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










Base de dados
Intervalo de ano de publicação
1.
Blood Adv ; 7(13): 3180-3191, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-36884296

RESUMO

This report identifies a novel variant form of the inherited bleeding disorder Glanzmann thrombasthenia, exhibiting only mild bleeding in a physically active individual. The platelets cannot aggregate ex vivo with physiologic agonists of activation, although microfluidic analysis with whole blood displays moderate ex vivo platelet adhesion and aggregation consistent with mild bleeding. Immunocytometry shows reduced expression of αIIbß3 on quiescent platelets that spontaneously bind/store fibrinogen, and activation-dependent antibodies (ligand-induced binding site-319.4 and PAC-1) report ß3 extension suggesting an intrinsic activation phenotype. Genetic analysis reveals a single F153Sß3 substitution within the ßI-domain from a heterozygous T556C nucleotide substitution of ITGB3 exon 4 in conjunction with a previously reported IVS5(+1)G>A splice site mutation with undetectable platelet messenger RNA accounting for hemizygous expression of S153ß3. F153 is completely conserved among ß3 of several species and all human ß-integrin subunits suggesting that it may play a vital role in integrin structure/function. Mutagenesis of αIIb-F153Sß3 also displays reduced levels of a constitutively activated αIIb-S153ß3 on HEK293T cells. The overall structural analysis suggests that a bulky aromatic, nonpolar amino acid (F,W)153ß3 is critical for maintaining the resting conformation of α2- and α1-helices of the ßI-domain because small amino acid substitutions (S,A) facilitate an unhindered inward movement of the α2- and α1-helices of the ßI-domain toward the constitutively active αIIbß3 conformation, while a bulky aromatic, polar amino acid (Y) hinders such movements and restrains αIIbß3 activation. The data collectively demonstrate that disruption of F153ß3 can significantly alter normal integrin/platelet function, although reduced expression of αIIb-S153ß3 may be compensated by a hyperactive conformation that promotes viable hemostasis.


Assuntos
Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Trombastenia , Humanos , Aminoácidos/genética , Células HEK293 , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/genética , Trombastenia/metabolismo
2.
Basic Res Cardiol ; 117(1): 24, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35469044

RESUMO

Impaired endothelium-dependent vasodilation has been suggested to be a key component of coronary microvascular dysfunction (CMD). A better understanding of endothelial pathways involved in vasodilation in human arterioles may provide new insight into the mechanisms of CMD. The goal of this study is to investigate the role of TRPV4, NOX4, and their interaction in human arterioles and examine the underlying mechanisms. Arterioles were freshly isolated from adipose and heart tissues obtained from 71 patients without coronary artery disease, and vascular reactivity was studied by videomicroscopy. In human adipose arterioles (HAA), ACh-induced dilation was significantly reduced by TRPV4 inhibitor HC067047 and by NOX 1/4 inhibitor GKT137831, but GKT137831 did not further affect the dilation in the presence of TRPV4 inhibitors. GKT137831 also inhibited TRPV4 agonist GSK1016790A-induced dilation in HAA and human coronary arterioles (HCA). NOX4 transcripts and proteins were detected in endothelial cells of HAA and HCA. Using fura-2 imaging, GKT137831 significantly reduced GSK1016790A-induced Ca2+ influx in the primary culture of endothelial cells and TRPV4-WT-overexpressing human coronary artery endothelial cells (HCAEC). However, GKT137831 did not affect TRPV4-mediated Ca2+ influx in non-phosphorylatable TRPV4-S823A/S824A-overexpressing HCAEC. In addition, treatment of HCAEC with GKT137831 decreased the phosphorylation level of Ser824 in TRPV4. Finally, proximity ligation assay (PLA) revealed co-localization of NOX4 and TRPV4 proteins. In conclusion, both TRPV4 and NOX4 contribute to ACh-induced dilation in human arterioles from patients without coronary artery disease. NOX4 increases TRPV4 phosphorylation in endothelial cells, which in turn enhances TRPV4-mediated Ca2+ entry and subsequent endothelium-dependent dilation in human arterioles.


Assuntos
Doença da Artéria Coronariana , Vasodilatação , Arteríolas/metabolismo , Doença da Artéria Coronariana/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Humanos , NADPH Oxidase 4/metabolismo , Fosforilação , Canais de Cátion TRPV , Vasodilatação/fisiologia
3.
J Thromb Haemost ; 19(10): 2417-2427, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34245090

RESUMO

BACKGROUND: Rotational thromboelastometry (ROTEM) has been commonly used to assess the viscoelastic properties of the blood clotting process in the clinic for patients with a hemostatic or prothrombotic disorder. OBJECTIVE: To evaluate the capability of ROTEM in assessing hemostatic properties in whole blood from various mouse models with genetic bleeding or clotting disease and the effect of factor VIII (FVIII) therapeutics in FVIIInull mice. METHODS: Mice with a genetic deficiency in either a coagulation factor or a platelet glycoprotein were used in this study. The properties of platelet- or plasma-FVIII were also assessed. Citrated blood from mice was recalcified and used for ROTEM analysis. RESULTS: We found that blood collected from the vena cava could generate reliable results from ROTEM analysis, but not blood collected from the tail vein, retro-orbital plexus, or submandibular vein. Age and sex did not significantly affect the hemostatic properties determined by ROTEM analysis. Clotting time (CT) and clot formation time (CFT) were significantly prolonged in FVIIInull (5- and 9-fold, respectively) and FIXnull (4- and 5.7-fold, respectively) mice compared to wild-type (WT)-C57BL/6J mice. Platelet glycoprotein (GP)IIIanull mice had significantly prolonged CFT (8.4-fold) compared to WT-C57BL/6J mice. CT and CFT in factor V (FV) Leiden mice were significantly shortened with an increased α-angle compared to WT-C57BL/6J mice. Using ROTEM analysis, we showed that FVIII expressed in platelets or infused into whole blood restored hemostasis of FVIIInull mice in a dose-dependent manner. CONCLUSION: ROTEM is a reliable and sensitive assay for assessing therapeutics on hemostatic properties in mouse models with a bleeding or clotting disorder.


Assuntos
Hemostáticos , Tromboelastografia , Animais , Modelos Animais de Doenças , Fator VIII/genética , Hemostasia , Humanos , Camundongos , Camundongos Endogâmicos C57BL
4.
Br J Pharmacol ; 178(3): 709-725, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33184836

RESUMO

BACKGROUND AND PURPOSE: The TRPV cation channels have emerged as important regulators of vascular tone. TRPV1 channels and endothelin-1 are independently associated with the pathophysiology of coronary vasospasm, but the relationship between their vasomotor functions remains unclear. We characterized the vasomotor function of TRPV1 channels in human arterioles and investigated regulation of their vasomotor function by endothelin-1. EXPERIMENTAL APPROACH: Human arterioles (mainly from adipose tissue) were threaded on two metal wires, equilibrated in a physiological buffer at 37°C and exposed to increasing concentrations of capsaicin, with or without SB366791 (TRPV1-selective inhibitor) or GF109203X (PKC-selective inhibitor). Some arterioles were pre-constricted with endothelin-1 or phenylephrine or high potassium buffer. TRPV1 mRNA and protein expression in human arteries were also assessed. KEY RESULTS: TRPV1 transcripts and proteins were detected in human resistance arteries. Capsaicin (1 µM) induced concentration-dependent constriction of endothelium-intact and endothelium-denuded human adipose arterioles (HAA), which was significantly inhibited by SB366791. Pre-constriction of HAA with endothelin-1, but not high potassium buffer or phenylephrine, significantly potentiated capsaicin (0.1 µM)-induced constriction. GF109203X significantly inhibited potentiation of capsaicin-induced constriction by endothelin-1. CONCLUSION AND IMPLICATIONS: TRPV1 channels are expressed in the human vasculature and affect vascular tone of human arterioles on activation. Their vasomotor function is modulated by endothelin-1, mediated in part by PKC. These findings reveal a novel interplay between endothelin-1 signalling and TRPV1 channels in human VSMC, adding to our understanding of the ion channel mechanisms that regulate human arteriolar tone and may also contribute to the pathophysiology of coronary vasospasm.


Assuntos
Endotelina-1 , Vasoconstrição , Tecido Adiposo , Arteríolas , Capsaicina/farmacologia , Humanos , Proteína Quinase C , Canais de Cátion TRPV
5.
J Biol Chem ; 293(14): 5307-5322, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29462784

RESUMO

Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable channel of the transient receptor potential (TRP) superfamily activated by diverse stimuli, including warm temperature, mechanical forces, and lipid mediators such as arachidonic acid (AA) and its metabolites. This activation is tightly regulated by protein phosphorylation carried out by various serine/threonine or tyrosine kinases. It remains poorly understood how phosphorylation differentially regulates TRPV4 activation in response to different stimuli. We investigated how TRPV4 activation by AA, an important signaling process in the dilation of coronary arterioles, is affected by protein kinase A (PKA)-mediated phosphorylation at Ser-824. Wildtype and mutant TRPV4 channels were expressed in human coronary artery endothelial cells (HCAECs). AA-induced TRPV4 activation was blunted in the S824A mutant but was enhanced in the phosphomimetic S824E mutant, whereas the channel activation by the synthetic agonist GSK1016790A was not affected. The low level of basal phosphorylation at Ser-824 was robustly increased by the redox signaling molecule hydrogen peroxide (H2O2). The H2O2-induced phosphorylation was accompanied by an enhanced channel activation by AA, and this enhanced response was largely abolished by PKA inhibition or S824A mutation. We further identified a potential structural context dependence of Ser-824 phosphorylation-mediated TRPV4 regulation involving an interplay between AA binding and the possible phosphorylation-induced rearrangements of the C-terminal helix bearing Ser-824. These results provide insight into how phosphorylation specifically regulates TRPV4 activation. Redox-mediated TRPV4 phosphorylation may contribute to pathologies associated with enhanced TRPV4 activity in endothelial and other systems.


Assuntos
Canais de Cátion TRPV/metabolismo , Canais de Cátion TRPV/fisiologia , Ácido Araquidônico/metabolismo , Canais de Cálcio/metabolismo , Células Cultivadas , Vasos Coronários/metabolismo , Cristalografia por Raios X , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células Endoteliais/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Fosforilação , Transdução de Sinais
6.
Blood ; 127(10): 1260-8, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26787735

RESUMO

Hematopoietic stem cells (HSCs) can be safely collected from the body, genetically modified, and re-infused into a patient with the goal to express the transgene product for an individual's lifetime. Hematologic defects that can be corrected with an allogeneic bone marrow transplant can theoretically also be treated with gene replacement therapy. Because some genetic disorders affect distinct cell lineages, researchers are utilizing HSC gene transfer techniques using lineage-specific endogenous gene promoters to confine transgene expression to individual cell types (eg, ITGA2B for inherited platelet defects). HSCs appear to be an ideal target for platelet gene therapy because they can differentiate into megakaryocytes which are capable of forming several thousand anucleate platelets that circulate within blood vessels to establish hemostasis by repairing vascular injury. Platelets play an essential role in other biological processes (immune response, angiogenesis) as well as diseased states (atherosclerosis, cancer, thrombosis). Thus, recent advances in genetic manipulation of megakaryocytes could lead to new and improved therapies for treating a variety of disorders. In summary, genetic manipulation of megakaryocytes has progressed to the point where clinically relevant strategies are being developed for human trials for genetic disorders affecting platelets. Nevertheless, challenges still need to be overcome to perfect this field; therefore, strategies to increase the safety and benefit of megakaryocyte gene therapy will be discussed.


Assuntos
Terapia Genética/métodos , Doenças Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Células Progenitoras de Megacariócitos/metabolismo , Megacariócitos/metabolismo , Animais , Doenças Hematológicas/genética , Doenças Hematológicas/metabolismo , Doenças Hematológicas/patologia , Humanos , Integrina alfa2/biossíntese , Integrina alfa2/genética , Células Progenitoras de Megacariócitos/patologia , Megacariócitos/patologia , Transgenes
7.
Cell Microbiol ; 17(7): 1021-36, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25604835

RESUMO

P66, a Borrelia burgdorferi surface protein with porin and integrin-binding activities, is essential for murine infection. The role of P66 integrin-binding activity in B. burgdorferi infection was investigated and found to affect transendothelial migration. The role of integrin binding, specifically, was tested by mutation of two amino acids (D205A,D207A) or deletion of seven amino acids (Del202-208). Neither change affected surface localization or channel-forming activity of P66, but both significantly reduced binding to αv ß3 . Integrin-binding deficient B. burgdorferi strains caused disseminated infection in mice at 4 weeks post-subcutaneous inoculation, but bacterial burdens were significantly reduced in some tissues. Following intravenous inoculation, the Del202-208 bacteria were below the limit of detection in all tissues assessed at 2 weeks post-inoculation, but bacterial burdens recovered to wild-type levels at 4 weeks post-inoculation. The delay in tissue colonization correlated with reduced migration of the Del202-208 strains across microvascular endothelial cells, similar to Δp66 bacteria. These results indicate that integrin binding by P66 is important to efficient dissemination of B. burgdorferi, which is critical to its ability to cause disease manifestations in incidental hosts and to its maintenance in the enzootic cycle.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Borrelia burgdorferi/fisiologia , Interações Hospedeiro-Patógeno , Integrina alfaVbeta3/metabolismo , Porinas/metabolismo , Substituição de Aminoácidos , Animais , Carga Bacteriana , Linhagem Celular , Células Endoteliais/microbiologia , Humanos , Camundongos Endogâmicos C3H , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Deleção de Sequência
8.
Blood ; 123(5): 753-7, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24335497

RESUMO

Megakaryocyte-specific transgene expression in patient-derived induced pluripotent stem cells (iPSCs) offers a new approach to study and potentially treat disorders affecting megakaryocytes and platelets. By using a Gp1ba promoter, we developed a strategy for achieving a high level of protein expression in human megakaryocytes. The feasibility of this approach was demonstrated in iPSCs derived from two patients with Glanzmann thrombasthenia (GT), an inherited platelet disorder caused by mutations in integrin αIIbß3. Hemizygous insertion of Gp1ba promoter-driven human αIIb complementary DNA into the AAVS1 locus of iPSCs led to high αIIb messenger RNA and protein expression and correction of surface αIIbß3 in megakaryocytes. Agonist stimulation of these cells displayed recovery of integrin αIIbß3 activation. Our findings demonstrate a novel approach to studying human megakaryocyte biology as well as functional correction of the GT defect, offering a potential therapeutic strategy for patients with diseases that affect platelet function.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Megacariócitos/metabolismo , Glicoproteínas de Membrana/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/genética , Transgenes , Expressão Gênica , Humanos , Complexo Glicoproteico GPIb-IX de Plaquetas , Regiões Promotoras Genéticas
9.
Nat Commun ; 4: 2773, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24253479

RESUMO

It is essential to improve therapies for controlling excessive bleeding in patients with haemorrhagic disorders. As activated blood platelets mediate the primary response to vascular injury, we hypothesize that storage of coagulation Factor VIII within platelets may provide a locally inducible treatment to maintain haemostasis for haemophilia A. Here we show that haematopoietic stem cell gene therapy can prevent the occurrence of severe bleeding episodes in dogs with haemophilia A for at least 2.5 years after transplantation. We employ a clinically relevant strategy based on a lentiviral vector encoding the ITGA2B gene promoter, which drives platelet-specific expression of human FVIII permitting storage and release of FVIII from activated platelets. One animal receives a hybrid molecule of FVIII fused to the von Willebrand Factor propeptide-D2 domain that traffics FVIII more effectively into α-granules. The absence of inhibitory antibodies to platelet-derived FVIII indicates that this approach may have benefit in patients who reject FVIII replacement therapies. Thus, platelet FVIII may provide effective long-term control of bleeding in patients with haemophilia A.


Assuntos
Plaquetas/fisiologia , Doenças do Cão/terapia , Fator VIII/genética , Terapia Genética/veterinária , Hemofilia A/veterinária , Hemostasia , Integrina alfa2/metabolismo , Animais , Doenças do Cão/genética , Cães , Regulação da Expressão Gênica/fisiologia , Terapia Genética/métodos , Hemofilia A/terapia , Humanos , Integrina alfa2/genética
10.
Semin Thromb Hemost ; 39(6): 642-55, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23929305

RESUMO

Glanzmann thrombasthenia (GT) is the principal inherited disease of platelets and the most commonly encountered disorder of an integrin. GT is characterized by spontaneous mucocutaneous bleeding and an exaggerated response to trauma caused by platelets that fail to aggregate when stimulated by physiologic agonists. GT is caused by quantitative or qualitative deficiencies of αIIbß3, an integrin coded by the ITGA2B and ITGB3 genes and which by binding fibrinogen and other adhesive proteins joins platelets together in the aggregate. Widespread genotyping has revealed that mutations spread across both genes, yet the reason for the extensive variation in both the severity and intensity of bleeding between affected individuals remains poorly understood. Furthermore, although genetic defects of ITGB3 affect other tissues with ß3 present as αvß3 (the vitronectin receptor), the bleeding phenotype continues to dominate. Here, we look in detail at mutations that affect (i) the ß-propeller region of the αIIb head domain and (ii) the membrane proximal disulfide-rich epidermal growth factor (EGF) domains of ß3 and which often result in spontaneous integrin activation. We also examine deep vein thrombosis as an unexpected complication of GT and look at curative procedures for the diseases, including allogeneic stem cell transfer and the potential for gene therapy.


Assuntos
Plaquetas/metabolismo , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/genética , Terapia Genética/métodos , Hemorragia/genética , Hemorragia/terapia , Humanos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transplante de Células-Tronco/métodos , Trombastenia/diagnóstico , Trombastenia/terapia , Transplante Autólogo
11.
J Am Heart Assoc ; 2(3): e000080, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23619744

RESUMO

BACKGROUND: Arachidonic acid (AA) and/or its enzymatic metabolites are important lipid mediators contributing to endothelium-derived hyperpolarizing factor (EDHF)-mediated dilation in multiple vascular beds, including human coronary arterioles (HCAs). However, the mechanisms of action of these lipid mediators in endothelial cells (ECs) remain incompletely defined. In this study, we investigated the role of the transient receptor potential vanilloid 4 (TRPV4) channel in AA-induced endothelial Ca(2+) response and dilation of HCAs. METHODS AND RESULTS: AA induced concentration-dependent dilation in isolated HCAs. The dilation was largely abolished by the TRPV4 antagonist RN-1734 and by inhibition of endothelial Ca(2+)-activated K(+) channels. In native and TRPV4-overexpressing human coronary artery ECs (HCAECs), AA increased intracellular Ca(2+) concentration ([Ca(2+)]i), which was mediated by TRPV4-dependent Ca(2+) entry. The AA-induced [Ca(2+)]i increase was inhibited by cytochrome P450 (CYP) inhibitors. Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. Apart from its effect on [Ca(2+)]i, AA induced endothelial hyperpolarization, and this effect was required for Ca(2+) entry through TRPV4. AA-induced and TRPV4-mediated Ca(2+) entry was also inhibited by the protein kinase A inhibitor PKI. TRPV4 exhibited a basal level of phosphorylation, which was inhibited by PKI. Patch-clamp studies indicated that AA activated TRPV4 single-channel currents in cell-attached and inside-out patches of HCAECs. CONCLUSIONS: AA dilates HCAs through a novel mechanism involving endothelial TRPV4 channel-dependent Ca(2+) entry that requires endothelial hyperpolarization, PKA-mediated basal phosphorylation of TRPV4, and direct activation of TRPV4 channels by AA.


Assuntos
Ácido Araquidônico/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/fisiologia , Arteríolas/citologia , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Células Cultivadas , Vasos Coronários/citologia , Endotélio Vascular/citologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
12.
Proc Natl Acad Sci U S A ; 108(23): 9583-8, 2011 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-21606353

RESUMO

Activated blood platelets mediate the primary response to vascular injury. Although molecular abnormalities of platelet proteins occur infrequently, taken collectively, an inherited platelet defect accounts for a bleeding diathesis in ≈1:20,000 individuals. One rare example of a platelet disorder, Glanzmann thrombasthenia (GT), is characterized by life-long morbidity and mortality due to molecular abnormalities in a major platelet adhesion receptor, integrin αIIbß3. Transfusion therapy is frequently inadequate because patients often generate antibodies to αIIbß3, leading to immune-mediated destruction of healthy platelets. In the most severe cases allogeneic bone marrow transplantation has been used, yet because of the risk of the procedure it has been limited to few patients. Thus, hematopoietic stem cell gene transfer was explored as a strategy to improve platelet function within a canine model for GT. Bleeding complications necessitated the use of a mild pretransplant conditioning regimen; therefore, in vivo drug selection was used to improve engraftment of autologously transplanted cells. Approximately 5,000 αIIbß3 receptors formed on 10% of platelets. These modest levels allowed platelets to adhere to αIIbß3's major ligand (fibrinogen), form aggregates, and mediate retraction of a fibrin clot. Remarkably, improved hemostatic function was evident, with ≤135-fold reduced blood loss, and improved buccal bleeding times decreased to 4 min for up to 5 y after transplant. One of four transplanted dogs developed a significant antibody response to αIIbß3 that was attenuated effectively with transient immune suppression. These results indicate that gene therapy could become a practical approach for treating inherited platelet defects.


Assuntos
Plaquetas/metabolismo , Doenças do Cão/terapia , Terapia Genética/métodos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/veterinária , Animais , Antígenos CD34/metabolismo , Tempo de Sangramento , Transplante de Células/métodos , Células Cultivadas , Doenças do Cão/genética , Cães , Citometria de Fluxo , Hemostasia , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/transplante , Mucosa Bucal/irrigação sanguínea , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/genética , Trombastenia/terapia , Transfecção , Transplante Autólogo
13.
Blood ; 117(18): 4946-52, 2011 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-21368290

RESUMO

Unfractionated heparin (UFH) is a widely used anticoagulant that has long been known to potentiate platelet responses to subthreshold doses of platelet agonists. UFH has been reported to bind and induce modest conformational changes in the major platelet integrin, αIIbß3, and induce minor changes in platelet morphology. The mechanism by which UFH elicits these platelet-activating effects, however, is not well understood. We found that both human and murine platelets exposed to UFH, either in solution or immobilized onto artificial surfaces, underwent biochemical and morphologic changes indicative of a potentiated state, including phosphorylation of key cytosolic signaling molecules and cytoskeletal changes leading to cell spreading. Low molecular weight heparin and the synthetic pentasaccharide, fondaparinux, had similar platelet-potentiating effects. Human or mouse platelets lacking functional integrin αIIbß3 complexes and human platelets pretreated with the fibrinogen receptor antagonists eptifibatide or abciximab failed to become potentiated by heparin, demonstrating that heparin promotes platelet responsiveness via its ability to initiate αIIbß3-mediated outside-in signaling. Taken together, these data provide novel insights into the mechanism by which platelets become activated after exposure to heparin and heparin-coated surfaces, and suggest that currently used glycoprotein IIb-IIIa inhibitors may be effective inhibitors of nonimmune forms of heparin-induced platelet activation.


Assuntos
Heparina/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Abciximab , Adulto , Animais , Anticorpos Monoclonais/administração & dosagem , Anticoagulantes/administração & dosagem , Sinergismo Farmacológico , Eptifibatida , Fondaparinux , Heparina/administração & dosagem , Humanos , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Técnicas In Vitro , Camundongos , Modelos Biológicos , Peptídeos/administração & dosagem , Inibidores da Agregação Plaquetária/administração & dosagem , Polissacarídeos/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
14.
Am J Physiol Heart Circ Physiol ; 298(2): H466-76, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19966050

RESUMO

The transient receptor potential vallinoid type 4 (TRPV4) channel has been implicated in the endothelial shear response and flow-mediated dilation, although the precise functions of this channel remain poorly understood. In the present study, we investigated the role of TRPV4 in shear stress-induced endothelial Ca(2+) entry and the potential link between this signaling response and relaxation of small resistance arteries. Using immunohistochemical analysis and RT-PCR, we detected strong expression of TRPV4 protein and mRNA in the endothelium in situ and endothelial cells freshly isolated from mouse small mesenteric arteries. The selective TRPV4 agonist GSK1016790A increased endothelial Ca(2+) and induced potent relaxation of small mesenteric arteries from wild-type (WT) but not TRPV4(-/-) mice. Luminal flow elicited endothelium-dependent relaxations that involved both nitric oxide and EDHFs. Both nitric oxide and EDHF components of flow-mediated relaxation were markedly reduced in TRPV4(-/-) mice compared with WT controls. Using a fura-2/Mn(2+) quenching assay, shear was observed to produce rapid Ca(2+) influx in endothelial cells, which was markedly inhibited by the TRPV4 channel blocker ruthenium red and TRPV4-specific short interfering RNA. Flow elicited a similar TRPV4-mediated Ca(2+) entry in HEK-293 cells transfected with TRPV4 channels but not in nontransfected cells. Collectively, these data indicate that TRPV4 may be a potential candidate of mechanosensitive channels in endothelial cells through which the shear stimulus is transduced into Ca(2+) signaling, leading to the release of endothelial relaxing factors and flow-mediated dilation of small resistance arteries.


Assuntos
Cálcio/metabolismo , Endotélio Vascular/metabolismo , Artérias Mesentéricas/metabolismo , Estresse Mecânico , Canais de Cátion TRPV/metabolismo , Vasodilatação/fisiologia , Animais , Fenômenos Biomecânicos , Sinalização do Cálcio/fisiologia , Linhagem Celular , Humanos , Rim/citologia , Rim/metabolismo , Leucina/análogos & derivados , Leucina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Óxido Nítrico/metabolismo , Sulfonamidas/farmacologia , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/genética , Transfecção
15.
J Clin Invest ; 118(8): 2929-38, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18654666

RESUMO

Fetomaternal alloimmune thrombocytopenia (FMAIT) is caused by maternal generation of antibodies specific for paternal platelet antigens and can lead to fetal intracranial hemorrhage. A SNP in the gene encoding integrin beta3 causes a clinically important maternal-paternal antigenic difference; Leu33 generates the human platelet antigen 1a (HPA-1a), whereas Pro33 generates HPA-1b. As a potential treatment to prevent fetal intracranial hemorrhage in HPA-1a alloimmunized pregnancies, we generated an antibody that blocks the binding of maternal HPA-1a-specific antibodies to fetal HPA-1a1b platelets by combining a high-affinity human HPA-1a-specific scFv (B2) with an IgG1 constant region modified to minimize Fcgamma receptor-dependent platelet destruction (G1Deltanab). B2G1Deltanab saturated HPA-1a+ platelets and substantially inhibited binding of clinical HPA-1a-specific sera to HPA-1a+ platelets. The response of monocytes to B2G1Deltanab-sensitized platelets was substantially less than their response to unmodified B2G1, as measured by chemiluminescence. In addition, B2G1Deltanab inhibited chemiluminescence induced by B2G1 and HPA-1a-specific sera. In a chimeric mouse model, B2G1 and polyclonal Ig preparations from clinical HPA-1a-specific sera reduced circulating HPA-1a+ platelets, concomitant with transient thrombocytopenia. As the Deltanab constant region is uninformative in mice, F(ab')2 B2G1 was used as a proof of principle blocking antibody and prevented the in vivo platelet destruction seen with B2G1 and polyclonal HPA-1a-specific antibodies. These results provide rationale for human clinical studies.


Assuntos
Anticorpos/imunologia , Antígenos de Plaquetas Humanas/imunologia , Trombocitopenia Neonatal Aloimune/terapia , Anticorpos/metabolismo , Antígenos de Plaquetas Humanas/genética , Plaquetas/imunologia , Plaquetas/metabolismo , Feminino , Humanos , Imunoglobulina G/química , Imunoglobulina G/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/metabolismo , Recém-Nascido , Integrina beta3 , Modelos Moleculares , Mutação , Contagem de Plaquetas , Gravidez , Ligação Proteica , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Proteínas Recombinantes/imunologia , Trombocitopenia Neonatal Aloimune/etiologia
16.
Blood ; 112(7): 2713-21, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18495954

RESUMO

Although genetic induction of factor VIII (FVIII) expression in platelets can restore hemostasis in hemophilia A mice, this approach has not been studied in the clinical setting of preexisting FVIII inhibitory antibodies to determine whether such antibodies would affect therapeutic engraftment. We generated a line of transgenic mice (2bF8) that express FVIII only in platelets using the platelet-specific alphaIIb promoter and bred this 2bF8 transgene into a FVIII(null) background. Bone marrow (BM) from heterozygous 2bF8 transgenic (2bF8(tg+/-)) mice was transplanted into immunized FVIII(null) mice after lethal or sublethal irradiation. After BM reconstitution, 85% of recipients survived tail clipping when the 1100-cGy (myeloablative) regimen was used, 85.7% of recipients survived when 660-cGy (nonmyeloablative) regimens were used, and 60% of recipients survived when the recipients were conditioned with 440 cGy. Our further studies showed that transplantation with 1% to 5% 2bF8(tg+/-) BM cells still improved hemostasis in hemophilia A mice with inhibitors. These results demonstrate that the presence of FVIII-specific immunity in recipients does not negate engraftment of 2bF8 genetically modified hematopoietic stem cells, and transplantation of these hematopoietic stem cells can efficiently restore hemostasis to hemophilic mice with preexisting inhibitory antibodies under either myeloablative or nonmyeloablative regimens.


Assuntos
Plaquetas/metabolismo , Fator VIII/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Hemofilia A/genética , Hemostasia , Imunidade , Animais , Bioensaio , Plaquetas/efeitos dos fármacos , Fator VIII/genética , Fator VIII/imunologia , Terapia Genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hemofilia A/terapia , Imunização , Camundongos , Agonistas Mieloablativos/farmacologia , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transformação Genética , Transgenes , Condicionamento Pré-Transplante , Transplante Isogênico
17.
J Clin Invest ; 116(7): 1974-82, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16823491

RESUMO

Inhibitory immune response to exogenously infused factor VIII (FVIII) is a major complication in the treatment of hemophilia A. Generation of such inhibitors has the potential to disrupt gene therapy for hemophilia A. We explore what we believe to be a novel approach to overcome this shortcoming. Human B-domain-deleted FVIII (hBDDFVIII) was expressed under the control of the platelet-specific alphaIIb promoter in platelets of hemophilic (FVIIInull) mice to create 2bF8trans mice. The FVIII transgene product was stored in platelets and released at the site of platelet activation. In spite of the lack of FVIII in the plasma of 2bF8trans mice, the bleeding phenotype of FVIIInull mice was corrected. More importantly, the bleeding phenotype was corrected in the presence of high inhibitory antibody titers introduced into the mice by infusion or by spleen cell transfer from recombinant hBDDFVIII-immunized mice. Our results demonstrate that this approach to the targeted expression of FVIII in platelets has the potential to correct hemophilia A, even in the presence of inhibitory immune responses to infused FVIII.


Assuntos
Anticorpos/imunologia , Plaquetas/fisiologia , Fator VIII/metabolismo , Fator VIII/uso terapêutico , Hemofilia A , Animais , Fator VIII/genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/terapia , Hemostasia/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Fenótipo , Ativação Plaquetária , Transgenes , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
18.
Blood ; 106(8): 2671-9, 2005 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15972454

RESUMO

Integrins mediate the adhesion of cells to each other and to the extracellular matrix during development, immunity, metastasis, thrombosis, and wound healing. Molecular defects in either the alpha- or beta-subunit can disrupt integrin synthesis, assembly, and/or binding to adhesive ligands. This is exemplified by the bleeding disorder, Glanzmann thrombasthenia (GT), where abnormalities of the platelet-specific integrin, alphaIIbbeta3, prevent platelet aggregation following vascular injury. We previously used a retrovirus vector containing a cDNA cassette encoding human integrin beta3 to restore integrin alphaIIbbeta3 on the surface of megakaryocytes derived from peripheral blood stem cells of GT patients. In the present study, bone marrow from beta3-deficient (beta3-/-) mice was transduced with the ITGbeta3-cassette to investigate whether the platelet progeny could establish hemostasis in vivo. A lentivirus transfer vector equipped with the human ITGA2B gene promoter confined transgene expression to the platelet lineage. Human beta3 formed a stable complex with murine alphaIIb, effectively restoring platelet function. Mice expressing significant levels of alphaIIbbeta3 on circulating platelets exhibited improved bleeding times. Intravenous immunoglobulin effectively diminished platelet clearance in animals that developed an antibody response to alphaIIbbeta3. These results indicate the feasibility of targeting platelets with genetic therapies for better management of patients with inherited bleeding disorders.


Assuntos
Plaquetas/metabolismo , Modelos Animais de Doenças , Terapia Genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/uso terapêutico , Trombastenia/genética , Trombastenia/terapia , Animais , Humanos , Imunoglobulinas/sangue , Imunoglobulinas/imunologia , Camundongos , Especificidade de Órgãos , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Regiões Promotoras Genéticas/genética , Trombastenia/metabolismo , Trombastenia/patologia
19.
Exp Hematol ; 31(12): 1357-62, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14662345

RESUMO

OBJECTIVE: The purpose of this study was to determine if nonmyeloablative bone marrow transplantation would induce stable hematopoietic chimerism that would correct the bleeding diathesis associated with type I Glanzmann's thrombasthenia (GT). METHODS: Three young dogs (less than 12 weeks of age) with GT were transplanted with DLA-matched bone marrow from littermates. Recipients received a sublethal dose (200 cGy) of total-body irradiation (TBI) prior to infusion with bone marrow (1-4 x 10(8) cells/kg). Recipient dogs were immunosuppressed with cyclosporine (15 mg/kg) and mycophenolate mofetil (10 mg/kg). Chimerism was determined by quantitation of donor microsatellite repeat polymorphisms in peripheral blood DNA and by flow cytometry to detect the presence of glycoproteins IIb and IIIa on platelets. Platelet function was assessed by a clot retraction test. RESULTS: One dog died one week posttransplant due to hemorrhage. Another dog died four weeks posttransplant from an unrecognized congenital heart defect and complications due to canine distemper virus infection. At the time of death, microsatellite analysis indicated 35 to 50% chimerism. Flow cytometry showed 20% of circulating platelets positive for glycoproteins IIb and IIIa. The third dog is alive and doing well approximately two years posttransplant. Hematopoietic chimerism has been sustained at 35 to 60% with approximately 30% of the platelets positive for glycoproteins IIb and IIIa. Platelet function is normal based on clot retraction. The animal does not have clinical signs of bleeding. CONCLUSIONS: These observations suggest that GT and perhaps other severe inherited platelet disorders can be corrected using nonmyeloablative bone marrow transplantation to establish partial chimerism with normal platelets in the platelet compartment.


Assuntos
Transplante de Medula Óssea/métodos , Ácido Micofenólico/análogos & derivados , Trombastenia/terapia , Animais , Ciclosporina/administração & dosagem , Cães , Terapia de Imunossupressão/métodos , Repetições de Microssatélites , Modelos Animais , Ácido Micofenólico/administração & dosagem , Testes de Função Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/análise , Quimeras de Transplante , Condicionamento Pré-Transplante/métodos , Resultado do Tratamento , Irradiação Corporal Total
20.
Blood ; 102(12): 4006-13, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12881300

RESUMO

Activated platelets release their granule content in a concentrated fashion at sites of injury. We examined whether ectopically expressed factor VIII in developing megakaryocytes would be stored in alpha-granules and whether its release from circulating platelets would effectively ameliorate bleeding in a factor VIIInull mice model. Using the proximal glycoprotein 1b alpha promoter to drive expression of a human factor VIII cDNA construct, transgenic lines were established. One line had detectable human factor VIII that colocalizes with von Willebrand factor in platelets. These animals had platelet factor VIII levels equivalent to 3% to 9% plasma levels, although there was no concurrent plasma human factor VIII detectable. When crossed onto a factor VIIInull background, whole blood clotting time was partially corrected, equivalent to a 3% correction level. In a cuticular bleeding time study, these animals also had only a partial correction, but in an FeCl3 carotid artery, thrombosis assay correction was equivalent to a 50% to 100% level. These studies show that factor VIII can be expressed and stored in platelet alpha-granules. Our studies also suggest that platelet-released factor VIII is at least as potent as an equivalent plasma level and perhaps even more potent in an arterial thrombosis model.


Assuntos
Plaquetas/metabolismo , Fator VIII/administração & dosagem , Fator VIII/biossíntese , Terapia Genética/métodos , Hemofilia A/terapia , Animais , Testes de Coagulação Sanguínea , Plaquetas/ultraestrutura , Artérias Carótidas , Grânulos Citoplasmáticos/química , Modelos Animais de Doenças , Fator VIII/genética , Hemorragia/prevenção & controle , Hemorragia/terapia , Humanos , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Deleção de Sequência , Trombose/prevenção & controle , Trombose/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...