Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Thromb Haemost ; 122(9): 1469-1478, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35717947

RESUMEN

Phosphomannomutase 2 (PMM2) deficiency is the most prevalent congenital disorder of glycosylation. It is associated with coagulopathy, including protein C deficiency. Since all components of the anticoagulant and cytoprotective protein C system are glycosylated, we sought to investigate the impact of an N-glycosylation deficiency on this system as a whole. To this end, we developed a PMM2 knockdown model in the brain endothelial cell line hCMEC/D3. The resulting PMM2low cells were less able to generate activated protein C (APC), due to lower surface expression of thrombomodulin and endothelial protein C receptor. The low protein levels were due to downregulated transcription of the corresponding genes (THBD and PROCR, respectively), which itself was related to downregulation of transcription regulators Krüppel-like factors 2 and 4 and forkhead box C2. PMM2 knockdown was also associated with impaired integrity of the endothelial cell monolayer-partly due to an alteration in the structure of VE-cadherin in adherens junctions. The expression of protease-activated receptor 1 (involved in the cytoprotective effects of APC on the endothelium) was not affected by PMM2 knockdown. Thrombin stimulation induced hyperpermeability in PMM2low cells. However, pretreatment of cells with APC before thrombin simulation was still associated with a barrier-protecting effect. Taken as a whole, our results show that the partial loss of PMM2 in hCMEC/D3 cells is associated with impaired activation of protein C and a relative increase in barrier permeability.


Asunto(s)
Proteína C , Trombina , Trastornos Congénitos de Glicosilación , Endotelio , Glicosilación , Humanos , Fosfotransferasas (Fosfomutasas)/deficiencia
2.
J Thromb Haemost ; 20(7): 1653-1664, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35445541

RESUMEN

BACKGROUND: Protein S (PS) is a natural anticoagulant acting as a cofactor for activated protein C (APC) in the proteolytic inactivation of activated factors V (FVa) and VIII (FVIIIa), but also for tissue factor pathway inhibitor α (TFPIα) in the inhibition of activated factor X (FXa). OBJECTIVE: For therapeutic purposes, we aimed at generating single-domain antibodies (sdAbs) that could specifically modulate the APC-cofactor activity of PS in vivo. METHODS: A llama-derived immune library of sdAbs was generated and screened on recombinant human PS by phage display. PS binders were tested in a global activated partial thromboplastin time (APTT)-based APC-cofactor activity assay. RESULTS: A PS-specific sdAb (PS003) was found to enhance the APC-cofactor activity of PS in our APTT-based assay, and this enhancing effect was greater for a bivalent form of PS003 (PS003biv). Further characterization of PS003biv demonstrated that PS003biv also enhanced the APC-cofactor activity of PS in a tissue factor (TF)-induced thrombin generation assay and stimulated APC in the inactivation of FVa, but not FVIIIa, in plasma-based assays. Furthermore, PS003biv was directed against the sex hormone-binding globulin (SHBG)-like domain but did not inhibit the binding of PS to C4b-binding protein (C4BP) and did not interfere with the TFPIα-cofactor activity of PS. In mice, PS003biv exerted an antithrombotic effect in a FeCl3 -induced thrombosis model, while not affecting physiological hemostasis in a tail-clip bleeding model. DISCUSSION: Altogether, these results showed that pharmacological enhancement of the APC-cofactor activity of PS through an original anti-PS sdAb might constitute a promising and safe antithrombotic strategy.


Asunto(s)
Proteína S , Anticuerpos de Dominio Único , Animales , Factor VIIIa/química , Fibrinolíticos/farmacología , Humanos , Ratones , Proteína C/metabolismo , Proteína S/metabolismo
3.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445542

RESUMEN

Endoglin (Eng) is an endothelial cell (EC) transmembrane glycoprotein involved in adhesion and angiogenesis. Eng mutations result in vessel abnormalities as observed in hereditary hemorrhagic telangiectasia of type 1. The role of Eng was investigated in endothelial functions and permeability under inflammatory conditions, focusing on the actin dynamic signaling pathway. Endothelial Colony-Forming Cells (ECFC) from human cord blood and mouse lung/aortic EC (MLEC, MAEC) from Eng+/+ and Eng+/- mice were used. ECFC silenced for Eng with Eng-siRNA and ctr-siRNA were used to test tubulogenesis and permeability +/- TNFα and +/- LIM kinase inhibitors (LIMKi). In silico modeling of TNFα-Eng interactions was carried out from PDB IDs 5HZW and 5HZV. Calcium ions (Ca2+) flux was studied by Oregon Green 488 in epifluorescence microscopy. Levels of cofilin phosphorylation and tubulin post-translational modifications were evaluated by Western blot. F-actin and actin-tubulin distribution/co-localization were evaluated in cells by confocal microscopy. Eng silencing in ECFCs resulted in a decrease of cell sprouting by 50 ± 15% (p < 0.05) and an increase in pseudo-tube width (41 ± 4.5%; p < 0.001) compared to control. Upon TNFα stimulation, ECFC Eng-siRNA displayed a significant higher permeability compared to ctr-siRNA (p < 0.01), which is associated to a higher Ca2+ mobilization (p < 0.01). Computational analysis suggested that Eng mitigated TNFα activity. F-actin polymerization was significantly increased in ECFC Eng-siRNA, MAEC+/-, and MLEC+/- compared to controls (p < 0.001, p < 0.01, and p < 0.01, respectively) as well as actin/tubulin distribution (p < 0.01). Furthermore, the inactive form of cofilin (P-cofilin at Ser3) was significantly decreased by 36.7 ± 4.8% in ECFC Eng-siRNA compared to ctr-siRNA (p < 0.001). Interestingly, LIMKi reproduced the absence of Eng on TNFα-induced ECFC-increased permeability. Our data suggest that Eng plays a critical role in the homeostasis regulation of endothelial cells under inflammatory conditions (TNFα), and loss of Eng influences ECFC-related permeability through the LIMK/cofilin/actin rearrangement-signaling pathway.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Permeabilidad de la Membrana Celular , Endoglina/metabolismo , Células Endoteliales/patología , Inflamación/patología , Quinasas Lim/metabolismo , Neovascularización Patológica/patología , Factores Despolimerizantes de la Actina/genética , Animales , Endoglina/genética , Células Endoteliales/metabolismo , Inflamación/genética , Inflamación/metabolismo , Quinasas Lim/genética , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo
4.
TH Open ; 5(2): e220-e229, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34189397

RESUMEN

The protein Z (PZ)-dependent plasma protease inhibitor (ZPI) is a glycoprotein that inhibits factor XIa and, in the presence of PZ, FXa. Recently, ZPI has been shown to be an acute-phase protein (APP). As usually APPs downregulate the harmful effects of inflammation, we tested whether ZPI could modulate the increase of cytokines observed in inflammatory states. We observed that recombinant human ZPI (rhZPI) significantly decreases the levels of interleukin (IL)-1, IL-6, and tumor necrosis factor- α (TNF-α) induced by lipopolysaccharide (LPS) in a whole blood model. This inhibitory effect was unaffected by the presence of PZ or heparin. A ZPI mutant within the reactive loop center ZPI (Y387A), lacking anticoagulant activity, still had an anti-inflammatory activity. Surprisingly, rhZPI did not inhibit the synthesis of IL-6 or TNF-α when purified monocytes were stimulated by LPS, whereas the inhibitory effect was evidenced when lymphocytes were added to monocytes. The requirement of lymphocytes could be due to the synthesis of CCL5 (RANTES), a chemokine mainly produced by activated lymphocytes which is induced by rhZPI, and which can reduce the production of proinflammatory cytokines in whole blood. Lastly, we observed that the intraperitoneal injection of rhZPI significantly decreased LPS-induced IL-6 and TNF-α production in mouse plasma.

5.
Physiol Rep ; 9(3): e14613, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33512067

RESUMEN

The Transient Receptor Potential Vanilloid 4 (TRPV4) of endothelial cells contributes to many important functions including the regulation of Ca2+ homeostasis, cell volume, endothelial barrier permeability, and smooth muscle tone. However, its role in the transition of endothelial cells toward a pro-inflammatory phenotype has not been studied so far. Using both arterial and venous endothelial cells, we first show that the pharmacological activation of TRPV4 channels with GSK1016790A, a potent TRPV4 agonist, triggers robust and sustained Ca2+ increases, which are blocked by both TRPV4 antagonists HC067047 and RN9893. TRPV4 activation also triggers the actin cytoskeleton and adherens junction (VE-Cadherin) rearrangement in both arterial and venous endothelial cells and leads to rapid decreases of trans-endothelial electrical resistance. In addition to its effect on endothelial barrier integrity, TRPV4 activation selectively increases ICAM-1 surface expression in arterial and venous endothelial cells, due to the stimulation of ICAM-1 gene expression through the NF-κB transcription factor. TRPV4 channel activation also induced apoptosis of venous and arterial endothelial cells, while TRPV4 blockade reduced apoptosis, even in the absence of TRPV4 activation. As altered barrier integrity, increased adhesion molecule expression and apoptosis are hallmarks of the pro-inflammatory state of endothelial cells, our results indicate that TRPV4 channel activity can induce the transition of both venous and arterial endothelial cells toward a pro-inflammatory phenotype.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Leucina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Sulfonamidas/farmacología , Canales Catiónicos TRPV/agonistas , Antígenos CD/metabolismo , Apoptosis , Cadherinas/metabolismo , Señalización del Calcio , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucina/farmacología , FN-kappa B/metabolismo , Permeabilidad , Fenotipo , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Canales Catiónicos TRPV/metabolismo
6.
Eur J Med Chem ; 209: 112873, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33038796

RESUMEN

In this work, a series of cyclic bridged analogs of isocombretastatin A-4 (isoCA-4) with phenyl or pyridine linkers were designed and synthesized. The synthesis of the desired analogs was performed by the formation of nitro-vinyl intermediates, followed by a Cadogan cyclization. Structure activity relationship (SAR) study demonstrates the critical role of the combination of quinaldine as ring A, pyridine as the linker, and indole as ring B in the same molecule, for the cytotoxic activity. Among all tested compounds, compound 42 showed the highest antiproliferative activity against a panel of cancer cell lines with average IC50 values of 5.6 nM. Also, compound 42 showed high antiproliferative activity against the MDR1-overexpressing K562R cell line; thus, it was 1.5- and 12-fold more active than the reference compounds, isoCA-4 and CA-4, respectively. Moreover, 42 displayed a strong antiproliferative activity against the colon-carcinoma cells (HT-29), which are resistant to combretastatin A-4 and isoCA-4, and it was found to be 8000-fold more active than natural CA-4. Compound 42 also effectively inhibited tubulin polymerization both in vitro and in cells, and induced cell cycle arrest in G2/M phase. Next, we demonstrated that compound 42 dose-dependently caused caspase-induced apoptosis of K562 cells through mitochondrial dysfunction. Finally, we evaluated the effect of compound 42 in human no cancer cells compared to the reference compound. We demonstrated that 42 was 73 times less cytotoxic than isoCA-4 in quiescent peripheral blood lymphocytes (PBLs). In summary, these results suggest that compound 42 represents a promising tubulin inhibitor worthy of further investigation.


Asunto(s)
Diseño de Fármacos , Estilbenos/química , Estilbenos/farmacología , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclización , Humanos , Simulación del Acoplamiento Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Estilbenos/síntesis química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química
7.
Blood ; 135(22): 1969-1982, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32276277

RESUMEN

Anticoagulant protein S (PS) in platelets (PSplt) resembles plasma PS and is released on platelet activation, but its role in thrombosis has not been elucidated. Here we report that inactivation of PSplt expression using the Platelet factor 4 (Pf4)-Cre transgene (Pros1lox/loxPf4-Cre+) in mice promotes thrombus propensity in the vena cava, where shear rates are low, but not in the carotid artery, where shear rates are high. At a low shear rate, PSplt functions as a cofactor for both activated protein C and tissue factor pathway inhibitor, thereby limiting factor X activation and thrombin generation within the growing thrombus and ensuring that highly activated platelets and fibrin remain localized at the injury site. In the presence of high thrombin concentrations, clots from Pros1lox/loxPf4-Cre- mice contract, but not clots from Pros1lox/loxPf4-Cre+ mice, because of highly dense fibrin networks. Thus, PSplt controls platelet activation as well as coagulation in thrombi in large veins, but not in large arteries.


Asunto(s)
Plaquetas/metabolismo , Proteína S/metabolismo , Trombosis/sangre , Animales , Tiempo de Sangría , Coagulación Sanguínea/genética , Coagulación Sanguínea/fisiología , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Activación Plaquetaria/genética , Activación Plaquetaria/fisiología , Agregación Plaquetaria/genética , Agregación Plaquetaria/fisiología , Factor Plaquetario 4/genética , Factor Plaquetario 4/metabolismo , Proteína S/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombosis/etiología , Trombosis/genética , Trombosis de la Vena/sangre , Trombosis de la Vena/etiología , Trombosis de la Vena/genética
8.
Hematology ; 24(1): 742-750, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31736432

RESUMEN

Deep vein thrombosis is a common disease associated with a variety of complications including post-thrombotic syndrome as a late complication. It is now clear that in addition to classical deep vein thrombosis triggers such as blood flow disturbance, hypercoagulability, and vessel wall changes, inflammation has a key role in the pathophysiology of deep vein thrombosis, and there is a close relationship between inflammation and coagulation. As attested by changes in several plasma biomarkers, inflammation may have a significant role in the development of post-thrombotic syndrome. Here, we review the link between inflammation and deep vein thrombosis and thus the potential value of anti-inflammatory and/or anticoagulant drugs in the treatment of deep vein thrombosis and the prevention of post-thrombotic syndrome.


Asunto(s)
Inflamación/complicaciones , Trombosis de la Vena/terapia , Humanos , Trombosis de la Vena/patología
9.
J Med Chem ; 61(15): 6574-6591, 2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30004697

RESUMEN

Designing multitarget drugs have raised considerable interest due to their advantages in the treatment of complex diseases such as cancer. Their design constitutes a challenge in antitumor drug discovery. The present study reports a dual inhibition of tubulin polymerization and HDAC activity. On the basis of 1,1-diarylethylenes ( isoCA-4) and belinostat, a series of hybrid molecules was successfully designed and synthesized. In particular compounds, 5f and 5h were proven to be potent inhibitors of both tubulin polymerization and HDAC8 leading to excellent antiproliferative activity.


Asunto(s)
Diseño de Fármacos , Histona Desacetilasas/metabolismo , Estilbenos/química , Estilbenos/farmacología , Tubulina (Proteína)/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Células HCT116 , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/química , Humanos , Células K562 , Conformación Proteica , Estilbenos/síntesis química , Tubulina (Proteína)/química
10.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29630665

RESUMEN

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotelio Vascular/patología , Proteínas de la Membrana/metabolismo , Infecciones Meningocócicas/complicaciones , Microvasos/patología , Proteína C/metabolismo , Púrpura Fulminante/etiología , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Adhesión Bacteriana , Coagulación Sanguínea/fisiología , Células Cultivadas , Receptor de Proteína C Endotelial/genética , Endotelio Vascular/metabolismo , Endotelio Vascular/microbiología , Humanos , Proteínas de la Membrana/genética , Infecciones Meningocócicas/microbiología , Microvasos/metabolismo , Microvasos/microbiología , Neisseria meningitidis/fisiología , Proteína C/genética , Púrpura Fulminante/metabolismo , Púrpura Fulminante/patología
11.
Blood ; 131(12): 1360-1371, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29317453

RESUMEN

Improved treatments are needed for hemophilia A and B, bleeding disorders affecting 400 000 people worldwide. We investigated whether targeting protein S could promote hemostasis in hemophilia by rebalancing coagulation. Protein S (PS) is an anticoagulant acting as cofactor for activated protein C and tissue factor pathway inhibitor (TFPI). This dual role makes PS a key regulator of thrombin generation. Here, we report that targeting PS rebalances coagulation in hemophilia. PS gene targeting in hemophilic mice protected them against bleeding, especially when intra-articular. Mechanistically, these mice displayed increased thrombin generation, resistance to activated protein C and TFPI, and improved fibrin network. Blocking PS in plasma of hemophilia patients normalized in vitro thrombin generation. Both PS and TFPIα were detected in hemophilic mice joints. PS and TFPI expression was stronger in the joints of hemophilia A patients than in those of hemophilia B patients when receiving on-demand therapy, for example, during a bleeding episode. In contrast, PS and TFPI expression was decreased in hemophilia A patients receiving prophylaxis with coagulation factor concentrates, comparable to osteoarthritis patients. These results establish PS inhibition as both controller of coagulation and potential therapeutic target in hemophilia. The murine PS silencing RNA approach that we successfully used in hemophilic mice might constitute a new therapeutic concept for hemophilic patients.


Asunto(s)
Coagulación Sanguínea , Proteínas Portadoras , Hemofilia A , Hemorragia , Animales , Proteínas de Unión al Calcio , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Fibrina/genética , Fibrina/metabolismo , Silenciador del Gen , Hemofilia A/sangre , Hemofilia A/genética , Hemofilia A/terapia , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patología , Hemorragia/prevención & control , Humanos , Ratones , Ratones Noqueados , Trombina/genética , Trombina/metabolismo
12.
Eur J Med Chem ; 143: 473-490, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29202409

RESUMEN

A novel series of tubulin polymerization inhibitors, based on fluorinated derivatives of isocombretastatin A-4 was synthesized with the goal of evaluating the effect of these compounds on the proliferative activity. The introduction of fluorine atom was performed on the phenyl ring or at the linker between the two aromatic rings. The modification of isoCA-4 by introduction of difluoromethoxy group at the para-position (3i) and substitution of the two protons of the linker by two fluorine atoms (3m), produced the most active compounds in the series, with IC50 values of 0.15-2.2 nM (3i) and 0.1-2 nM (3m) respectively, against a panel of six cancer cell lines. Compounds 3i and 3m had greater antiproliferative activity in comparison with references CA-4 or isoCA-4, the presence of fluorine group leads to a significant enhancement of the antiproliferative activity. Molecular docking studies indicated that compounds 3i and 3m occupy the colchicine binding site of tubulin. Evaluation of cytotoxicity in Human noncancer cells indicated that the compounds 3i and 3m were practically ineffective in quiescent peripheral blood lymphocytes, and may have a selective antiproliferative activity against cancer cells. Analyses of cell cycle distribution, and morphological microtubules organization showed that compound 3m induced G2/M phase arrest and, dramatically disrupted the microtubule network.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Flúor/química , Estilbenos/farmacología , Tubulina (Proteína)/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Estructura Molecular , Polimerizacion/efectos de los fármacos , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad
14.
Anal Chim Acta ; 947: 58-65, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27846990

RESUMEN

Antithrombin (AT) is a plasma glycoprotein which possesses anticoagulant and anti-inflammatory properties. AT exhibits various forms, among which are native, latent and heterodimeric ones. We studied the potential of capillary electrophoresis-mass spectrometry (CE-MS) using a sheath liquid interface, electrospray ionization (ESI), and a quadrupole-time-of-flight (Q-TOF) mass spectrometer to separate and quantify the different AT forms. For CE separation, a neutral polyvinyl alcohol (PVA) coated capillary was employed. The protein conformation was preserved by using a background electrolyte (BGE) at physiological pH. A sheath liquid of isopropanol-water 50:50 (v/v) with 14 mM ammonium acetate delivered at a flow rate of 120 µL h-1 resulted in optimal signal intensities. Each AT form exhibited a specific mass spectrum, allowing unambiguous distinction. Several co-injection experiments proved that latent AT had a higher electrophoretic mobility (µep) than native AT, and that these conformers could associate to form a heterodimer during the CE analysis. The developed CE-MS method enabled the detection and quantitation of latent and heterodimeric forms in a commercial AT preparation stored at room temperature for three weeks.


Asunto(s)
Proteínas Antitrombina/química , Electroforesis Capilar/métodos , Espectrometría de Masas/métodos , Multimerización de Proteína , Proteínas Antitrombina/aislamiento & purificación , Modelos Moleculares , Estructura Cuaternaria de Proteína , Temperatura
15.
Thromb Haemost ; 116(3): 452-60, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27412396

RESUMEN

In the absence of specific antidote to fondaparinux, two modified forms of antithrombin (AT), one recombinant inactive (ri-AT) and the other chemically inactivated (chi-AT), were designed to antagonise AT-mediated anticoagulants, e. g. heparins or fondaparinux. These inactive ATs were previously proven to effectively neutralise anticoagulant activity associated with heparin derivatives in vitro and in vivo, as assessed by direct measurement of anti-FXa activity. This study was undertaken to evaluate in vitro the effectivity of inactive ATs to reverse anticoagulation by heparin derivatives and to compare them with non-specific fondaparinux reversal agents, like recombinant-activated factor VII (rFVIIa) or activated prothrombin-complex concentrate (aPCC), in a thrombin-generation assay (TGA). Addition of fondaparinux (3 µg/ml) to normal plasma inhibited thrombin generation by prolonging lag time (LT) as much as 244 % and lowering endogenous thrombin potential (ETP) to 17 % of their control (normal plasma) values. Fondaparinux-anticoagulant activity was reversed by ri-AT and chi-AT, as reflected by the corrections of LT up to 117 % and 114 % of its control value, and ETP recovery to 78 % and 63 %, respectively. Unlike ri-AT that had no effect on thrombin generation in normal plasma, chi-AT retained anticoagulant activity that minimises its reversal capacity. However, both ATs were more effective than rFVIIa or aPCC at neutralising fondaparinux and, unlike non-specific antidotes, inactive ATs specifically reversed AT-mediated anticoagulant activities, as suggested by their absence of procoagulant activity in anticoagulant-free plasma.


Asunto(s)
Antídotos/metabolismo , Antitrombinas/metabolismo , Polisacáridos/antagonistas & inhibidores , Trombina/biosíntesis , Anticoagulantes/administración & dosificación , Antídotos/análisis , Antitrombinas/análisis , Análisis Químico de la Sangre/métodos , Relación Dosis-Respuesta a Droga , Factor VIIa/análisis , Factor VIIa/metabolismo , Inhibidores del Factor Xa/análisis , Inhibidores del Factor Xa/metabolismo , Fondaparinux , Hemostáticos/análisis , Hemostáticos/metabolismo , Heparina/administración & dosificación , Heparina de Bajo-Peso-Molecular/antagonistas & inhibidores , Humanos , Técnicas In Vitro , Trombina/análisis
16.
Adv Exp Med Biol ; 898: 333-52, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161235

RESUMEN

Calcium ions (Ca(2+)) are versatile messengers that need to be tidily regulated in time and space in order to create a large number of signals. The coupling between Ca(2+) entry and Ca(2+) refilling is playing a central role in this Ca(2+) homeostasis. Since the capacitative Ca(2+) entry has been described, different mechanisms have been proposed in order to explain how the Ca(2+) entry could be under control of intracellular store Ca(2+) depletion. Today, in addition of STIM1 and Orai1, the two major elements of SOCe, increasing attention is put on the role of the transient receptor potential canonical (TRPC), that can form protein clusters with Orai1, and Sarco/endoplasmic reticulum Ca(2+)ATPases (SERCAs), that refill the stores and are also located in the same environment than SOC clusters. Altogether, these proteins elaborate either Ca(2+) microdomains in the vicinity of the membrane or larger Ca(2+) increases overtaking the whole cell. The coupling between Ca(2+) entry and Ca(2+) refilling can possibly act much further away from the plasma membrane. Ca(2+), uptaken by SERCAs, have been described to move faster and further in the ER than in the cytosol and to create specific signal that depends on Ca(2+) entry but at longer distance from it. The complexity of such created Ca(2+) currents resides in the heteromeric nature of channels as well as the presence of different intracellular stores controlled by SERCA2b and SERCA3, respectively. A role for mitochondria has also been explored. To date, mitochondria are other crucial compartments that play an important role in Ca(2+) homeostasis. Although mitochondria mostly interact with intracellular stores, coupling of Ca(2+) entry and mitochondria cannot be completely rule out.


Asunto(s)
Calcio/metabolismo , Animales , Humanos , Transporte Iónico , Mitocondrias/enzimología , Mitocondrias/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Canales Catiónicos TRPC/metabolismo
17.
Electrophoresis ; 37(12): 1696-703, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26989842

RESUMEN

Antithrombin (AT) is a human plasma glycoprotein that possesses anticoagulant and anti-inflammatory properties. However, the native (active) form of AT is unstable and undergoes conformational changes, leading to latent, cleaved, and heterodimeric forms. The presence of these alternative forms mostly inactive can highly impact the quality and therapeutic activity of pharmaceutical AT preparations. We developed a capillary zone electrophoresis method, based on a neutral polyethylene oxide-coated capillary and a buffer close to physiological conditions, enabling the separation of more than eight forms of AT. Several peaks were identified as native, latent, and heterodimeric forms. The CZE method was reproducible with intraday relative standard deviations less than 0.5 and 2% for migration times and peak areas, respectively. The method was applied to the comparison of AT preparations produced by five competitive pharmaceutical companies, and statistical tests were performed. Important differences in the proportion of each form were highlighted. In particular, one AT preparation was shown to contain a high quantity of heterodimer, and two preparations contained high quantities of latent form. In addition, one AT preparation exhibited additional forms, not yet identified.


Asunto(s)
Antitrombina III/aislamiento & purificación , Electroforesis Capilar/métodos , Tampones (Química) , Dimerización , Composición de Medicamentos , Electroforesis Capilar/instrumentación , Polietilenglicoles , Conformación Proteica , Isoformas de Proteínas/aislamiento & purificación
18.
J Pharm Biomed Anal ; 111: 64-70, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25863018

RESUMEN

With the aim to determine the binding affinity of a new generation of recombinant antithrombin (AT) toward heparin, we developed a dynamic equilibrium-affinity capillary electrophoresis (DE-ACE) method. This method allows the determination of an AT-heparin binding constant (Kd) directly from the cell culture supernatant used to produce the AT variants. Eight measurements per AT variant are sufficient to determine an accurate Kd (uncertainty ≤ 22%, regression coefficient ≥ 0.97), which is not significantly different from the value obtained from a higher number of measurements. Due to the relatively short time required to determine the Kd of one AT variant (2h), this method has the potential for being a low throughput screening method. The method was validated by analyzing five AT variants, whose Kd have been reported in the literature using fluorescence spectroscopy. Finally, the method was applied to estimate the Kd of one new AT variant and one AT conformer, a latent form, that exhibits a significant loss of affinity.


Asunto(s)
Antitrombinas/química , Heparina/química , Técnicas de Cultivo de Célula/métodos , Electroforesis Capilar/métodos , Humanos , Cinética , Espectrometría de Fluorescencia/métodos
19.
Circulation ; 124(8): 930-9, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21810657

RESUMEN

BACKGROUND: Formation of platelet plug initiates hemostasis after vascular injury and triggers thrombosis in ischemic disease. However, the mechanisms leading to the formation of a stable thrombus are poorly understood. Connexins comprise a family of proteins that form gap junctions enabling intercellular coordination of tissue activity, a process termed gap junctional intercellular communication. METHODS AND RESULTS: In the present study, we show that megakaryocytes and platelets express connexin 37 (Cx37). Deletion of the Cx37 gene in mice shortens bleeding time and increases thrombus propensity. Aggregation is increased in murine Cx37(-/-) platelets or in murine Cx37(+/+) and human platelets treated with gap junction blockers. Intracellular microinjection of neurobiotin, a Cx37-permeant tracer, revealed gap junctional intercellular communication in platelet aggregates, which was impaired in Cx37(-/-) platelets and in human platelets exposed to gap junction blockers. Finally, healthy subjects homozygous for Cx37-1019C, a prognostic marker for atherosclerosis, display increased platelet responses compared with subjects carrying the Cx37-1019T allele. Expression of these polymorphic channels in communication-deficient cells revealed a decreased permeability of Cx37-1019C channels for neurobiotin. CONCLUSIONS: We propose that the establishment of gap junctional communication between Cx37-expressing platelets provides a mechanism to limit thrombus propensity. To our knowledge, these data provide the first evidence incriminating gap junctions in the pathogenesis of thrombosis.


Asunto(s)
Plaquetas/fisiología , Conexinas/fisiología , Megacariocitos/fisiología , Trombosis/genética , Trombosis/fisiopatología , Adolescente , Adulto , Animales , Biotina/análogos & derivados , Biotina/farmacocinética , Tiempo de Sangría , Conexinas/genética , Regulación hacia Abajo/fisiología , Uniones Comunicantes/fisiología , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Agregación Plaquetaria/fisiología , Polimorfismo Genético/fisiología , Adulto Joven , Proteína alfa-4 de Unión Comunicante
20.
Blood ; 115(16): 3390-7, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20139094

RESUMEN

Growth arrest-specific gene 6 (Gas6) is expressed in antigen-presenting cells and endothelial cells (ECs) but not in T cells. When wild-type (WT) or Gas6(-/-) mice received allogeneic non-T cell-depleted bone marrow cells, hepatic graft-versus-host disease (GVHD) was alleviated in Gas6(-/-) recipients regardless of donor genotype, but not in WT recipients. T-cell infiltration was more prominent and diffuse in WT than in Gas6(-/-) recipients' liver. When mice received 0.5 x 10(6) allogeneic T cells with T cell-depleted allogeneic bone marrow, clinical signs indicated that GVHD was less severe in Gas6(-/-) than in WT recipients, as shown by a significant improvement of the survival and reduced liver GVHD. These data demonstrate that donor cells were not involved in the protection mechanism. In addition, lack of Gas6 in antigen-presenting cells did not affect WT or Gas6(-/-) T-cell proliferation. We therefore assessed the response of WT or Gas6(-/-) ECs to tumor necrosis factor-alpha. Lymphocyte transmigration was less extensive through Gas6(-/-) than WT ECs and was not accompanied by increases in adhesion molecule levels. Thus, the lack of Gas6 in ECs impaired donor T-cell transmigration into the liver, providing a rationale for considering Gas6 pathway as a potential nonimmunosuppressive target to minimize GVHD in patients receiving allogeneic hematopoietic stem cell transplantation.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Hígado/inmunología , Animales , Separación Celular , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Células Endoteliales/metabolismo , Citometría de Flujo , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Hígado/patología , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA