Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 324
Filtrar
1.
Blood Rev ; 47: 100774, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33213987

RESUMO

Thrombopoietin receptor agonist (TPO-RA) treatment increases the thrombosis rate in immune thrombocytopenia (ITP). We hypothesize that TPO-RAs influence platelet function, global and secondary hemostasis and/or fibrinolysis. A systematic review was performed. If possible, data were compared between responders (relevant increase in platelet count), and non-responders. Twelve observational studies with 305 patients were included (responders (127/150 (85%))). There were indications that TPO-RA treatment enhanced platelet function, with respect to platelet-monocyte aggregates, soluble P-selectin, GPVI expression, and adhesion under flow. Studies addressing global and secondary hemostasis and fibrinolysis were scarce. Overall, no changes were found during TPO-RA treatment, apart from an accelerated clot formation and conflicting data on levels of plasminogen activator inhibitor (PAI)-1. The parameters that increased have previously been associated with thrombosis in other patient groups, and might contribute to the increased rate of thrombosis observed in TPO-RA-treated ITP patients.


Assuntos
Plaquetas/metabolismo , Hemostáticos/uso terapêutico , Agregação Plaquetária/efeitos dos fármacos , Púrpura Trombocitopênica Idiopática , Receptores de Trombopoetina/agonistas , Plaquetas/patologia , Fibrinólise/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Hemostáticos/efeitos adversos , Humanos , Monócitos/metabolismo , Monócitos/patologia , Selectina-P/biossíntese , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Glicoproteínas da Membrana de Plaquetas/biossíntese , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/metabolismo , Púrpura Trombocitopênica Idiopática/patologia , Receptores de Trombopoetina/metabolismo , Trombose/induzido quimicamente , Trombose/metabolismo , Trombose/patologia
2.
Int J Lab Hematol ; 42(4): 371-379, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32202052

RESUMO

INTRODUCTION: Hemorrhage and infection are two main causes of death in patients with myelodysplastic syndromes (MDS), and it is becoming increasingly clear that platelet dysfunction can also affect the process of hemostasis and anti-infection. The aim of this study was to evaluate activation function and immune-related function of platelets in MDS. METHODS: We included 29 MDS patients and divided them into different subgroups (low-risk group and high-risk group; untreated group and treated group; pretransfusion group and post-transfusion group) according to IPSS-R score, hypomethylating agents (HMAs) therapy, and platelet transfusion history. Platelet light scatter properties, expression of CD41a, activation-associated phenotypes (CD62p and CD63), and immune-associated phenotypes (CD154 and TLR4) were detected by multiparameter flow cytometry. RESULTS: Expression of CD41a was decreased (P < .05), and no difference was found in platelet light scatter properties between MDS patients and healthy subjects (P > .05). Significantly decreased expression frequency and intensity of activation phenotype CD63 were found in patients with MDS (P < .05). Low-risk MDS showed lower expression frequency while high-risk MDS showed reduced mean fluorescence intensity (MFI) of CD63. Decreased expression of CD154 and TLR4 was found in MDS patients (P < .05) which was significantly elevated after HMAs therapy (P < .05). Particularly, MFI of CD154 and TLR4 reduced in high-risk MDS patients (P < .05). CONCLUSION: Myelodysplastic syndromes patients displayed defective expression of both activation- and immune-associated platelet phenotypes, with differential mechanisms between low-risk and high-risk groups regarding phenotype alterations. The findings confirmed impaired platelet phenotypes in MDS which may assist in the diagnosis and identification of MDS patients.


Assuntos
Plaquetas/metabolismo , Regulação da Expressão Gênica , Síndromes Mielodisplásicas/sangue , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Plaquetas/imunologia , Plaquetas/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/patologia , Síndromes Mielodisplásicas/terapia , Glicoproteínas da Membrana de Plaquetas/imunologia
3.
Molecules ; 24(19)2019 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-31561469

RESUMO

Atroxlysin-III (Atr-III) was purified from the venom of Bothrops atrox. This 56-kDa protein bears N-linked glycoconjugates and is a P-III hemorrhagic metalloproteinase. Its cDNA-deduced amino acid sequence reveals a multidomain structure including a proprotein, a metalloproteinase, a disintegrin-like and a cysteine-rich domain. Its identity with bothropasin and jararhagin from Bothrops jararaca is 97% and 95%, respectively. Its enzymatic activity is metal ion-dependent. The divalent cations, Mg2+ and Ca2+, enhance its activity, whereas excess Zn2+ inhibits it. Chemical modification of the Zn2+-complexing histidine residues within the active site by using diethylpyrocarbonate (DEPC) inactivates it. Atr-III degrades plasma fibronectin, type I-collagen, and mainly the α-chains of fibrinogen and fibrin. The von Willebrand factor (vWF) A1-domain, which harbors the binding site for GPIb, is not hydrolyzed. Platelets interact with collagen via receptors for collagen, glycoprotein VI (GPVI), and α2ß1 integrin. Neither the α2ß1 integrin nor its collagen-binding A-domain is fragmented by Atr-III. In contrast, Atr-III cleaves glycoprotein VI (GPVI) into a soluble ~55-kDa fragment (sGPVI). Thereby, it inhibits aggregation of platelets which had been stimulated by convulxin, a GPVI agonist. Selectively, Atr-III targets GPVI antagonistically and thus contributes to the antithrombotic effect of envenomation by Bothrops atrox.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Venenos de Crotalídeos/enzimologia , Crotalinae , Metaloproteases/farmacologia , Glicoproteínas da Membrana de Plaquetas/biossíntese , Sequência de Aminoácidos , Animais , Crotalinae/metabolismo , Matriz Extracelular , Metaloproteases/química , Metaloproteases/genética , Metaloproteases/isolamento & purificação , Modelos Moleculares , Filogenia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/química , Conformação Proteica , Proteólise , Relação Estrutura-Atividade
4.
Platelets ; 28(5): 498-508, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27778530

RESUMO

Platelet storage is associated with deleterious changes leading to the loss of platelet reactivity and response. During storage, platelets experience increased expression and shedding of P-selectin and CD40L as specific markers of platelet activation, whereas GPIbα decreases due to ectodomain shedding. As an important adhesive receptor, GPVI contributes significantly to thrombus formation while its expression and shedding levels during storage of platelet products have not been well characterized yet. This study investigated the modulation of GPVI during platelet storage. For this study, samples obtained from 10 PRP-platelet concentrates (PCs) were subjected to flow-cytometry analysis to examine the expression of platelet activation markers and GPVI on days 1, 3, and 5 post-storage. To examine the levels of etcodomain shedding of these molecules, microparticle (MP)-free supernatants were also analyzed by either ELISA or Western blot methods. According to results, the expression levels of P-selectin and CD40L as well as the amounts of their soluble forms significantly increased during storage. The expression of GPIbα and GPVI decreased whereas their shedding significantly increased post-storage. The expression and shedding levels of these two receptors were significantly correlated. Negative correlations between the expressions of GPIbα or GPVI and P-selectin have been observed whereas their shedding levels were significantly relevant together. In a control study, the use of biotinylated platelet resuspended in Tyrode's buffer in the presence of ionophore with/without EDTA, confirmed the role of calcium in receptors shedding. In citrated PRP-PCs, recalcification of platelets also enhanced shedding levels of both GPIbα and GPVI. Intriguingly, the shedding levels of GPVI in stored PRP-PCs were much higher than those of ionophore-treated controls obtained from washed platelets. The ratios of sGPVI in stored platelet to ionophore-treated controls were also at least six times higher than those of GPIbα during storage. In conclusion, here we showed significant decreases of GPVI expression associated with its increasing levels of shedding during storage, suggesting GPVI as a valid marker of platelet storage lesion. Importantly, we found higher levels of GPVI shedding in stored platelets than those of ionophore-treated non-stored control samples. This suggests whereas platelet receptor shedding is mainly modulated by calcium-dependent signals, either platelet-surface interactions with the container walls during storage or induced shear stress under long-term agitation, might be also involved in the excessive shedding of GPVI during the storage of PCs.


Assuntos
Plaquetas/metabolismo , Preservação de Sangue , Regulação da Expressão Gênica , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/biossíntese , Biomarcadores/metabolismo , Plaquetas/citologia , Citometria de Fluxo , Humanos , Domínios Proteicos
5.
Int J Oncol ; 49(1): 181-8, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27176648

RESUMO

Platelet-activating factor (PAF) and its receptor (PAFR), have been reported to participate in many cellular processes of cancer. However, little is known about their function in prostate cancer. In the present study, we found that PAFR was overexpressed in prostate cancer cells. PAF stimulation dose-dependently promoted the invasion, migration and growth of prostate cancer cells in vitro, while knockdown of PAFR inhibited the effect of PAF on prostate cancer cells. We further found that PAFR promoted prostate cancer cell growth and metastasis in vivo. Moreover, we found that PAFR activation increased MMP-3 expression and decreased E-cadherin expression of prostate cancer cells in vitro and in vivo. Finally, we found that PAFR time-dependently induced activation of ERK1/2, and ERK1/2 pathway contributed to PAFR-mediated prostate cancer cell invasion, migration and growth. Together, our findings demonstrate that PAFR can activate ERK1/2 pathway, and subsequently increase MMP-3 expression and decrease E-cadherin expression, which finally promote prostate cancer cell growth, invasion and metastasis. Thus, PAFR may act as a potential target for therapeutic use of prostate cancer.


Assuntos
Caderinas/biossíntese , Proteínas de Transporte/genética , Metaloproteinase 3 da Matriz/biossíntese , Glicoproteínas da Membrana de Plaquetas/biossíntese , Neoplasias da Próstata/genética , Receptores Acoplados a Proteínas G/biossíntese , Caderinas/genética , Movimento Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Metaloproteinase 3 da Matriz/genética , Invasividade Neoplásica/genética , Fosforilação , Glicoproteínas da Membrana de Plaquetas/genética , Neoplasias da Próstata/patologia , Receptores Acoplados a Proteínas G/genética
6.
Respirology ; 21(3): 504-10, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26662379

RESUMO

BACKGROUND AND OBJECTIVE: PAFr is a cell adhesion site for specific bacteria, notably non-typeable Haemophilus influenzae (NTHi) and Streptococcus pneumoniae. We previously published that PAFr expression is significantly upregulated in the large airways of smokers, especially in COPD. We have now investigated PAFr expression in the epithelium and Rbm of small airways and in the alveolar compartment in smokers and patients with both COPD and small airway disease. METHODS: We evaluated PAFr expression cross-sectionally in resected lung tissue from: eight smokers with normal lung function (NLFS); 10 with smoking-related small airway narrowing only; eight COPD smokers; 10 COPD ex-smokers, and compared these with nine control tissues. Anti-PAFr immunostaining was quantified using computer-aided image analysis. RESULTS: Significantly increased PAFr expression in small airway epithelium of all clinical groups was found compared with controls (P < 0.01). Moreover, epithelial PAFr expression was upregulated in COPD smokers compared with NLFS (P < 0.05), but not when compared with COPD ex-smokers or patients with only small airways disease. Smoking history (pack-year) correlated significantly with PAFr expression in the currently smoking individuals, especially in NLFS (r = 0.9; P < 0.002). An increase above normal in PAFr-expressing cells in the airway epithelial Rbm was only significant in COPD smokers (P < 0.007). An upregulation of PAFr-expressing cell in alveolar epithelium was uniformly found in all clinical groups compared with normal control (P < 0.01). CONCLUSION: Epithelial PAFr expression is upregulated in small airways and alveoli in smokers and COPD. Increased expression of PAFr could be crucial in facilitating acute and chronic respiratory infection with specific respiratory pathogens.


Assuntos
Plaquetas/metabolismo , Regulação da Expressão Gênica , Glicoproteínas da Membrana de Plaquetas/genética , Alvéolos Pulmonares/metabolismo , Doença Pulmonar Obstrutiva Crônica/genética , Receptores Acoplados a Proteínas G/genética , Fumar/genética , Regulação para Cima , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Glicoproteínas da Membrana de Plaquetas/biossíntese , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , RNA/genética , Receptores Acoplados a Proteínas G/biossíntese , Mucosa Respiratória/metabolismo , Fumar/metabolismo , Ativação Transcricional
7.
Cell Microbiol ; 16(8): 1244-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24612488

RESUMO

ExoU is an important virulence factor in acute Pseudomonas aeruginosa infections. Here, we unveiled the mechanisms of ExoU-driven NF-κB activation by using human airway cells and mice infected with P. aeruginosa strains. Several approaches showed that PAFR was crucially implicated in the activation of the canonical NF-κB pathway. Confocal microscopy of lungs from infected mice revealed that PAFR-dependent NF-κB activation occurred mainly in respiratory epithelial cells, and reduced p65 nuclear translocation was detected in mice PAFR-/- or treated with the PAFR antagonist WEB 2086. Several evidences showed that ExoU-induced NF-κB activation regulated PAFR expression. First, ExoU increased p65 occupation of PAFR promoter, as assessed by ChIP. Second, luciferase assays in cultures transfected with different plasmid constructs revealed that ExoU promoted p65 binding to the three κB sites in PAFR promoter. Third, treatment of cell cultures with the NF-κB inhibitor Bay 11-7082, or transfection with IκBα negative-dominant, significantly decreased PAFR mRNA. Finally, reduction in PAFR expression was observed in mice treated with Bay 11-7082 or WEB 2086 prior to infection. Together, our data demonstrate that ExoU activates NF-κB by PAFR signalling, which in turns enhances PAFR expression, highlighting an important mechanism of amplification of response to this P. aeruginosa toxin.


Assuntos
Proteínas de Bactérias/metabolismo , Fator de Ativação de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Pseudomonas aeruginosa/patogenicidade , Receptores Acoplados a Proteínas G/genética , Fator de Transcrição RelA/metabolismo , Animais , Azepinas/farmacologia , Toxinas Bacterianas/metabolismo , Linhagem Celular , Ativação Enzimática , Feminino , Regulação da Expressão Gênica , Humanos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Fator de Ativação de Plaquetas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/biossíntese , Regiões Promotoras Genéticas , Ligação Proteica , Infecções por Pseudomonas/patologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/genética , Triazóis/farmacologia
8.
Infect Immun ; 82(2): 587-93, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478074

RESUMO

Streptococcus pneumoniae is the leading cause of respiratory infection worldwide. Although oral hygiene has been considered a risk factor for developing pneumonia, the relationship between oral bacteria and pneumococcal infection is unknown. In this study, we examined the synergic effects of Prevotella intermedia, a major periodontopathic bacterium, on pneumococcal pneumonia. The synergic effects of the supernatant of P. intermedia (PiSup) on pneumococcal pneumonia were investigated in mice, and the stimulation of pneumococcal adhesion to human alveolar (A549) cells by PiSup was assessed. The effects of PiSup on platelet-activating factor receptor (PAFR) transcript levels in vitro and in vivo were analyzed by quantitative real-time PCR, and the differences between the effects of pneumococcal infection induced by various periodontopathic bacterial species were verified in mice. Mice inoculated with S. pneumoniae plus PiSup exhibited a significantly lower survival rate, higher bacterial loads in the lungs, spleen, and blood, and higher inflammatory cytokine levels in the bronchoalveolar lavage fluid (macrophage inflammatory protein 2 and tumor necrosis factor alpha) than those infected without PiSup. In A549 cells, PiSup increased pneumococcal adhesion and PAFR transcript levels. PiSup also increased lung PAFR transcript levels in mice. Similar effects were not observed in the supernatants of Porphyromonas gingivalis or Fusobacterium nucleatum. Thus, P. intermedia has the potential to induce severe bacteremic pneumococcal pneumonia with enhanced pneumococcal adhesion to lower airway cells.


Assuntos
Bacteriemia/patologia , Interações Microbianas , Glicoproteínas da Membrana de Plaquetas/biossíntese , Pneumonia Pneumocócica/complicações , Pneumonia Pneumocócica/patologia , Prevotella intermedia/patogenicidade , Receptores Acoplados a Proteínas G/biossíntese , Streptococcus pneumoniae/patogenicidade , Animais , Bacteriemia/microbiologia , Aderência Bacteriana , Linhagem Celular , Células Epiteliais/microbiologia , Fusobacterium nucleatum/patogenicidade , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Glicoproteínas da Membrana de Plaquetas/genética , Pneumonia Pneumocócica/microbiologia , Porphyromonas gingivalis/patogenicidade , Prevotella intermedia/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/genética , Streptococcus pneumoniae/fisiologia , Análise de Sobrevida , Regulação para Cima
9.
Pancreas ; 41(8): 1319-24, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22617709

RESUMO

OBJECTIVES: The aim of this study was to assess the functional state of platelets in patients with mild acute pancreatitis and severe acute pancreatitis (S-AP). METHODS: The number of platelets and their morphological parameters were measured with Advia 2120. ß-Thromboglobulin and platelet factor 4 concentrations were determined by enzyme-linked immunosorbent assay method. To evaluate the expression of platelet glycoproteins, flow cytometry method was used. RESULTS: At the time of admission, a multiparameter evaluation of the platelets' function in AP patients showed enhanced platelet activation, which was reflected by an increase in the number of large platelets, concentration of degranulation markers (platelet factor 4 and ß-thromboglobulin), expression of glycoprotein (Gp) IIb/IIIa, and decreased mean platelet component. Only in S-AP patients at day 1 a decreased number of platelets and high expression of P-selectin and GpIa were observed, which may suggest their prognostic value. At day 30, the procoagulation state was still present in S-AP patients, because of increased platelets and number of large platelets as well as high GpIIb/IIIa expression. CONCLUSIONS: These results may indicate an important role of platelet activation in the pathogenesis of acute pancreatitis and the development of complications in S-AP.


Assuntos
Plaquetas/fisiologia , Pancreatite/sangue , Ativação Plaquetária/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/uso terapêutico , Plaquetas/efeitos dos fármacos , Ciprofloxacina/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Masculino , Metronidazol/uso terapêutico , Pessoa de Meia-Idade , Selectina-P/biossíntese , Pancreatite/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Fator Plaquetário 4/análise , Glicoproteínas da Membrana de Plaquetas/biossíntese , Prognóstico , Índice de Gravidade de Doença , beta-Tromboglobulina/análise
10.
Mediators Inflamm ; 2012: 175408, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22570511

RESUMO

Melanoma cells express the platelet-activating factor receptor (PAFR) and, thus, respond to PAF, a bioactive lipid produced by both tumour cells and those in the tumour microenvironment such as macrophages. Here, we show that treatment of a human melanoma SKmel37 cell line with cisplatin led to increased expression of PAFR and its accumulation. In the presence of exogenous PAF, melanoma cells were significantly more resistant to cisplatin-induced cell death. Inhibition of PAFR-dependent signalling pathways by a PAFR antagonist (WEB2086) showed chemosensitisation of melanoma cells in vitro. Nude mice were inoculated with SKmel37 cells and treated with cisplatin and WEB2086. Animals treated with both agents showed significantly decreased tumour growth compared to the control group and groups treated with only one agent. PAFR accumulation and signalling are part of a prosurvival program of melanoma cells, therefore constituting a promising target for combination therapy for melanomas.


Assuntos
Antineoplásicos/farmacologia , Azepinas/farmacologia , Regulação Neoplásica da Expressão Gênica , Melanoma/metabolismo , Glicoproteínas da Membrana de Plaquetas/biossíntese , Receptores Acoplados a Proteínas G/biossíntese , Triazóis/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azepinas/administração & dosagem , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Feminino , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transdução de Sinais , Triazóis/administração & dosagem
11.
Blood ; 119(18): 4311-20, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22431567

RESUMO

Ligand-induced ectodomain shedding of glycoprotein VI (GPVI) is a metalloproteinase-dependent event. We examined whether shear force, in the absence of GPVI ligand, was sufficient to induce shedding of GPVI. Human-citrated platelet-rich plasma or washed platelets were subjected to increasing shear rates in a cone-plate viscometer, and levels of intact and cleaved GPVI were examined by Western blot and ELISA. Pathophysiologic shear rates (3000-10 000 seconds(-1)) induced platelet aggregation and metalloproteinase-dependent appearance of soluble GPVI ectodomain, and GPVI platelet remnant. Shedding of GPVI continued after transient exposure to shear. Blockade of α(IIb)ß(3), GPIbα, or intracellular signaling inhibited shear-induced platelet aggregation but minimally affected shear-induced shedding of GPVI. Shear-induced GPVI shedding also occurred in platelet-rich plasma or washed platelets isolated from a von Willebrand disease type 3 patient with no detectable VWF, implying that shear-induced activation of platelet metalloproteinases can occur in the absence of GPVI and GPIbα ligands. Significantly elevated levels of sGPVI were observed in 10 patients with stable angina pectoris, with well-defined single vessel coronary artery disease and mean intracoronary shear estimates at 2935 seconds(-1) (peak shear, 19 224 seconds(-1)). Loss of GPVI in platelets exposed to shear has potential implications for the stability of a forming thrombus at arterial shear rates.


Assuntos
Plaquetas/química , Estenose Coronária/sangue , Hemorreologia , Glicoproteínas da Membrana de Plaquetas/química , Estresse Mecânico , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/fisiologia , Proteína ADAM10 , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/fisiologia , Angina Estável/sangue , Viscosidade Sanguínea , Colágeno/fisiologia , Estenose Coronária/genética , Dipeptídeos/farmacologia , Regulação para Baixo , Feminino , Humanos , Ácidos Hidroxâmicos/farmacologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Pessoa de Meia-Idade , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas , Glicoproteínas da Membrana de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/genética , Plasma Rico em Plaquetas , Estrutura Terciária de Proteína , Doença de von Willebrand Tipo 3/sangue
12.
J Thromb Haemost ; 10(6): 1152-64, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22448969

RESUMO

BACKGROUND: Bone-marrow-derived progenitor cells are important in myocardial repair mechanisms following prolonged ischemia. Cell-based therapy of diseased myocardium is limited by a low level of tissue engraftment. OBJECTIVES: The aim of this study was the development of the bifunctional protein αCD133-glycoprotein (GP)VI as an effective treatment for supporting vascular and myocardial repair mechanisms. RESULTS: We have generated and characterized a bifunctional molecule (αCD133-GPVI) that binds both to the subendothelium of the injured microvasculature and to CD133(+) progenitor cells with high affinity. αCD133-GPVI enhances progenitor cell adhesion to extracellular matrix proteins and differentiation into mature endothelial cells. In vivo studies showed that αCD133-GPVI favors adhesion of circulating progenitor cells to the injured vessel wall (intravital microscopy). Also, treatment of mice undergoing experimental myocardial infarction with αCD133-GPVI-labeled progenitor cells reduces infarction size and preserves myocardial function. CONCLUSIONS: The bifunctional trapping protein αCD133-GPVI represents a novel and promising therapeutic option for limiting heart failure of the ischemic myocardium.


Assuntos
Antígenos CD/imunologia , Células Endoteliais/transplante , Terapia Genética , Glicoproteínas/imunologia , Infarto do Miocárdio/terapia , Miocárdio/patologia , Peptídeos/imunologia , Glicoproteínas da Membrana de Plaquetas/biossíntese , Regeneração , Anticorpos de Cadeia Única/biossíntese , Transplante de Células-Tronco , Antígeno AC133 , Animais , Sítios de Ligação , Adesão Celular , Diferenciação Celular , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Infarto do Miocárdio/genética , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/imunologia , Miocárdio/metabolismo , Glicoproteínas da Membrana de Plaquetas/genética , Proteínas Recombinantes/biossíntese , Anticorpos de Cadeia Única/genética , Fatores de Tempo , Transfecção , Função Ventricular Esquerda
13.
Int Arch Allergy Immunol ; 158(1): 18-26, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22212397

RESUMO

BACKGROUND: Racemic albuterol is a 50:50 mixture of the (R)- and (S)-enantiomers of albuterol. Its clinical efficacy resides in the (R)-enantiomer (levalbuterol). Studies have shown that (S)-albuterol induces human bronchial smooth muscle cell (HBSMC) proliferation via a pathway linked to platelet-activating factor (PAF), but the underlying mechanism by which (S)-albuterol augments PAF effects is not clear. In this study, we compared effect of levalbuterol and (S)-albuterol on PAF receptor (PAFr)-mediated signaling and PAF metabolism by HBSMCs after incubation with the albuterol isomers. METHODS: PAF binding and inositol phosphate (IP(3)) release were studied on adherent cultured cells. PAFr protein expression was measured by Western blotting, PAF synthesis and catabolism were measured in membrane and cytosolic proteins of cells incubated with albuterol isomers. RESULTS: Compared to control conditions, (S)-albuterol increased PAF binding by 70% after 30 min of preincubation and by 150% after 24 h of preincubation. Levalbuterol had no effect on PAF binding under both conditions. (S)-albuterol also augmented PAF stimulation of IP(3) release, while levalbuterol and the racemic mixture had no effect. WEB 2170, a PAFr antagonist, inhibited the ability of (S)-albuterol to increase PAF binding or stimulate IP(3) release. (S)-albuterol stimulated PAFr protein expression. With PAF metabolism, (S)-albuterol treatment augmented PAF synthesis, but significantly inhibited PAF catabolism. CONCLUSIONS: Our data suggest that one mechanism by which (S)-albuterol stimulates HBSMC proliferation involves upregulation of PAFr-mediated effects including increased PAF synthesis and decreased PAF catabolism.


Assuntos
Albuterol/farmacologia , Brônquios/efeitos dos fármacos , Broncodilatadores/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Ativação de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Azepinas/farmacologia , Brônquios/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Miócitos de Músculo Liso/metabolismo , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/biossíntese , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/efeitos dos fármacos , Triazóis/farmacologia
14.
Cryobiology ; 63(3): 229-34, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21945819

RESUMO

The current study aims to optimize the compositions of platelet activation-inhibitors for a loading solution of lyophilizing protectants and to establish a series of perfect pretreatment methods for platelet lyophilization. The optimal combination of six kinds of inhibitors and loading solutions of lyophilizing protectants, including prostaglandin E1 (PGE1), adenosine, L-arginine, phyticacid, bivalirudin, and cilostazol, was analyzed using the orthogonal experimental design. The values of the expression rates of p-selectin (CD62p) and platelet membrane glycoprotein (PAC-1), as well as of platelet and mean platelet volume (MPV), were selected as indices of platelet activation. The values of CD62p and Pac-1 induced by thrombin were determined as indices of platelet reactivity. The maximal aggregation and slide platelet aggregation test (SPAT) induced by the inducer were calculated as indices of the aggregation function of platelets. Level I of the loading condition factor had no adverse action on MPV, CD62p, PAC-1, SPAT, and the maximum platelet aggregation rate. Level II of factors PGE1, L-arginine, phycicacid sodium, and Bivalirudin could inhibit the activation of platelets and enable them to retain their function. The results show that the optimal solution compounding was the third group. The loading solution, which includes plasma, 1 µM prostaglandin E1, 5 mM L-arginine, 0.5 mM phyticacid, and 0.5 µM bivalirudin, could prevent the activation damage of platelets before lyophilization.


Assuntos
Plaquetas/efeitos dos fármacos , Preservação de Sangue/métodos , Excipientes/farmacologia , Liofilização/métodos , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Adenosina/metabolismo , Adenosina/farmacologia , Algoritmos , Alprostadil/metabolismo , Alprostadil/farmacologia , Arginina/metabolismo , Arginina/farmacologia , Biomarcadores/análise , Plaquetas/metabolismo , Tamanho Celular , Cilostazol , Hirudinas/metabolismo , Hirudinas/farmacologia , Humanos , Selectina-P/análise , Selectina-P/biossíntese , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Glicoproteínas da Membrana de Plaquetas/análise , Glicoproteínas da Membrana de Plaquetas/biossíntese , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Tetrazóis/metabolismo , Tetrazóis/farmacologia
15.
J Immunol ; 186(9): 5254-60, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21421855

RESUMO

Basophils are the rarest leukocytes in human blood, but they are now recognized as one of the most important immunomodulatory as well as effector cells in allergic inflammation. Leptin, a member of the IL-6 cytokine family, has metabolic effects as an adipokine, and it is also known to participate in the pathogenesis of inflammatory reactions. Because there is an epidemiologic relationship between obesity and allergy, we examined whether basophil functions are modified by leptin. We found that human basophils express leptin receptor (LepR) at both the mRNA and surface protein levels, which were upregulated by IL-33. Leptin exerted strong effects on multiple basophil functions. It induced a strong migratory response in human basophils, similar in potency to that of basophil-active chemokines. Also, leptin enhanced survival of human basophils, although its potency was less than that of IL-3. Additionally, CD63, a basophil activation marker expressed on the cell surface, was upregulated by leptin, an effect that was neutralized by blocking of LepR. Assessments of basophil degranulation and cytokine synthesis found that leptin showed a strong priming effect on human basophil degranulation in response to FcεRI aggregation and induced Th2, but not Th1, cytokine production by the cells. In summary, the present findings indicate that leptin may be a key molecule mediating the effects of adipocytes on inflammatory cells such as basophils by binding to LepR and activating the cellular functions, presumably exacerbating allergic inflammation.


Assuntos
Basófilos/imunologia , Degranulação Celular/imunologia , Movimento Celular/imunologia , Citocinas/biossíntese , Leptina/imunologia , Antígenos CD/biossíntese , Basófilos/citologia , Basófilos/metabolismo , Separação Celular , Sobrevivência Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Leptina/metabolismo , Glicoproteínas da Membrana de Plaquetas/biossíntese , Receptores para Leptina/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tetraspanina 30
16.
J Clin Invest ; 121(3): 941-55, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21285515

RESUMO

Allergic asthma is characterized by airway hyperresponsiveness, inflammation, and a cellular infiltrate dominated by eosinophils. Numerous epidemiological studies have related the exacerbation of allergic asthma with an increase in ambient inhalable particulate matter from air pollutants. This is because inhalable particles efficiently deliver airborne allergens deep into the airways, where they can aggravate allergic asthma symptoms. However, the cellular mechanisms by which inhalable particulate allergens (pAgs) potentiate asthmatic symptoms remain unknown, in part because most in vivo and in vitro studies exploring the pathogenesis of allergic asthma use soluble allergens (sAgs). Using a mouse model of allergic asthma, we found that, compared with their sAg counterparts, pAgs triggered markedly heightened airway hyperresponsiveness and pulmonary eosinophilia in allergen-sensitized mice. Mast cells (MCs) were implicated in this divergent response, as the differences in airway inflammatory responses provoked by the physical nature of the allergens were attenuated in MC-deficient mice. The pAgs were found to mediate MC-dependent responses by enhancing retention of pAg/IgE/FcεRI complexes within lipid raft­enriched, CD63(+) endocytic compartments, which prolonged IgE/FcεRI-initiated signaling and resulted in heightened cytokine responses. These results reveal how the physical attributes of allergens can co-opt MC endocytic circuitry and signaling responses to aggravate pathological responses of allergic asthma in mice.


Assuntos
Alérgenos/química , Asma/metabolismo , Hiper-Reatividade Brônquica/imunologia , Regulação da Expressão Gênica , Hipersensibilidade/metabolismo , Imunoglobulina E/metabolismo , Mastócitos/citologia , Poluentes Atmosféricos , Animais , Antígenos CD/biossíntese , Modelos Animais de Doenças , Endocitose , Inflamação , Lipídeos/química , Masculino , Microdomínios da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Glicoproteínas da Membrana de Plaquetas/biossíntese , Tetraspanina 30
17.
J Immunol ; 186(5): 2800-8, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270405

RESUMO

CXCR4 expression is critical for localization of centroblasts in the dark zone of germinal centers (GCs), and centrocytes downregulate CXCR4 and thus leave the dark zone to reside in the light zone. However, mechanisms governing CXCR4 downregulation on centrocytes are not known. In this study, we show that the amount of intracellular CXCR4 in centroblasts was similar to that in centrocytes, suggesting differential control of CXCR4 protein expression in these GC B cells. Restimulation of activated B cells with IL-21, which is a major cytokine produced by T follicular helper cells, accelerated CXCR4 internalization by inducing endocytosis-related GRK6 expression. Although CXCR4 expression was downregulated on GC B cells by IL-21 stimulation, CXCR4(low) centrocytes developed in the spleens of IL-21R-deficient mice, suggesting other mechanisms for downregulation. The level of CD63 (which recruits CXCR4 to late endosome in CD4 T cells) in centrocytes was more than that in centroblasts and was strikingly elevated in activated Bcl6-deficient B cells. Bcl6, a transcriptional repressor, was detected on the chromatin of the CD63 gene in resting B cells, therefore CD63 is a molecular target of Bcl6. Downregulation of CD63 mRNA in activated Bcl6-deficient B cells by small interfering RNA upregulated CXCR4 expression on the B cells. Furthermore, addition of Bcl6 inhibitor to activated B cell cultures increased CD63 mRNA expression in (and downregulated CXCR4 expression on) those activated B cells. Thus, CXCR4 can be downregulated on activated B cells by IL-21-induced endocytosis and CD63-mediated endosomal recruitment, and these mechanisms may contribute to downregulation of CXCR4 on centrocytes.


Assuntos
Antígenos CD/fisiologia , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Regulação para Baixo/imunologia , Interleucinas/fisiologia , Ativação Linfocitária/imunologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/biossíntese , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Subpopulações de Linfócitos B/citologia , Células Cultivadas , Endocitose/imunologia , Endossomos/imunologia , Endossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteínas da Membrana de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/genética , Receptores CXCR4/metabolismo , Tetraspanina 30
19.
Mol Genet Metab ; 101(4): 400-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20813571

RESUMO

Hypoxia augments PAF receptor (PAFr) binding and PAFr protein expression in venous SMC (SMC-PV). We compared effect of acute and prolonged hypoxia (pO(2)<40 torr) on PAFr-mediated responses in arterial SMC (SMC-PA) and SMC-PV. Cells were studied for 30 min (acute) or for 48 h (prolonged) hypoxia and compared to normoxic (pO(2) ~100 torr) conditions. PAF binding was quantified in fmol/10(6) cells (mean ± SEM). PAF binding in normoxia were SMC-PA, 5.2 ± 0.2 and in SMC-PV, 19.3 ± 1.1; values in acute hypoxia were SMC-PA, 7.7 ± 0.4 and in SMC-PV, 27.8 ± 1.7. Prolonged hypoxia produced 6-fold increase in binding in SMC-PA, but only 2-fold increase in SMC-PV, but binding in SMC-PV was still higher. Acute hypoxia augmented inositol phosphate release by 50% and 40% in SMC-PA and SMC-PV, respectively. During normoxia, PAFr mRNA expression by both cell types was similar, but expression in hypoxia by SMC-PA was greater. In SMC-PA, hypoxia and PAF augmented intracellular calcium flux. Re-exposure of cells to 30 min normoxia after 48 h hypoxia decreased binding by 45-60%, suggesting immediate down-regulation of hypoxia-induced PAFr-mediated effects. We speculate that re-oxygenation immediately reverses hypoxia effect probably due to oxygen tension-dependent reversibility of PAFr activation and suggest that exposure of the neonate to prolonged state of hypoxia will vilify oxygen exchange capacity of the neonatal lungs.


Assuntos
Hipóxia/metabolismo , Músculo Liso Vascular/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Regulação para Baixo , Feminino , Feto , Hipóxia/genética , Fosfatos de Inositol/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Oxigênio/metabolismo , Oxigênio/farmacologia , Fator de Ativação de Plaquetas/farmacologia , Glicoproteínas da Membrana de Plaquetas/biossíntese , Glicoproteínas da Membrana de Plaquetas/genética , Gravidez , Artéria Pulmonar/citologia , Artéria Pulmonar/metabolismo , Veias Pulmonares/citologia , Veias Pulmonares/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/genética , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...