; 20(1)2018 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-30577630

RESUMEN

Studies, including ours, have shown that pro-oxidative stressors, such as chemotherapeutic agents, generate oxidized lipids with agonistic platelet-activating factor (PAF) activity. Importantly, recent reports have implicated that these PAF-agonists are transported extracellularly via microvesicle particles (MVPs). While the role of PAF-receptor (PAF-R) has been implicated in mediating chemotherapy effects, its significance in chemotherapy-mediated MVP release in pancreatic cancer has not been studied. The current studies determined the functional significance of PAF-R in gemcitabine chemotherapy-mediated MVP release in human pancreatic cancer cells. Using PAF-R-expressing (PANC-1) and PAF-R-deficient (Hs766T) cells, we demonstrate that gemcitabine induces MVP release in a PAF-R-dependent manner. Blocking of PAF-R via PAF-R antagonist or inhibition of MVP generation via inhibitor of acid sphingomyelinase (aSMase) enzyme, significantly attenuated gemcitabine-mediated MVP release from PANC-1 cells, however, exerted no effects in Hs766T cells. Notably, MVPs from gemcitabine-treated PANC-1 cells, contained a measurable amount of PAF-agonists. Mechanistically, pretreatment with ERK1/2 or p38 inhibitors significantly abrogated gemcitabine-mediated MVP release, indicating the involvement of mitogen-activated protein kinase (MAPK) pathway in PAF-R-dependent gemcitabine-mediated MVP release. These findings demonstrate the significance of PAF-R in gemcitabine-mediated MVP release, as well as the rationale of evaluating PAF-R targeting agents with gemcitabine against pancreatic cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Micropartículas Derivadas de Células/metabolismo , Desoxicitidina/análogos & derivados , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias Pancreáticas/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Línea Celular Tumoral , Desoxicitidina/farmacología , Humanos , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo , Gemcitabina
8.
Int J Mol Sci ; 19(10)2018 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-30314274

RESUMEN

Platelet-activating factor-receptor (PAF-R) agonists are pleiotropic lipid factors that influence multiple biological processes, including the induction and resolution of inflammation as well as immunosuppression. PAF-R agonists have been shown to modulate tumorigenesis and/or tumor growth in various skin cancer models by suppressing either cutaneous inflammation and/or anti-tumoral adaptive immunity. We have previously shown that a chronic systemic PAF-R agonist administration of mice enhances the growth of subcutaneously implanted melanoma tumors. Conversely, chronic topical applications of a PAF-R agonist suppressed non-melanoma skin cancer (NMSC) in a topical chemical carcinogenesis model (dimethylbenz[a]anthracene/phorbol 12-myristate 13-acetate (DMBA/PMA)) in-part via anti-inflammatory effects. These results indicate that the context of PAF-R agonist exposure via either chronic cutaneous or systemic administration, result in seemingly disparate effects on tumor promotion. To further dissect the contextual role of PAF-R agonism on tumorigenesis, we chronically administered systemic PAF-R agonist, carbamoyl-PAF (CPAF) to mice under a cutaneous chemical carcinogenesis protocol, recently characterized to initiate both NMSC and melanocytic nevus formation that can progress to malignant melanoma. Our results showed that while systemic CPAF did not modulate melanocytic nevus formation, it enhanced the growth of NMSC tumors.


Asunto(s)
Glicoproteínas de Membrana Plaquetaria/agonistas , Receptores Acoplados a Proteínas G/agonistas , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Animales , Carcinógenos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones , Neoplasias Cutáneas/etiología , Carga Tumoral
9.
J Thromb Haemost ; 16(11): 2315-2321, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30179298

RESUMEN

Essentials Mitochondrial hyperpolarization enhances the conversion of platelets to a procoagulant phenotype. Mitochondrial calcium uniporter (MCU) function is essential in procoagulant platelet formation. Mitochondrial calcium uniporter deletion does not impact other aspects of platelet activation. Ablation of MCU results in the emergence of a permeability transition pore-independent pathway. SUMMARY: Background Procoagulant platelets comprise a phenotypically distinct subpopulation of activated platelets with high-level phosphatidylserine externalization. When initiated by co-stimulation with thrombin and a glycoprotein VI (GPVI) agonist, the transition to the procoagulant phenotype is mediated by extracellular calcium entry and mitochondrial permeability transition pore (mPTP) formation. Objectives The intracellular mechanisms coordinating these distinct cytoplasmic and mitochondrial processes remain unclear. The mitochondrial calcium uniporter (MCU) protein is a central component of the transmembrane ion channel that allows the passage of Ca2+ from the cytosol into the mitochondrial matrix. Here we investigate the role of the MCU in the regulation of procoagulant platelet formation. Results Procoagulant platelet formation was directly correlated with pre-stimulatory mitochondrial transmembrane potential, a key determinant of calcium flux from the cytoplasm to the mitochondria. The role of MCU in the regulation of procoagulant platelet formation was investigated using MCU null platelets. Procoagulant platelet formation in MCU null platelets was significantly decreased coincident with decreased mPTP formation. In contrast, neither granule release nor initial integrin activation was altered in response to stimulation. In the genomic absence of MCU, developmental induction of an alternative intracellular pathway partially rescued procoagulant platelet formation. Conclusion These results identify a key role for the mitochondrial calcium uptake channel in the regulation of mPTP-mediated procoagulant platelet formation and suggest a novel pharmacologic target for procoagulant-platelet-related pathologies.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Activación Plaquetaria/efectos de los fármacos , Animales , Antimicina A/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Citosol/metabolismo , Humanos , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Péptidos/metabolismo , Fenotipo , Fosfatidilserinas/metabolismo , Glicoproteínas de Membrana Plaquetaria/agonistas , Rotenona/farmacología , Transducción de Señal/efectos de los fármacos , Trombina/agonistas
10.
Haematologica ; 103(12): 2097-2108, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30026342

RESUMEN

Ibrutinib and acalabrutinib are irreversible inhibitors of Bruton tyrosine kinase used in the treatment of B-cell malignancies. They bind irreversibly to cysteine 481 of Bruton tyrosine kinase, blocking autophosphorylation on tyrosine 223 and phosphorylation of downstream substrates including phospholipase C-γ2. In the present study, we demonstrate that concentrations of ibrutinib and acalabrutinib that block Bruton tyrosine kinase activity, as shown by loss of phosphorylation at tyrosine 223 and phospholipase C-γ2, delay but do not block aggregation in response to a maximally-effective concentration of collagen-related peptide or collagen. In contrast, 10- to 20-fold higher concentrations of ibrutinib or acalabrutinib block platelet aggregation in response to glycoprotein VI agonists. Ex vivo studies on patients treated with ibrutinib, but not acalabrutinib, showed a reduction of platelet aggregation in response to collagen-related peptide indicating that the clinical dose of ibrutinib but not acalabrutinib is supramaximal for Bruton tyrosine kinase blockade. Unexpectedly, low concentrations of ibrutinib inhibited aggregation in response to collagen-related peptide in patients deficient in Bruton tyrosine kinase. The increased bleeding seen with ibrutinib over acalabrutinib is due to off-target actions of ibrutinib that occur because of unfavorable pharmacodynamics.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Agammaglobulinemia/tratamiento farmacológico , Plaquetas/efectos de los fármacos , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Glicoproteínas de Membrana Plaquetaria/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa/genética , Agammaglobulinemia Tirosina Quinasa/metabolismo , Agammaglobulinemia/sangre , Agammaglobulinemia/genética , Benzamidas/administración & dosificación , Benzamidas/metabolismo , Plaquetas/metabolismo , Proteínas Portadoras/administración & dosificación , Enfermedades Genéticas Ligadas al Cromosoma X/sangre , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Humanos , Mutación , Péptidos/administración & dosificación , Piperidinas , Activación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Glicoproteínas de Membrana Plaquetaria/agonistas , Inhibidores de Proteínas Quinasas/metabolismo , Pirazinas/administración & dosificación , Pirazinas/metabolismo , Pirazoles/administración & dosificación , Pirazoles/metabolismo , Pirimidinas/administración & dosificación , Pirimidinas/metabolismo
11.
Nat Struct Mol Biol ; 25(6): 488-495, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29808000

RESUMEN

Platelet-activating-factor receptor (PAFR) responds to platelet-activating factor (PAF), a phospholipid mediator of cell-to-cell communication that exhibits diverse physiological effects. PAFR is considered an important drug target for treating asthma, inflammation and cardiovascular diseases. Here we report crystal structures of human PAFR in complex with the antagonist SR 27417 and the inverse agonist ABT-491 at 2.8-Å and 2.9-Å resolution, respectively. The structures, supported by molecular docking of PAF, provide insights into the signal-recognition mechanisms of PAFR. The PAFR-SR 27417 structure reveals an unusual conformation showing that the intracellular tips of helices II and IV shift outward by 13 Å and 4 Å, respectively, and helix VIII adopts an inward conformation. The PAFR structures, combined with single-molecule FRET and cell-based functional assays, suggest that the conformational change in the helical bundle is ligand dependent and plays a critical role in PAFR activation, thus greatly extending knowledge about signaling by G-protein-coupled receptors.


Asunto(s)
Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Sitios de Unión , Cristalografía por Rayos X , Transferencia Resonante de Energía de Fluorescencia , Humanos , Enlace de Hidrógeno , Imidazoles/farmacología , Indoles/farmacología , Ligandos , Simulación del Acoplamiento Molecular , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Conformación Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Tiazoles/farmacología
12.
Thromb Haemost ; 117(8): 1588-1600, 2017 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-28536721

RESUMEN

The platelet receptors glycoprotein (Gp)VI, integrin α2ß1 and GpIb/V/IX mediate platelet adhesion and activation during thrombogenesis. Increases of intracellular Ca2+ ([Ca2+]i) are key signals during platelet activation; however, their relative importance in coupling different collagen receptors to functional responses under shear conditions remains unclear. To study shear-dependent, receptor-specific platelet responses, we used collagen or combinations of receptor-specific collagen-mimetic peptides as substrates for platelet adhesion and activation in whole human blood under arterial flow conditions and compared real-time and endpoint parameters of thrombus formation alongside [Ca2+]i measurements using confocal imaging. All three collagen receptors coupled to [Ca2+]i signals, but these varied in amplitude and temporal pattern alongside variable integrin activation. GpVI engagement produced large, sustained [Ca2+]i signals leading to real-time increases in integrins α2ß1- and αIIbß3-mediated platelet adhesion. αIIbß3-dependent platelet aggregation was dependent on P2Y12 signalling. Co-engagement of α2ß1 and GpIb/V/IX generated transient [Ca2+]i spikes and low amplitude [Ca2+]i responses that potentiated GpVI-dependent [Ca2+]i signalling. Therefore α2ß1, GpIb/V/IX and GpVI synergise to generate [Ca2+]i signals that regulate platelet behaviour and thrombus formation. Antagonism of secondary signalling pathways reveals distinct, separate roles for αIIbß3 in stable platelet adhesion and aggregation.


Asunto(s)
Plaquetas/metabolismo , Señalización del Calcio , Hemorreología , Integrina alfa2beta1/metabolismo , Activación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Plaquetas/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Colágeno/farmacología , Humanos , Integrina alfa2beta1/agonistas , Microscopía Confocal , Péptidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Adhesividad Plaquetaria , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/agonistas , Factores de Tiempo
13.
Arterioscler Thromb Vasc Biol ; 37(5): 823-835, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28336561

RESUMEN

OBJECTIVE: Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a collagen receptor that belongs to the inhibitory immunoreceptor tyrosine-based inhibition motif-containing receptor family. It is an inhibitor of signaling via the immunoreceptor tyrosine-based activation motif-containing collagen receptor complex, glycoprotein VI-FcRγ-chain. It is expressed on hematopoietic cells, including immature megakaryocytes, but is not detectable on platelets. Although the inhibitory function of LAIR-1 has been described in leukocytes, its physiological role in megakaryocytes and in particular in platelet formation has not been explored. In this study, we investigate the role of LAIR-1 in megakaryocyte development and platelet production by generating LAIR-1-deficient mice. APPROACH AND RESULTS: Mice lacking LAIR-1 exhibit a significant increase in platelet counts, a prolonged platelet half-life in vivo, and increased proplatelet formation in vitro. Interestingly, platelets from LAIR-1-deficient mice exhibit an enhanced reactivity to collagen and the glycoprotein VI-specific agonist collagen-related peptide despite not expressing LAIR-1, and mice showed enhanced thrombus formation in the carotid artery after ferric chloride injury. Targeted deletion of LAIR-1 in mice results in an increase in signaling downstream of the glycoprotein VI-FcRγ-chain and integrin αIIbß3 in megakaryocytes because of enhanced Src family kinase activity. CONCLUSIONS: Findings from this study demonstrate that ablation of LAIR-1 in megakaryocytes leads to increased Src family kinase activity and downstream signaling in response to collagen that is transmitted to platelets, rendering them hyper-reactive specifically to agonists that signal through Syk tyrosine kinases, but not to G-protein-coupled receptors.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , Activación Plaquetaria , Receptores Inmunológicos/deficiencia , Trombocitosis/sangre , Trombosis/sangre , Animales , Plaquetas/efectos de los fármacos , Proteínas Portadoras/farmacología , Células Cultivadas , Cloruros , Modelos Animales de Enfermedad , Activación Enzimática , Compuestos Férricos , Predisposición Genética a la Enfermedad , Megacariocitos/efectos de los fármacos , Ratones Noqueados , Péptidos/farmacología , Fenotipo , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores de IgG/sangre , Receptores Inmunológicos/genética , Transducción de Señal/efectos de los fármacos , Trombocitosis/genética , Trombosis/inducido químicamente , Trombosis/genética , Familia-src Quinasas/sangre
14.
Arterioscler Thromb Vasc Biol ; 37(5): 812-822, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28254816

RESUMEN

OBJECTIVE: Platelets have been found to express intracellular nuclear receptors including the retinoid X receptors (RXRα and RXRß). Treatment of platelets with ligands of RXR has been shown to inhibit platelet responses to ADP and thromboxane A2; however, the effects on responses to other platelet agonists and the underlying mechanism have not been fully characterized. APPROACH AND RESULTS: The effect of 9-cis-retinoic acid, docosahexaenoic acid and methoprene acid on collagen receptor (glycoprotein VI [GPVI]) agonists and thrombin-stimulated platelet function; including aggregation, granule secretion, integrin activation, calcium mobilization, integrin αIIbß3 outside-in signaling and thrombus formation in vitro and in vivo were determined. Treatment of platelets with RXR ligands resulted in attenuation of platelet functional responses after stimulation by GPVI agonists or thrombin and inhibition of integrin αIIbß3 outside-in signaling. Treatment with 9-cis-retinoic acid caused inhibition of thrombus formation in vitro and an impairment of thrombosis and hemostasis in vivo. Both RXR ligands stimulated protein kinase A activation, measured by VASP S157 phosphorylation, that was found to be dependent on both cAMP and nuclear factor κ-light-chain-enhancer of activated B cell activity. CONCLUSIONS: This study identifies a widespread, negative regulatory role for RXR in the regulation of platelet functional responses and thrombus formation and describes novel events that lead to the upregulation of protein kinase A, a known negative regulator of many aspects of platelet function. This mechanism may offer a possible explanation for the cardioprotective effects described in vivo after treatment with RXR ligands.


Asunto(s)
Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Hemostasis/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Receptores X Retinoide/agonistas , Trombosis/prevención & control , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/metabolismo , Señalización del Calcio/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Ligandos , Masculino , Ratones , Proteínas de Microfilamentos/metabolismo , FN-kappa B/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores X Retinoide/metabolismo , Sistemas de Mensajero Secundario/efectos de los fármacos , Trombina/farmacología , Trombosis/sangre , Factores de Tiempo
15.
Haematologica ; 102(3): e89-e92, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27979925
16.
Oncotarget ; 7(15): 20788-800, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-26959112

RESUMEN

Pro-oxidative stressors can suppress host immunity due to their ability to generate oxidized lipid agonists of the platelet-activating factor-receptor (PAF-R). As radiation therapy also induces reactive oxygen species, the present studies were designed to define whether ionizing radiation could generate PAF-R agonists and if these lipids could subvert host immunity. We demonstrate that radiation exposure of multiple tumor cell lines in-vitro, tumors in-vivo, and human subjects undergoing radiation therapy for skin tumors all generate PAF-R agonists. Structural characterization of radiation-induced PAF-R agonistic activity revealed PAF and multiple oxidized glycerophosphocholines that are produced non-enzymatically. In a murine melanoma tumor model, irradiation of one tumor augmented the growth of the other (non-treated) tumor in a PAF-R-dependent process blocked by a cyclooxygenase-2 inhibitor. These results indicate a novel pathway by which PAF-R agonists produced as a byproduct of radiation therapy could result in tumor treatment failure, and offer important insights into potential therapeutic strategies that could improve the overall antitumor effectiveness of radiation therapy regimens.


Asunto(s)
Antioxidantes/farmacología , Melanoma/terapia , Factor de Activación Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/agonistas , Receptores Acoplados a Proteínas G/agonistas , Neoplasias Cutáneas/terapia , Rayos Ultravioleta , Animales , Femenino , Humanos , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Estrés Oxidativo , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/secundario , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Arterioscler Thromb Vasc Biol ; 36(5): 846-54, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26988594

RESUMEN

OBJECTIVE: Reactive oxygen species (ROS) are known to regulate platelet activation; however, the mechanisms of ROS production during platelet activation remain unclear. Platelets express different isoforms of nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidases (NOXs). Here, we investigated the role of NOX1 and NOX2 in ROS generation and platelet activation using NOX1 and NOX2 knockout mice. APPROACH AND RESULTS: NOX1(-/Y) platelets showed selective defects in G-protein-coupled receptor-mediated platelet activation induced by thrombin and thromboxane A2 analog U46619, but were not affected in platelet activation induced by collagen-related peptide, a glycoprotein VI agonist. In contrast, NOX2(-/-) platelets showed potent inhibition of collagen-related peptide-induced platelet activation, and also showed partial inhibition of thrombin-induced platelet activation. Consistently, production of ROS was inhibited in NOX1(-/Y) platelets stimulated with thrombin, but not collagen-related peptide, whereas NOX2(-/-) platelets showed reduced ROS generation induced by collagen-related peptide or thrombin. Reduced ROS generation in NOX1/2-deficient platelets is associated with impaired activation of Syk and phospholipase Cγ2, but minimally affected mitogen-activated protein kinase pathways. Interestingly, laser-induced arterial thrombosis was impaired but the bleeding time was not affected in NOX2(-/-) mice. Wild-type thrombocytopenic mice injected with NOX2(-/-) platelets also showed defective arterial thrombosis, suggesting an important role for platelet NOX2 in thrombosis in vivo but not hemostasis. CONCLUSIONS: NOX1 and NOX2 play differential roles in different platelet activation pathways and in thrombosis. ROS generated by these enzymes promotes platelet activation via the Syk/phospholipase Cγ2/calcium signaling pathway.


Asunto(s)
Plaquetas/enzimología , Glicoproteínas de Membrana/sangre , NADH NADPH Oxidorreductasas/sangre , NADPH Oxidasas/sangre , Activación Plaquetaria , Especies Reactivas de Oxígeno/sangre , Trombosis/sangre , Trombosis/enzimología , Animales , Plaquetas/efectos de los fármacos , Antígeno CD11b/sangre , Señalización del Calcio , Modelos Animales de Enfermedad , Activación Enzimática , Predisposición Genética a la Enfermedad , Hemostasis , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , NADH NADPH Oxidorreductasas/deficiencia , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , Fenotipo , Fosfolipasa C gamma/sangre , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/sangre , Quinasa Syk/sangre , Trombina/metabolismo , Trombosis/genética , Factores de Tiempo
18.
J Nutr Biochem ; 26(9): 978-85, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26013469

RESUMEN

Hepatic diseases are comorbidities caused by obesity and are influenced by diet composition. The aim of this study was to evaluate the kinetics of metabolic and inflammatory liver dysfunction induced by a high-refined carbohydrate-containing (HC) diet and to determine how platelet-activating factor (PAF) modulates the liver lipid content of mice. BALB/c mice were fed a chow or HC diet for the following experimental periods: 1 and 3 days, 1, 2, 4, 6, 8, 10 and 12 weeks. Wild-type (WT) and PAF receptor-deficient (PAFR(-/-)) mice were fed the same diets for 8 weeks. Mice fed with HC diet showed higher triglycerides and cholesterol levels, fibrosis and inflammation in the liver. The number of neutrophils migrating into the liver was also increased in mice fed with HC diet. However, transaminase levels did not change. PAFR(-/-) mice fed with HC diet showed more steatosis, oxidative stress and higher transaminases levels associated with lower inflammation than WT mice. The consumption of HC diet altered the metabolic and inflammatory response in the liver and was worse in PAFR(-/-) mice. We suggest that PAF regulates liver lipid content and dyslipidemia, protecting the mice from lipotoxicity and liver damage.


Asunto(s)
Carbohidratos de la Dieta/efectos adversos , Metabolismo de los Lípidos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factor de Activación Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/agonistas , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Animales , Colesterol/sangre , Colesterol/metabolismo , Dislipidemias/etiología , Dislipidemias/inmunología , Dislipidemias/metabolismo , Dislipidemias/patología , Manipulación de Alimentos , Peroxidación de Lípido , Hígado/inmunología , Hígado/patología , Cirrosis Hepática/etiología , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Infiltración Neutrófila , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/sangre , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Factores de Tiempo , Triglicéridos/sangre , Triglicéridos/metabolismo
19.
Mediators Inflamm ; 2015: 820543, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25922565

RESUMEN

Chemotherapy is used to treat numerous cancers including melanoma. However, its effectiveness in clinical settings is often hampered by various mechanisms. Previous studies have demonstrated that prooxidative stressor-mediated generation of oxidized lipids with platelet-activating factor-receptor (PAF-R) agonistic activity induces systemic immunosuppression that augments the growth of experimental melanoma tumors. We have recently shown that treatment of murine B16F10 melanoma cells in vitro or tumors implanted into syngeneic mice and treated intratumorally with various chemotherapeutic agents generated PAF-R agonists in a process blocked by antioxidants. Notably, these intratumoral chemotherapy-generated PAF-R agonists augmented the growth of secondary (untreated) tumors in a PAF-R dependent manner. As both localized and systemic chemotherapies are used based on tumor localization/stage and metastases, the current studies were sought to determine effects of PAF-R agonists on systemic chemotherapy against experimental melanoma. Here, we show that systemic chemotherapy with etoposide (ETOP) attenuates the growth of melanoma tumors when given subsequent to the tumor cell implantation. Importantly, this ETOP-mediated suppression of melanoma tumor growth was blocked by exogenous administration of a PAF-R agonist, CPAF. These findings indicate that PAF-R agonists not only negatively affect the ability of localized chemotherapy but also compromise the efficacy of systemic chemotherapy against murine melanoma.


Asunto(s)
Antineoplásicos/farmacología , Factor de Activación Plaquetaria/análogos & derivados , Glicoproteínas de Membrana Plaquetaria/agonistas , Receptores Acoplados a Proteínas G/agonistas , Animales , Antineoplásicos/uso terapéutico , Antioxidantes/química , Etopósido/uso terapéutico , Femenino , Terapia de Inmunosupresión , Melanoma/tratamiento farmacológico , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Trasplante de Neoplasias , Factor de Activación Plaquetaria/uso terapéutico , Transducción de Señal
20.
Mol Med Rep ; 12(1): 394-400, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25695262

RESUMEN

Melanoma cells often express platelet-activating factor receptor (PAF-R), which has been demonstrated to increase metastatic behavior. However, the effect of PAF-R on the responsiveness of melanoma to naturally occurring cytotoxic agents remains to be elucidated. The present study aimed to determine the relative cytotoxicity and mechanism of benzyl isothiocyanate (BITC), a component of cruciferous vegetables, in melanoma cells expressing PAF-R. To evaluate the importance of PAF-R signaling in melanoma cell growth, PAF-R-negative murine B16F10 cells were transduced with a retrovirus containing the cDNA for PAF-R to generate cells stably expressing PAF-R (B16-PAF-R) or an empty vector (MSCV) to generate PAF-R-deficient B16-MSCV control cells. Activation of PAF-R, using the PAF-R agonist, 1-hexadecyl-2-N-methylcarbamoyl-3-glycerophosphocholine, induced an increase in the proliferation of B16-PAF-R cells compared with the B16-MSCV cells. Reverse transcription quantitative polymerase chain reaction revealed the presence of functional PAF-R in human melanoma SK23MEL cells, but not in SK5MEL cells. The present study investigated the effect of BITC treatments on the survival of murine and human melanoma cells, in the presence or absence of functional PAF-R. The results revealed that treatment with BITC decreased the survival rate of the PAF-R-positive and negative murine and human melanoma cells. However, the expression of PAF-R substantially augmented BITC-mediated cytotoxicity in the PAF-R-positive cells at lower concentrations compared with the PAF-R-negative cells. In order to determine the underlying mechanism, flow cytometric analysis was used, which demonstrated a significant increase in the generation of reactive oxygen species (ROS) in the B16-PAF-R cells compared with the B16-MSCV cells, which enhanced apoptosis by BITC, as measured by increased caspase-3/7 luminescence. Notably, the BITC-mediated decreased cell survival rate, increased ROS and increased apoptosis in the B16-PAF-R cells were significantly attenuated by the antioxidant, vitamin C, indicating ROS involvement. Additionally, the WEB2086 PAF-R antagonist, inhibited the BITC-mediated enhancement of apoptosis in the B16-PAF-R cells, indicating a role for PAF-R-signaling in the BITC-mediated effects. These findings indicated that the selectivity of BITC towards PAF­R in melanoma offers a promising chemopreventive agent for PAF-R-positive melanoma treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Isotiocianatos/toxicidad , Melanoma/patología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Azepinas/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Melanoma/metabolismo , Ratones , Factor de Activación Plaquetaria/análogos & derivados , Factor de Activación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/agonistas , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...