Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Clin Lab ; 68(8)2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35975485

RESUMEN

BACKGROUND: Protein S is a central regulator of coagulation as it critically participates in down-regulation of both extrinsic and intrinsic pathways of the coagulation cascade. In this review, we aim to provide an update on protein S and its anticoagulant functions as a central hemostatic regulator. METHODS: Electronic databases including, Google, Google Scholar, PMC, PubMed, Science Direct, and Scopus were rigorously searched using the terms protein S, hemostasis, natural anticoagulants, regulators of coagulation, and coagulation inhibitors for the completion of this descriptive review. RESULTS: Literature review shows that protein S is a potent cofactor for activated protein C (APC) in the regulation of the intrinsic pathway and a cofactor for tissue factor pathway inhibitor (TFPI) in the regulation of the extrinsic pathway. The strong association between protein S deficiency either hereditary or acquired and increased risk for venous thrombosis indicates the important and central role of protein S in controlling the initiation and propagation phase of coagulation cascade and that protein S is an important determinant for optimal activity of both APC and TFPI in coagulation regulation. CONCLUSIONS: Available evidence suggests that the role of protein S in the down-regulation of blood coagulation is mainly mediated through its high affinity binding to negatively charged phospholipid surfaces. This high affinity binding to negatively charged phospholipids helps bring the anticoagulant proteins to the membranes, resulting in efficient and targeted regulation of coagulation. In the shade of current COVID-19 pandemic, protein S deficiency has been found to be a leading cause of thrombotic complications associated with COVID-19.


Asunto(s)
Coagulación Sanguínea , Deficiencia de Proteína S , Proteína S , Anticoagulantes/farmacología , COVID-19 , Humanos , Proteína S/fisiología
2.
Adv Exp Med Biol ; 1074: 577-583, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29721990

RESUMEN

MerTK is required for photoreceptor outer segment (POS) phagocytosis by retinal pigment epithelial (RPE) cells, a diurnal function essential for vision maintenance. In vivo, MerTK is stimulated at the time of the phagocytic peak through an intracellular signaling pathway. However, MerTK ligands Gas6 and Protein S are expressed in both RPE cells and photoreceptors, and at least one of them required for phagocytosis to occur. Still, their exact role in the retina was not clear until recently. This review combines results from different studies to shed the light on a tissue-specific regulation of MerTK function by its ligands. Indeed, with opposite effects on RPE phagocytosis and changes in their expression levels around the time of POS uptake, Gas6 and Protein S may contribute to the tight control of the acute phagocytic peak in the retina.


Asunto(s)
Apoptosis/fisiología , Proteínas del Ojo/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Fagocitosis/fisiología , Proteína S/fisiología , Retina/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Animales , Células Cultivadas , Ritmo Circadiano , Activación Enzimática , Humanos , Ligandos , Macrófagos/metabolismo , Ratones , Ratas , Retina/citología , Segmento Externo de la Célula en Bastón/metabolismo , Transducción de Señal/fisiología , Tirosina Quinasa c-Mer/deficiencia , Tirosina Quinasa c-Mer/fisiología
3.
Blood ; 123(16): 2460-9, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24596417

RESUMEN

TAM receptors (Tyro3, Axl, and Mer) belong to a family of receptor tyrosine kinases that have important effects on hemostasis and inflammation. Also, they affect cell proliferation, survival, adhesion, and migration. TAM receptors can be activated by the vitamin K-dependent proteins Gas6 and protein S. Protein S is more commonly known as an important cofactor for protein C as well as a direct inhibitor of multiple coagulation factors. To our knowledge, the functions of Gas6 are limited to TAM receptor activation. When activated, the TAM receptors have effects on primary hemostasis and coagulation and display an anti-inflammatory or a proinflammatory effect, depending on cell type. To comprehend the effects that the TAM receptors and their ligands have on hemostasis and inflammation, we compare studies that report the different phenotypes displayed by mice with deficiencies in the genes of this receptor family and its ligands (protein S(+/-), Gas6(-/-), TAM(-/-), and variations of these). In this manner, we aim to display which features are attributable to the different ligands. Because of the effects TAM receptors have on hemostasis, inflammation, and cancer growth, their modulation could make interesting therapeutic targets in thromboembolic disease, atherosclerosis, sepsis, autoimmune disease, and cancer.


Asunto(s)
Hemostasis/genética , Inflamación/genética , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteína S/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Humanos , Inflamación/metabolismo , Ratones , Proteínas Proto-Oncogénicas/fisiología , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
4.
Blood ; 117(24): 6685-93, 2011 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-21508412

RESUMEN

Protein S has an important anticoagulant function by acting as a cofactor for activated protein C (APC). We recently reported that the EGF1 domain residue Asp95 is critical for APC cofactor function. In the present study, we examined whether additional interaction sites within the Gla domain of protein S might contribute to its APC cofactor function. We examined 4 residues, composing the previously reported "Face1" (N33S/P35T/E36A/Y39V) variant, as single point substitutions. Of these protein S variants, protein S E36A was found to be almost completely inactive using calibrated automated thrombography. In factor Va inactivation assays, protein S E36A had 89% reduced cofactor activity compared with wild-type protein S and was almost completely inactive in factor VIIIa inactivation; phospholipid binding was, however, normal. Glu36 lies outside the ω-loop that mediates Ca(2+)-dependent phospholipid binding. Using mass spectrometry, it was nevertheless confirmed that Glu36 is γ-carboxylated. Our finding that Gla36 is important for APC cofactor function, but not for phospholipid binding, defines a novel function (other than Ca(2+) coordination/phospholipid binding) for a Gla residue in vitamin K-dependent proteins. It also suggests that residues within the Gla and EGF1 domains of protein S act cooperatively for its APC cofactor function.


Asunto(s)
Ácido 1-Carboxiglutámico/fisiología , Proteína C/metabolismo , Proteína S/metabolismo , Proteína S/fisiología , Ácido 1-Carboxiglutámico/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos/fisiología , Sitios de Unión/genética , Dominio Catalítico/genética , Células Cultivadas , Factor VIIIa/metabolismo , Factor Va/metabolismo , Humanos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Proteína C/agonistas , Proteína C/fisiología , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteína S/química , Proteína S/genética , Estructura Terciaria de Proteína , Relación Estructura-Actividad
5.
Arterioscler Thromb Vasc Biol ; 32(10): 2387-93, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22904276

RESUMEN

OBJECTIVE: Protein S is a vitamin K-dependent plasma protein that functions in the feedback regulation of thrombin generation. Our goal was to determine how protein S regulates the intrinsic pathway of blood coagulation. METHODS AND RESULTS: We used plasma, including platelet-rich plasma, and in vitro methods to determine how the intrinsic pathway of blood coagulation is regulated by protein S. We obtained the following results: (1) activated partial thromboplastin time assays with protein S-supplemented plasma confirmed that protein S prolongs clotting time; (2) a modified activated partial thromboplastin time assay with factor IX (fIX)-deficient plasma confirmed that protein S affects fIX-initiated clotting; (3) a fIXa/factor VIIIa (fVIIIa)-mediated thrombin generation assay with either platelet-rich plasma or factor-deficient plasma, initiated with a limiting amount of tissue factor, was regulated by protein S; (4) in the presence of phosphatidylserine vesicles, protein S inhibited fIXa in the absence and presence of fVIIIa; and (5) protein S altered only the K(M) for factor X activation by fIXa in the absence of fVIIIa and both k(cat) and K(M) in the presence of fVIIIa. CONCLUSIONS: From our findings, it can be concluded that protein S inhibits fIXa in the presence or absence of fVIIIa in an activated protein C-independent way.


Asunto(s)
Coagulación Sanguínea/fisiología , Factor IXa/antagonistas & inhibidores , Factor VIIIa/antagonistas & inhibidores , Proteína C/fisiología , Proteína S/fisiología , Factor IXa/fisiología , Factor VIIIa/fisiología , Retroalimentación Fisiológica/fisiología , Humanos , Técnicas In Vitro , Tiempo de Tromboplastina Parcial , Transducción de Señal/fisiología , Trombina/fisiología
6.
Cell Mol Life Sci ; 69(9): 1391-414, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22076650

RESUMEN

Ample clinical and preclinical evidence indicates that macrophages interact with tumor cells as well as with virtually all populations of host cells present in the tumor microenvironment. This crosstalk can strongly promote malignancy, but also has in principle the potential to inhibit tumor growth. Thus, it is of the utmost importance to improve our understanding of the mechanisms driving the pro- and antimalignant behavior of tumor-associated macrophages (TAMs) in order to develop better anticancer therapies. In this review, we discuss the biological consequences of reciprocal interactions between TAMs, cancer cells, endothelial cells, fibroblasts and other leukocyte subfractions within tumors. It was recently elucidated that tumors specifically educate macrophages to secrete growth arrest-specific gene 6 (Gas6), the common ligand of the Tyro3, Axl, Mer receptor (TAMR) family. In turn, Gas6 fosters tumor growth by promoting cancer cell proliferation. Therefore, the Gas6-TAMR axis might represent a novel target for disrupting tumor-macrophage crosstalk. We summarize here what is known about TAMR and their ligands in (human) cancer biology. In order to shed more light on the role of macrophages in human cancer, we additionally provide an overview of what is currently known about the prognostic impact of TAMs in human cancer.


Asunto(s)
Macrófagos/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Microambiente Tumoral/fisiología , Células Endoteliales/patología , Células Endoteliales/fisiología , Femenino , Fibroblastos/patología , Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Leucocitos/patología , Leucocitos/fisiología , Ligandos , Macrófagos/clasificación , Macrófagos/inmunología , Macrófagos/patología , Masculino , Modelos Biológicos , Neoplasias/patología , Neoplasias/fisiopatología , Pronóstico , Proteína S/fisiología
7.
Semin Thromb Hemost ; 38(2): 129-34, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22422327

RESUMEN

Blood coagulation in vivo is triggered by the tissue factor (TF) pathway. The major physiological regulator of this pathway is tissue factor pathway inhibitor (TFPI), a Kunitz-type inhibitor that regulates the activity of the TF-factor VIIa complex in a factor Xa-dependent manner, thus controlling the generation of thrombin and ultimately, fibrin. Although some of the in vivo and in vitro effects of TFPI have been described for nearly a century, the bulk of the research that has elucidated the physiology of this inhibitor has only occurred in the past 25 years. Despite this, many questions remain. This review will highlight the recent advances in knowledge related to TFPI, with an emphasis on new insights into its physiology, association with disease, and possible use as a therapeutic anticoagulant.


Asunto(s)
Lipoproteínas/fisiología , Empalme Alternativo , Animales , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Factor VIIa/metabolismo , Humanos , Lipoproteínas/química , Lipoproteínas/genética , Lipoproteínas/uso terapéutico , Proteína S/fisiología , Inhibidores de Serina Proteinasa/fisiología , Tromboplastina
8.
J Neurosci ; 30(46): 15521-34, 2010 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21084607

RESUMEN

The anticoagulant factor protein S (PS) protects neurons from hypoxic/ischemic injury. However, molecular mechanisms mediating PS protection in injured neurons remain unknown. Here, we show mouse recombinant PS protects dose-dependently mouse cortical neurons from excitotoxic NMDA-mediated neuritic bead formation and apoptosis by activating the phosphatidylinositol 3-kinase (PI3K)-Akt pathway (EC(50) = 26 ± 4 nm). PS stimulated phosphorylation of Bad and Mdm2, two downstream targets of Akt, which in neurons subjected to pathological overstimulation of NMDA receptors (NMDARs) increased the antiapoptotic Bcl-2 and Bcl-X(L) levels and reduced the proapoptotic p53 and Bax levels. Adenoviral transduction with a kinase-deficient Akt mutant (Ad.Akt(K179A)) resulted in loss of PS-mediated neuronal protection, Akt activation, and Bad and Mdm2 phosphorylation. Using the TAM receptors tyrosine kinases Tyro3-, Axl-, and Mer-deficient neurons, we showed that PS protected neurons lacking Axl and Mer, but not Tyro3, suggesting a requirement of Tyro3 for PS-mediated protection. Consistent with these results, PS dose-dependently phosphorylated Tyro3 on neurons (EC(50) = 25 ± 3 nm). In an in vivo model of NMDA-induced excitotoxic lesions in the striatum, PS dose-dependently reduced the lesion volume in control mice (EC(50) = 22 ± 2 nm) and protected Axl(-/-) and Mer(-/-) transgenic mice, but not Tyro3(-/-) transgenic mice. Using different structural PS analogs, we demonstrated that the C terminus sex hormone-binding globulin-like (SHBG) domain of PS is critical for neuronal protection in vitro and in vivo. Thus, our data show that PS protects neurons by activating the Tyro3-PI3K-Akt pathway via its SHGB domain, suggesting potentially a novel neuroprotective approach for acute brain injury and chronic neurodegenerative disorders associated with excessive activation of NMDARs.


Asunto(s)
Neuronas/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína S/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Globulina de Unión a Hormona Sexual/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/toxicidad , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
Semin Thromb Hemost ; 37(4): 355-61, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21805441

RESUMEN

Blood coagulation and complement pathways are two important natural defense systems. The high affinity interaction between the anticoagulant vitamin K-dependent protein S and the complement regulator C4b-binding protein (C4BP) is a direct physical link between the two systems. In human plasma, ~70% of total protein S circulates in complex with C4BP; the remaining is free. The anticoagulant activity of protein S is mainly expressed by the free form, although the protein S-C4BP complex has recently been shown to have some anticoagulant activity. The high affinity binding of protein S to C4BP provides C4BP with the ability to bind to negatively charged phospholipid membranes, which serves the purpose of localizing complement regulatory activity close to the membrane. Even though C4BP does not directly affect the coagulation system, it still influences the regulation of blood coagulation through its interaction with protein S. This is particularly important in states of inherited deficiency of protein S where the tight binding of protein S to C4BP results in a pronounced and selective drop in concentration of free protein S, whereas the concentration of protein S in complex with C4BP remains relatively unchanged. This review summarizes the current knowledge on C4BP with respect to its association with thrombosis and hemostasis.


Asunto(s)
Proteína de Unión al Complemento C4b/metabolismo , Hemostasis/fisiología , Trombosis/metabolismo , Animales , Humanos , Proteína S/metabolismo , Proteína S/fisiología
10.
Crit Care Med ; 38(2 Suppl): S18-25, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20083909

RESUMEN

OBJECTIVE: To review new findings about the function of the protein C system during inflammation and coagulation. MAIN FINDINGS: Coagulation proteases and their cofactors modify the outcome of severe inflammation by engaging signaling-competent cell surface receptors. The central effector protease of the protein C pathway, activated protein C, interacts with the endothelial cell protein C receptor, protease-activated receptors, and other receptors to exert multiple effects on hemostasis and immune cell function. Thrombomodulin controls the complement arm of the innate immune system in a thrombin-dependent manner through activation of the thrombin activatable inhibitor of fibrinolysis, and in a thrombin-independent, constitutive manner via its lectin-like extracellular domain; and inhibits the inflammatory effects of high-mobility box group 1 protein. Protein S not only suppresses coagulation as an enhancing cofactor for the coagulation inhibitors activated protein C and tissue factor pathway inhibitor but also is also a physiologic ligand for the Tyro/axl/Mer-family of receptor tyrosine kinases that mediate an anti-inflammatory regulatory loop of dendritic cell and monocyte inflammatory function. CONCLUSIONS: The immune-regulatory capacity of the protein C pathway and its individual components emerge as the dominant action of this pathway in the setting of severe inflammation.


Asunto(s)
Inflamación/fisiopatología , Proteína C/fisiología , Antígenos CD/fisiología , Coagulación Sanguínea/fisiología , Proteínas del Sistema Complemento/fisiología , Enfermedad Crítica , Receptor de Proteína C Endotelial , Hemostasis/fisiología , Humanos , Inmunidad Celular/fisiología , Inmunidad Innata/fisiología , Proteína S/fisiología , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología , Trombomodulina/fisiología
11.
FASEB J ; 23(7): 2244-53, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19244162

RESUMEN

Protein S (PS) is a cofactor for activated protein C (APC), which inactivates coagulation factors (F) Va and VIIIa. Deficiency of protein C or PS is associated with risk of thrombosis. We found that PS also has APC-independent anticoagulant activity (PS-direct) and directly inhibits thrombin generated by FXa/FVa (prothrombinase complex). Here we report that PS contains Zn(2+) that is required for PS-direct and that is lost during certain purification procedures. Immunoaffinity-purified PS contained 1.4 +/- 0.6 Zn(2+)/mol, whereas MonoQ-purified and commercial PS contained 0.15 +/- 0.15 Zn(2+)/mol. This may explain the controversy regarding the validity of PS-direct. Zn(2+) content correlated positively with PS-direct in prothrombinase assays and clotting assays, but APC-cofactor activity of PS was independent of Zn(2+) content. PS-direct and Zn(2+) were restored to inactive PS under mildly denaturing conditions. Conversely, o-phenanthroline reversibly impaired the PS-direct of active PS. Zn(2+)-containing PS bound FXa more efficiently (K(d)(app)=9.3 nM) than Zn(2+)-deficient PS (K(d)(app)=110 nM). PS bound TFPI efficiently, independently of Zn(2+) content (K(d)(app)=21 nM). Antibodies that block PS-direct preferentially recognized Zn(2+)-containing PS, suggesting conformation differences at or near the interface of 2 laminin G-like domains near the PS C terminus. Thus, Zn(2+) is required for PS-direct and efficient FXa binding and may play a role in stabilizing PS conformation.


Asunto(s)
Factor Xa/metabolismo , Lipoproteínas/metabolismo , Proteína C/metabolismo , Proteína S/química , Zinc/fisiología , Anticoagulantes , Coagulación Sanguínea , Pruebas de Coagulación Sanguínea , Unión Proteica , Conformación Proteica , Proteína S/aislamiento & purificación , Proteína S/fisiología , Zinc/química
12.
Arterioscler Thromb Vasc Biol ; 29(12): 2015-20, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19661488

RESUMEN

In the last decades evidence was obtained that protein S not only acts as cofactor of activated protein C (APC) in the downregulation of coagulation, but also expresses anticoagulant activity in the absence of APC. The search for the mechanism(s) underlying the APC-independent anticoagulant activity of protein S was hampered by the fact that protein S exhibited 2 seemingly identical anticoagulant activities in model systems and in plasma. Later it was shown that the anticoagulant activity of purified protein S in model systems was dependent on the concentration of phospholipid vesicles and was explained by low amounts of protein S multimers generated during purification that effectively inhibited phospholipid-dependent coagulation reactions via competition for phospholipid binding sites. Plasma does not contain multimers, and the anticoagulant activity of protein S in plasma was not affected by the phospholipid concentration but was dependent on the amount of tissue factor (TF) used for initiation of thrombin generation. This led to the discovery that protein S acts as cofactor of tissue factor pathway inhibitor (TFPI) which stimulates the inhibition of factor Xa by TFPI approximately 10-fold. The current review describes the background of the TFPI-cofactor activity of protein S as well as the rationale for the observation that the TFPI/protein S system particularly inhibits the TF pathway at low procoagulant stimuli.


Asunto(s)
Lipoproteínas/fisiología , Proteína S/fisiología , Anticoagulantes/metabolismo , Coagulación Sanguínea/fisiología , Factor VIIa/antagonistas & inhibidores , Inhibidores del Factor Xa , Humanos , Cinética , Modelos Biológicos , Proteína S/química , Estructura Cuaternaria de Proteína
14.
Curr Opin Hematol ; 15(5): 529-36, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18695379

RESUMEN

PURPOSE OF REVIEW: Protein S has been one of the least mechanistically understood amongst the vitamin K-dependent coagulation proteins, and diagnosis of protein S deficiency and quantification of the associated thrombotic risk are not straightforward. In this review, the regulation of thrombin generation by protein S and the pathophysiological implications of protein S deficiency are discussed in the light of recent findings on the anticoagulant function(s) of protein S. RECENT FINDINGS: Protein S expresses both activated protein C-dependent and activated protein C-independent anticoagulant activities, but the former is generally believed to be lost upon binding of protein S to C4b-binding protein. Recently it has been shown that protein S acts as a cofactor of tissue factor pathway inhibitor in the down regulation of factor X-activation, which provides a mechanistic basis for the activated protein C-independent anticoagulant activity of protein S in plasma. In addition, reevaluation of the role of the protein S/C4b-binding protein complex has demonstrated that C4b-binding protein-bound protein S does express activated protein C-cofactor activity, especially during the inactivation of factor Va Leiden. SUMMARY: These findings underscore the central role of protein S in the regulation of coagulation and may have important implications for the evaluation of the thrombotic risk associated with protein S deficiency.


Asunto(s)
Coagulación Sanguínea , Proteína S/fisiología , Humanos , Deficiencia de Proteína S/fisiopatología , Trombina/metabolismo
16.
J Clin Invest ; 95(5): 1987-93, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7738165

RESUMEN

The antithrombotic effects of bovine activated protein C (APC) and protein S were investigated in a rabbit model of microarterial thrombosis. Because of the species specificity of the APC-protein S interaction, bovine APC expresses potent anticoagulant activity in rabbit plasma only when bovine protein S is also present. This provided a way to assess the contribution of bovine protein S to the antithrombotic effect of bovine APC. Rabbits were infused with boluses of activated protein C (0.1, 0.2, 0.4, or 0.8 mg/kg), protein S (0.5 mg/kg), or activated protein C (0.1 or 0.01 mg/kg) plus protein S (0.5 mg/kg). APC alone produced a dose-dependent antithrombotic effect, but only the group receiving the highest dose differed significantly from controls. While a low dose of activated protein C (0.1 mg/kg) alone had no antithrombotic effect, together with protein S (0.5 mg/kg) it produced a potent response. The presented results demonstrate the in vivo significance of protein S as a cofactor to activated protein C. The data show that a potent antithrombotic effect, without hemorrhagic side effects or significant systemic anticoagulation, may be achieved by low doses of activated protein C when combined with protein S.


Asunto(s)
Arterias/fisiología , Proteína C/farmacología , Proteína S/farmacología , Trombosis/prevención & control , Animales , Arterias/efectos de los fármacos , Arterias/fisiopatología , Bovinos , Ensayo de Inmunoadsorción Enzimática , Femenino , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Músculo Liso Vascular/fisiopatología , Proteína C/fisiología , Proteína S/fisiología , Conejos , Reperfusión , Factores de Tiempo
17.
FEBS Lett ; 579(27): 6023-6, 2005 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-16229836

RESUMEN

Protein S, a cofactor for activated protein C (aPC) to inactivate coagulation factors, also plays a pivotal role in inflammation. Based on our recent findings that aPC and protein S modifies tissue plasminogen activator (tPA)-catalyzed activation of Glu-plasminogen (Glu-plg), we analyzed possible role of protein S in cell-associated plasminogen activation and invasive potential of inflammatory cells. Monocyte-like THP-1 cells, to which both plasminogen and tPA bind, enhanced tPA-catalyzed plasminogen activation, which was partially abolished by protein S but not by aPC. Protein S attenuated both the plasminogen binding to THP-1 cells and associated their invasive potential through Matrigel.


Asunto(s)
Movimiento Celular , Monocitos/fisiología , Plasminógeno/antagonistas & inhibidores , Proteína S/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Humanos , Monocitos/efectos de los fármacos , Plasminógeno/metabolismo , Proteína C/metabolismo , Proteína S/metabolismo , Proteína S/farmacología
18.
J Thromb Haemost ; 3(12): 2712-20, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16359509

RESUMEN

BACKGROUND: Protein S is a vitamin K-dependent protein with anticoagulant properties. It contains a so-called thrombin-sensitive region (TSR), which is susceptible to cleavage by coagulation factor Xa (FXa) and thrombin. Upon cleavage, the anticoagulant activity of protein S is abolished. OBJECTIVE: The aim of the present study was to determine whether protein S is cleaved within the TSR during activation of the coagulation system under near physiological conditions. RESULTS: In a reconstituted coagulation system containing apart from protein S only procoagulant constituents and synthetic phospholipid vesicles, protein S was cleaved at Arg60 by the FXa generated (3 mol min(-1) mol(-1) enzyme). FXa-catalyzed cleavage of protein S, however, was inhibited by factor Va and prothrombin by more than 70%. During clotting of recalcified citrated plasma in the presence of a synthetic lipid membrane, no FXa-catalyzed proteolysis of protein S was observed. Substituting platelets for phospholipid vesicles resulted both in the reconstituted system and in plasma in cleavage of the TSR. Cleavage was at Arg60 and was observed upon platelet activation, irrespective of the presence of FXa (13 pmol min(-1) 10(-8) platelets). No cleavage by thrombin was observed in either the reconstituted coagulation system or clotting plasma. CONCLUSION: These findings suggest that in vivo the anticoagulant activity of protein S is not down-regulated by FXa or thrombin during activation of coagulation. Our results rather suggest a role for a platelet protease in down-regulating the anticoagulant activity of protein S during the hemostatic response.


Asunto(s)
Factor Xa/metabolismo , Hemostasis , Proteína S/metabolismo , Trombina/metabolismo , Sitios de Unión , Coagulación Sanguínea , Plaquetas/citología , Plaquetas/fisiología , Células Cultivadas , Endotelio Vascular/citología , Humanos , Cinética , Liposomas/farmacología , Proteína S/fisiología
19.
Arterioscler Thromb Vasc Biol ; 22(4): 530-8, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11950687

RESUMEN

The generation of thrombin by the prothrombinase complex constitutes an essential step in hemostasis, with thrombin being crucial for the amplification of blood coagulation, fibrin formation, and platelet activation. In the prothrombinase complex, the activated form of coagulation factor V (FVa) is an essential cofactor to the enzyme-activated factor X (FXa), FXa being virtually ineffective in the absence of its cofactor. Besides its procoagulant potential, intact factor V (FV) has an anticoagulant cofactor capacity functioning in synergy with protein S and activated protein C (APC) in APC-catalyzed inactivation of the activated form of factor VIII. The expression of anticoagulant cofactor function of FV is dependent on APC-mediated proteolysis of intact FV. Thus, FV has the potential to function in procoagulant and anticoagulant pathways, with its functional properties being modulated by proteolysis exerted by procoagulant and anticoagulant enzymes. The procoagulant enzymes factor Xa and thrombin are both able to activate circulating FV to FVa. The activity of FVa is, in turn, regulated by APC together with its cofactor protein S. In fact, the regulation of thrombin formation proceeds primarily through the upregulation and downregulation of FVa cofactor activity, and failure to control FVa activity may result in either bleeding or thrombotic complications. A prime example is APC resistance, which is the most common genetic risk factor for thrombosis. It is caused by a single point mutation in the FV gene (factor V(Leiden)) that not only renders FVa less susceptible to the proteolytic inactivation by APC but also impairs the anticoagulant properties of FV. This review gives a description of the dualistic character of FV and describes the gene-gene and gene-environment interactions that are important for the involvement of FV in the etiology of venous thromboembolism.


Asunto(s)
Factor V/fisiología , Trombosis de la Vena/sangre , Resistencia a la Proteína C Activada/genética , Coagulación Sanguínea/fisiología , Factor V/biosíntesis , Factor V/genética , Factor V/metabolismo , Factor VIII/genética , Factor VIII/fisiología , Factor Va/genética , Factor Va/fisiología , Factor Xa/metabolismo , Factor Xa/fisiología , Humanos , Fenotipo , Mutación Puntual , Proteína C/genética , Proteína C/fisiología , Proteína S/fisiología , Trombina/metabolismo , Trombosis de la Vena/genética , Población Blanca
20.
Semin Hematol ; 34(3): 256-64, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9241710

RESUMEN

Familial thrombosis has long been considered as an autosomal dominant trait, caused by a dominant gene defect with a reduced penetrance for the disease. Recently, this view has changed and today familial thrombophilia is considered as a complex genetic disorder caused by the segregation of two or more gene defects (known and unknown) in a family. Here we briefly discuss the known genetic defects (protein C, protein S, and antithrombin deficiency and activated protein C resistance associated with factor V Leiden) with special focus on the relation between gene mutation and plasma abnormality and on the association between abnormality and thrombosis in affected families and in the population. Finally, the evidence is reviewed that indicates familial thrombosis as an oligogenetic disorder and on the basis of these data strategies are discussed for the identification of new genetic risk factors for thrombosis via a genetic approach.


Asunto(s)
Tromboflebitis/genética , Trombosis/genética , Antitrombina III/genética , Antitrombina III/fisiología , Deficiencia de Antitrombina III , Factor V/genética , Factor V/fisiología , Salud de la Familia , Humanos , Proteína C/genética , Proteína C/fisiología , Proteína S/genética , Proteína S/fisiología , Tromboflebitis/sangre , Tromboflebitis/fisiopatología , Trombosis/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA