Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Thromb Haemost ; 21(5): 1200-1213, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36696212

RESUMEN

BACKGROUND: Titanium (Ti) and its alloys are widely used in manufacturing medical devices because of their strength and resistance to corrosion. Although Ti compounds are considered compatible with blood, they appear to support plasma contact activation and may be thrombogenic. OBJECTIVES: The objective of this study was to compare Ti and titanium nitride (TiN) with known activators of contact activation (kaolin and silica) in plasma-clotting assays and to assess binding and activation of factor XII, (FXII), factor XI (FXI), prekallikrein, and high-molecular-weight kininogen (HK) with Ti/TiN. METHODS: Ti-based nanospheres and foils were compared with kaolin, silica, and aluminum in plasma-clotting assays. Binding and activation of FXII, prekallikrein, HK, and FXI to surfaces was assessed with western blots and chromogenic assays. RESULTS: Using equivalent surface amounts, Ti and TiN were comparable with kaolin and superior to silica, for inducing coagulation and FXII autoactivation. Similar to many inducers of contact activation, Ti and TiN are negatively charged; however, their effects on FXII are not neutralized by the polycation polybrene. Antibodies to FXII, prekallikrein, or FXI or coating Ti with poly-L-arginine blocked Ti-induced coagulation. An antibody to FXII reduced FXII and PK binding to Ti, kallikrein generation, and HK cleavage. CONCLUSION: Titanium compounds induce contact activation with a potency comparable with that of kaolin. Binding of FXII with Ti shares some features with FXII binding to soluble polyanions but may have unique features. Inhibitors targeting FXII or FXI may be useful in mitigating Ti-induced contact activation in patients with titanium-based implants that are exposed to blood.


Asunto(s)
Caolín , Precalicreína , Humanos , Factor XI/metabolismo , Factor XII/metabolismo , Precalicreína/metabolismo , Titanio
2.
JRSM Cardiovasc Dis ; 9: 2048004020906994, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32110389

RESUMEN

PURPOSE: Chronic thromboembolic pulmonary hypertension is characterized by incomplete thrombus resolution following acute pulmonary embolism, leading to pulmonary hypertension and right ventricular dysfunction. Conditions such as thrombophilias, dysfibrinogenemias, and inflammatory states have been associated with chronic thromboembolic pulmonary hypertension, but molecular mechanisms underlying this disease are poorly understood. We sought to characterize the molecular and functional features associated with chronic thromboembolic pulmonary hypertension using a multifaceted approach. METHODS: We utilized functional assays to compare clot lysis times between chronic thromboembolic pulmonary hypertension patients and multiple controls. We then performed immunohistochemical characterization of tissue from chronic thromboembolic pulmonary hypertension, pulmonary arterial hypertension, and healthy controls, and examined RNA expression patterns of cultured lymphocytes and pulmonary arterial specimens. We then confirmed RNA expression changes using immunohistochemistry, immunofluorescence, and Western blotting in pulmonary arterial tissue. RESULTS: Clot lysis times in chronic thromboembolic pulmonary hypertension patients are similar to multiple controls. Chronic thromboembolic pulmonary hypertension endarterectomized tissue has reduced expression of both smooth muscle and endothelial cell markers. RNA expression profiles in pulmonary arteries and peripheral blood lymphocytes identified differences in RNA transcript levels related to inflammation and growth factor signaling, which we confirmed using immunohistochemistry. Gene expression data also suggested significant alterations in metabolic pathways, and immunofluorescence and Western blot experiments confirmed that unglycosylated CD36 and adiponectin expression were increased in chronic thromboembolic pulmonary hypertension versus controls. CONCLUSIONS: Our data do not support impaired clot lysis underlying chronic thromboembolic pulmonary hypertension, but did demonstrate distinct molecular patterns present both in peripheral blood and in pathologic specimens of chronic thromboembolic pulmonary hypertension patients suggesting that altered metabolism may play a role in chronic thromboembolic pulmonary hypertension pathogenesis.

3.
Blood ; 135(8): 558-567, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31800958

RESUMEN

Prekallikrein (PK) is the precursor of the trypsin-like plasma protease kallikrein (PKa), which cleaves kininogens to release bradykinin and converts the protease precursor factor XII (FXII) to the enzyme FXIIa. PK and FXII undergo reciprocal conversion to their active forms (PKa and FXIIa) by a process that is accelerated by a variety of biological and artificial surfaces. The surface-mediated process is referred to as contact activation. Previously, we showed that FXII expresses a low level of proteolytic activity (independently of FXIIa) that may initiate reciprocal activation with PK. The current study was undertaken to determine whether PK expresses similar activity. Recombinant PK that cannot be converted to PKa was prepared by replacing Arg371 with alanine at the activation cleavage site (PK-R371A, or single-chain PK). Despite being constrained to the single-chain precursor form, PK-R371A cleaves high-molecular-weight kininogen (HK) to release bradykinin with a catalytic efficiency ∼1500-fold lower than that of kallikrein cleavage of HK. In the presence of a surface, PK-R371A converts FXII to FXIIa with a specific activity ∼4 orders of magnitude lower than for PKa cleavage of FXII. These results support the notion that activity intrinsic to PK and FXII can initiate reciprocal activation of FXII and PK in solution or on a surface. The findings are consistent with the hypothesis that the putative zymogens of many trypsin-like proteases are actually active proteases, explaining their capacity to undergo processes such as autoactivation and to initiate enzyme cascades.


Asunto(s)
Coagulación Sanguínea , Bradiquinina/metabolismo , Precalicreína/metabolismo , Sustitución de Aminoácidos , Animales , Factor XII/metabolismo , Células HEK293 , Humanos , Quininógeno de Alto Peso Molecular/metabolismo , Ratones Endogámicos C57BL , Precalicreína/química , Precalicreína/genética , Proteolisis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
J Thromb Haemost ; 17(9): 1449-1460, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31125187

RESUMEN

BACKGROUND: The homologous plasma proteins prekallikrein and factor XI (FXI) circulate as complexes with high molecular weight kininogen. Although evidence supports an interaction between the prekallikrein-kininogen complexes and vascular endothelium, there is conflicting information regarding FXI binding to endothelium. OBJECTIVE: To study the interaction between FXI and blood vessels in mice. METHODS: C57Bl/6 wild-type or F11-/- mice in which variants of FXI were expressed by hydrodynamic tail vein injection, received intravenous infusions of saline, heparin, polyphosphates, protamine, or enzymes that digest glycosaminoglycans (GAGs). Blood was collected after infusion and plasma was analyzed by western blot for FXI. RESULTS AND CONCLUSIONS: Plasma FXI increased 5- to 10-fold in wild-type mice after infusion of heparin, polyphosphates, protamine, or GAG-digesting enzymes, but not saline. Similar treatments resulted in a much smaller change in plasma FXI levels in rats, and infusions of large boluses of heparin did not change FXI levels appreciably in baboons or humans. The releasable FXI fraction was reconstituted in F11-/- mice by expressing murine FXI, but not human FXI. We identified a cluster of basic residues on the apple 4 domain of mouse FXI that is not present in other species. Replacing the basic residues with alanine prevented the interaction of mouse FXI with blood vessels, whereas introducing the basic residues into human FXI allowed it to bind to blood vessels. Most FXI in mice is noncovalently associated with GAGs on blood vessel endothelium and does not circulate in plasma.


Asunto(s)
Endotelio Vascular/metabolismo , Factor XI/metabolismo , Glicosaminoglicanos/sangre , Animales , Sitios de Unión , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/inducido químicamente , Cloruros/toxicidad , Factor XI/química , Deficiencia del Factor XI/sangre , Compuestos Férricos/toxicidad , Heparina/farmacología , Humanos , Quininógenos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Papio , Precalicreína/metabolismo , Unión Proteica , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Especificidad de la Especie , Electricidad Estática
5.
Blood ; 133(10): 1152-1163, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30591525

RESUMEN

The plasma proteins factor XII (FXII) and prekallikrein (PK) undergo reciprocal activation to the proteases FXIIa and kallikrein by a process that is enhanced by surfaces (contact activation) and regulated by the serpin C1 inhibitor. Kallikrein cleaves high-molecular-weight kininogen (HK), releasing the vasoactive peptide bradykinin. Patients with hereditary angioedema (HAE) experience episodes of soft tissue swelling as a consequence of unregulated kallikrein activity or increased prekallikrein activation. Although most HAE cases are caused by reduced plasma C1-inhibitor activity, HAE has been linked to lysine/arginine substitutions for Thr309 in FXII (FXII-Lys/Arg309). Here, we show that FXII-Lys/Arg309 is susceptible to cleavage after residue 309 by coagulation proteases (thrombin and FXIa), resulting in generation of a truncated form of FXII (δFXII). The catalytic efficiency of δFXII activation by kallikrein is 15-fold greater than for full-length FXII. The enhanced rate of reciprocal activation of PK and δFXII in human plasma and in mice appears to overwhelm the normal inhibitory function of C1 inhibitor, leading to increased HK cleavage. In mice given human FXII-Lys/Arg309, induction of thrombin generation by infusion of tissue factor results in enhanced HK cleavage as a consequence of δFXII formation. The effects of δFXII in vitro and in vivo are reproduced when wild-type FXII is bound by an antibody to the FXII heavy chain (HC; 15H8). The results contribute to our understanding of the predisposition of patients carrying FXII-Lys/Arg309 to angioedema after trauma, and reveal a regulatory function for the FXII HC that normally limits PK activation in plasma.


Asunto(s)
Factor XII/química , Factor XIa/química , Angioedema Hereditario Tipo III/sangre , Angioedema Hereditario Tipo III/genética , Angioedemas Hereditarios , Animales , Arginina/química , Coagulación Sanguínea , Bradiquinina/sangre , Catálisis , Proteína Inhibidora del Complemento C1/química , Factor XIIa/química , Células HEK293 , Humanos , Quininógenos/sangre , Lisina/química , Ratones , Ratones Endogámicos C57BL , Calicreína Plasmática/química , Precalicreína/química , Unión Proteica , Proteínas Recombinantes/química , Propiedades de Superficie , Trombina/genética
6.
Oncotarget ; 10(66): 7031-7042, 2019 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-31903163

RESUMEN

Growth arrest-specific 6 (Gas6) has been implicated in carcinogenesis through activation of its receptors, particularly MerTK. To investigate whether Gas6 plays a role in resistance to NF-κB inhibitors, which have not proven to be effective agents for lung cancer therapy, we studied lung cancer models induced by urethane injection or expression of mutant Kras (KrasG12D). We found that Gas6 is primarily produced by macrophages during tumorigenesis and that Gas6 is negatively regulated by NF-κB. Since Gas6 is a vitamin K dependent protein, we used low-dose warfarin to block Gas6 production and showed that this treatment inhibited tumorigenesis in both the urethane and KrasG12D models, most prominently in mice with targeted deletion of IKKß in myeloid cells (IKKßΔMye mice). In addition, MerTK deficient mice had reduced urethane-induced tumorigenesis. Inhibition of the Gas6-MerTK pathway in all these models reduced macrophages and neutrophils in the lungs of tumor-bearing mice. Analysis of mouse lung tumors revealed MerTK staining on tumor cells and in vitro studies showed that Gas6 increased proliferation of human lung cancer cell lines. To assess the therapeutic potential for combination treatment targeting NF-κB and Gas6-MerTK, we injected Lewis Lung Carcinoma cells subcutaneously and treated mice with Bay 11-70852 (NF-κB inhibitor) and/or Foretinib (MerTK inhibitor). While individual treatments were ineffective, combination therapy markedly reduced tumor growth, blocked tumor cell proliferation, reduced tumor-associated macrophages, and increased CD4+ T cells. Together, our studies unmask a role for Gas6-MerTK signaling in lung carcinogenesis and indicate that up-regulation of Gas6 production in macrophages could be a major mechanism of resistance to NF-κB inhibitors.

7.
Res Pract Thromb Haemost ; 2(1): 168-173, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29354798

RESUMEN

BACKGROUND: Factor XII (FXII) Locarno is a natural variant with proline replacing Arg353 at the activation cleavage site, preventing conversion to the fully active protease factor XIIa (FXIIa). Recently, we showed that FXII restricted to a single chain form (sc-FXII) by replacing Arg353 with alanine expresses proteolytic activity that is enhanced by cofactors such as polyphosphate. AIM: To determine if the Pro353 substitution affects the activity of sc-FXII. METHODS: Wild type FXII (FXII-WT), FXII-R353A, and FXII Locarno (FXII-R353P) were tested for their abilities to activate prekallikrein, and to induce thrombin generation and coagulation in plasma in a factor XI-dependent manner. RESULTS: FXII-WT is converted to FXIIa by autoactivation in the presence of polyphosphate, and by incubation with kallikrein. FXII-R353P and FXII-R353A were not converted to FXIIa by these methods. Despite this, FXII-R353A converts prekallikrein to kallikrein, and the reaction is enhanced by polyphosphate. FXII-R353P also converts prekallikrein to kallikrein, but at a slower rate than FXII-R353A. In FXII-deficient plasma induced to clot with silica, FXII-R353A is a better promoter of factor XI-dependent thrombin generation and coagulation than FXII-R353P. CONCLUSION: The activity of sc-FXII is sensitive to perturbations in the activation loop, which contains residue 353. Homology modeling based on the crystal structure of the FXII homolog tissue plasminogen activator suggests that Pro353 introduces changes in the shape and flexibility of the activation loop that disrupt key interactions that support an active conformation in sc-FXII.

8.
Thromb Res ; 161: 94-105, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29223926

RESUMEN

Factor XI (FXI) is the zymogen of a plasma protease, factor XIa (FXIa), that contributes to thrombin generation during blood coagulation by proteolytic activation of several coagulation factors, most notably factor IX (FIX). FXI is a homolog of prekallikrein (PK), a component of the plasma kallikrein-kinin system. While sharing structural and functional features with PK, FXI has undergone adaptive changes that allow it to contribute to blood coagulation. Here we review current understanding of the biology and enzymology of FXI, with an emphasis on structural features of the protein as they relate to protease function.


Asunto(s)
Factor XI/genética , Factor XI/metabolismo , Humanos , Procesamiento Proteico-Postraduccional
9.
Curr Opin Hematol ; 24(5): 411-418, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28604413

RESUMEN

PURPOSE OF REVIEW: Exposure of blood to foreign surfaces induces reciprocal conversion of the plasma proteins factor XII (fXII) and plasma prekallikrein (PPK) to the proteases α-fXIIa and α-kallikrein. This process, called contact activation, has a range of effects on host defence mechanisms, including promoting coagulation. The nature of the triggering mechanism for contact activation is debated. One hypothesis predicts that fXII has protease activity, either intrinsically or upon surface-binding, that initiates contact activation. We tested this by assessing the proteolytic activity of a recombinant fXII variant that cannot be converted to α-fXIIa. RECENT FINDINGS: The proteolytic activity of fXII-T (for 'triple' mutant), a variant with alanine substitutions for arginine at activation cleavage sites (Arg334, Arg344, and Arg353) was tested with known α-fXIIa substrates. FXII-T activates PPK in solution, and the reaction is enhanced by polyphosphate, an inducer of contact activation released from platelets. In the presence of polyphosphate, fXII-T converts fXII to α-fXIIa, and also converts the coagulation protein factor XI to its active form. SUMMARY: The findings support the hypothesis that contact activation is initiated through activity intrinsic to single-chain fXII, and indicate that preexisting α-fXIIa is not required for induction of contact activation.


Asunto(s)
Factor XIIa/metabolismo , Calicreína Plasmática/metabolismo , Precalicreína/metabolismo , Factor XIIa/genética , Humanos , Calicreína Plasmática/genética , Precalicreína/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 312(5): H907-H918, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28235789

RESUMEN

Immune activation in chronic systolic heart failure (HF) correlates with disease severity and prognosis. Recombinant neuregulin-1 (rNRG-1) is being developed as a possible therapy for HF, based on the activation of ERBB receptors in cardiac cells. Work in animal models of HF led us to hypothesize that there may be direct effects of NRG-1 on immune system activation and inflammation. We investigated the expression of ERBB receptors and the effect of rNRG-1 isoform glial growth factor 2 (GGF2) in subpopulations of peripheral blood mononuclear cells (PB MNCs) in subjects with HF. We found that human monocytes express both ERBB2 and ERBB3 receptors, with high interindividual variability among subjects. Monocyte surface ERBB3 and TNF-α mRNA expression were inversely correlated in subjects with HF but not in human subjects without HF. GGF2 activation of ERBB signaling ex vivo inhibited LPS-induced TNF-α production, specifically in the CD14lowCD16+ population of monocytes in a phosphoinositide 3-kinase-dependent manner. GGF2 suppression of TNF-α correlated directly with the expression of ERBB3. In vivo, a single dose of intravenous GGF2 reduced TNF-α expression in PB MNCs of HF subjects participating in a phase I safety study of GGF2. These results support a role for ERBB3 signaling in the regulation of TNF-α production from CD14lowCD16+ monocytes and a need for further investigation into the clinical significance of NRG-1/ERBB signaling as a modulator of immune system function.NEW & NOTEWORTHY This study identified a novel role of neuregulin-1 (NRG-1)/ERBB signaling in the control of proinflammatory activation of monocytes. These results further improve our fundamental understanding of cardioprotective effects of NRG-1 in patients with heart failure.


Asunto(s)
Receptores ErbB/biosíntesis , Inflamación/fisiopatología , Monocitos , Transducción de Señal , Receptores ErbB/efectos de los fármacos , Receptores ErbB/genética , Femenino , Humanos , Técnicas In Vitro , Activación de Macrófagos , Masculino , Persona de Mediana Edad , Neurregulina-1/metabolismo , Neurregulina-1/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor ErbB-2/biosíntesis , Receptor ErbB-2/genética , Receptor ErbB-3/biosíntesis , Receptor ErbB-3/genética , Proteínas Recombinantes/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis
11.
Thromb Haemost ; 117(4): 671-681, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28124063

RESUMEN

The plasma zymogens factor XI (fXI) and prekallikrein (PK) are activated by factor XIIa (fXIIa) during contact activation. Polyanions such as DNA and RNA may contribute to thrombosis and inflammation partly by enhancing PK and fXI activation. We examined PK and fXI activation in the presence of nucleic acids, and determine the effects of the cofactor high molecular weight kininogen (HK) on the reactions. In the absence of HK, DNA and RNA induced fXI autoactivation. Proteases known to activate fXI (fXIIa and thrombin) did not enhance this process appreciably. Nucleic acids had little effect on PK activation by fXIIa in the absence of HK. HK had significant but opposite effects on PK and fXI activation. HK enhanced fXIIa activation of PK in the presence of nucleic acids, but blocked fXI autoactivation. Thrombin and fXIIa could overcome the HK inhibitory effect on autoactivation, indicating these proteases are necessary for nucleic acid-induced fXI activation in an HK-rich environment such as plasma. In contrast to PK, which requires HK for optimal activation, fXI activation in the presence of nucleic acids depends on anion binding sites on the fXI molecule. The corresponding sites on PK are not necessary for PK activation. Our results indicate that HK functions as a cofactor for PK activation in the presence of nucleic acids in a manner consistent with classic models of contact activation. However, HK has, on balance, an inhibitory effect on nucleic acid-supported fXI activation and may function as a negative regulator of fXI activation.


Asunto(s)
Coagulación Sanguínea , ADN/metabolismo , Factor XIa/metabolismo , Quininógeno de Alto Peso Molecular/metabolismo , Precalicreína/metabolismo , ARN/metabolismo , Activación Enzimática , Factor XIIa/metabolismo , Humanos , Trombina/metabolismo , Factores de Tiempo
12.
Blood ; 129(11): 1527-1537, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28069606

RESUMEN

When blood is exposed to variety of artificial surfaces and biologic substances, the plasma proteins factor XII (FXII) and prekallikrein undergo reciprocal proteolytic conversion to the proteases αFXIIa and α-kallikrein by a process called contact activation. These enzymes contribute to host-defense responses including coagulation, inflammation, and fibrinolysis. The initiating event in contact activation is debated. To test the hypothesis that single-chain FXII expresses activity that could initiate contact activation, we prepared human FXII variants lacking the Arg353 cleavage site required for conversion to αFXIIa (FXII-R353A), or lacking the 3 known cleavage sites at Arg334, Arg343, and Arg353 (FXII-T, for "triple" mutant), and compared their properties to wild-type αFXIIa. In the absence of a surface, FXII-R353A and FXII-T activate prekallikrein and cleave the tripeptide S-2302, demonstrating proteolytic activity. The activity is several orders of magnitude weaker than that of αFXIIa. Polyphosphate, an inducer of contact activation, enhances PK activation by FXII-T, and facilitates FXII-T activation of FXII and FXI. In plasma, FXII-T and FXII-R353A, but not FXII lacking the active site serine residue (FXII-S544A), shortened the clotting time of FXII-deficient plasma and enhanced thrombin generation in a surface-dependent manner. The effect was not as strong as for wild-type FXII. Our results support a model for induction of contact activation in which activity intrinsic to single-chain FXII initiates αFXIIa and α-kallikrein formation on a surface. αFXIIa, with support from α-kallikrein, subsequently accelerates contact activation and is responsible for the full procoagulant activity of FXII.


Asunto(s)
Coagulación Sanguínea , Factor XII/metabolismo , Proteolisis , Dominio Catalítico/genética , Factor XIIa/metabolismo , Humanos , Calicreínas/metabolismo , Propiedades de Superficie
13.
J Appl Lab Med ; 2(3): 380-385, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33636847

RESUMEN

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a hypercoagulable state caused by a transient antibody to heparin-bound platelet factor 4 (PF4). Treatment involves discontinuing heparin and administering a nonheparin anticoagulant. Procedures requiring heparin, such as cardiopulmonary bypass, are preferably delayed until the offending antibody is no longer detectable. For patients with a high-titer anti-PF4-heparin antibody and who require exposure to heparin, therapeutic plasma exchange (TPE) has been used to remove the antibody. Recent work indicates that a functional assay for detecting platelet-activating antibodies in HIT patients, the serotonin release assay (SRA), is preferable to ELISAs for anti-PF4-heparin antibodies for following the effectiveness of plasma exchange. METHODS: Two cases of acute heparin-induced thrombocytopenia managed with plasma exchange before emergent cardiac surgery were evaluated with SRAs using a range of heparin concentrations that included those used in cardiopulmonary bypass. RESULTS: We observed that a single round of plasma exchange led to greater reduction in platelet reactivity at heparin concentrations between 1 and 3 U/mL than at lower concentrations, consistent with the impression that heparin-PF4-antibody complexes form optimally within a limited heparin concentration range. CONCLUSIONS: The findings suggest there may be a range of heparin concentration in which cardiac surgery may be safely performed in HIT patients, and that a single TPE in an emergent setting may lower antibody concentration sufficiently to lower platelet reactivity in the presence of heparin.

14.
PLoS One ; 11(4): e0152968, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27046148

RESUMEN

Sepsis, a systemic inflammatory response to infection, is often accompanied by abnormalities of blood coagulation. Prior work with a mouse model of sepsis induced by cecal ligation and puncture (CLP) suggested that the protease factor XIa contributed to disseminated intravascular coagulation (DIC) and to the cytokine response during sepsis. We investigated the importance of factor XI to cytokine and coagulation responses during the first 24 hours after CLP. Compared to wild type littermates, factor XI-deficient (FXI-/-) mice had a survival advantage after CLP, with smaller increases in plasma levels of TNF-α and IL-10 and delayed IL-1ß and IL-6 responses. Plasma levels of serum amyloid P, an acute phase protein, were increased in wild type mice 24 hours post-CLP, but not in FXI-/- mice, supporting the impression of a reduced inflammatory response in the absence of factor XI. Surprisingly, there was little evidence of DIC in mice of either genotype. Plasma levels of the contact factors factor XII and prekallikrein were reduced in WT mice after CLP, consistent with induction of contact activation. However, factor XII and PK levels were not reduced in FXI-/- animals, indicating factor XI deficiency blunted contact activation. Intravenous infusion of polyphosphate into WT mice also induced changes in factor XII, but had much less effect in FXI deficient mice. In vitro analysis revealed that factor XIa activates factor XII, and that this reaction is enhanced by polyanions such polyphosphate and nucleic acids. These data suggest that factor XI deficiency confers a survival advantage in the CLP sepsis model by altering the cytokine response to infection and blunting activation of the contact (kallikrein-kinin) system. The findings support the hypothesis that factor XI functions as a bidirectional interface between contact activation and thrombin generation, allowing the two processes to influence each other.


Asunto(s)
Coinfección/metabolismo , Citocinas/metabolismo , Deficiencia del Factor XI/metabolismo , Péptido Hidrolasas/metabolismo , Sepsis/metabolismo , Animales , Coinfección/enzimología , Activación Enzimática , Ratones , Sepsis/enzimología
15.
Thromb Res ; 140: 118-124, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26950760

RESUMEN

Studies with animal models implicate the plasma proteases factor XIIa (FXIIa) and α-kallikrein in arterial and venous thrombosis. As congenital deficiencies of factor XII (FXII) or prekallikrein (PK), the zymogens of FXIIa and α-kallikrein respectively, do not cause bleeding disorders, inhibition of these enzymes may have therapeutic benefit without compromising hemostasis. The relative contributions of FXIIa and α-kallikrein to thrombosis in animal models are not clear. We compared mice lacking FXII or PK to wild type mice in established models of arterial thrombosis. Wild type mice developed carotid artery occlusion when the vessel was exposed to a 3.5% solution of ferric chloride (FeCl3). FXII-deficient mice were resistant to occlusion at 5% FeCl3 and partially resistant at 10% FeCl3. PK-deficient mice were resistant at 3.5% FeCl3 and partially resistant at 5% FeCl3. Mice lacking high molecular weight kininogen, a cofactor for PK activation and activity, were also partially resistant to thrombosis at 5% FeCl3. Induction of carotid artery thrombosis with Rose Bengal was delayed in FXII-deficient mice compared to wild type or PK-deficient animals. In human plasma supplemented with silica, DNA or collagen to induce contact activation, an antibody to the FXIIa active site was more effective at preventing thrombin generation than an antibody to the α-kallikrein active site. Similarly, the FXIIa antibody was more effective at reducing fibrin formation in human blood flowing through collagen coated-tubes. The findings suggest that inhibitors of FXIIa will have more potent anti-thrombotic effects than inhibitors of α-kallikrein.


Asunto(s)
Trastornos de la Coagulación Sanguínea/complicaciones , Deficiencia del Factor XII/complicaciones , Factor XII/genética , Precalicreína/deficiencia , Precalicreína/genética , Trombosis/etiología , Trombosis/genética , Animales , Coagulación Sanguínea/efectos de los fármacos , Trastornos de la Coagulación Sanguínea/genética , Factor XII/antagonistas & inhibidores , Deficiencia del Factor XII/genética , Eliminación de Gen , Humanos , Calicreínas/antagonistas & inhibidores , Calicreínas/genética , Ratones , Ratones Endogámicos C57BL , Precalicreína/antagonistas & inhibidores , Factores Protectores , Trombosis/prevención & control
16.
Biochemistry ; 54(36): 5578-88, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26295742

RESUMEN

With a newer, more selective and efficacious cytosolic phospholipase A2α (cPLA2α) inhibitor available, we revisited the role of cPLA2α activity in platelet activation and discovered that a component of platelet signaling, even larger than previously appreciated, relies on this enzyme. In a whole blood shear-based flow chamber assay, giripladib, a cPLA2α inhibitor, reduced platelet adhesion and accumulation on collagen. Moreover, giripladib differentially affected P-selectin expression and GPIIbIIIa activation depending on the agonist employed. While protease-activated receptor 1 (PAR1)-mediated platelet activation was unaffected by giripladib, the levels of PAR4- and GPVI-mediated platelet activation were significantly reduced. Meanwhile, the thromboxane A2 receptor antagonist SQ29548 had no effect on PAR-, GPVI-, or puriniergic receptor-mediated platelet activation, suggesting that another eicosanoid produced downstream of arachidonic acid liberation by cPLA2α was responsible for this large component of PAR4- and GPVI-mediated platelet activation. In parallel, we profiled PAR-mediated changes in glycerophospholipid (GPL) mass with and without giripladib to better understand cPLA2α-mediated lipid metabolism. Phosphatidylcholine and phosphatidylethanolamine (PE) demonstrated the largest consumption of mass during thrombin stimulation. Additionally, we confirm phosphatidylinositol as a major substrate of cPLA2α. A comparison of PAR1- and PAR4-induced metabolism revealed the consumption of more putative arachidonyl-PE species downstream of PAR1 activation. Instead of enhanced cPLA2α activity and therefore more arachidonic acid liberation downstream of PAR4, these results indicate the major role that cPLA2α activity plays in platelet function and suggest that a novel eicosanoid is produced in response to platelet activation that represents a large component of PAR4- and GPVI-mediated responses.


Asunto(s)
Plaquetas/enzimología , Fosfolipasas A2 Grupo IV/sangre , Lípidos/sangre , Benzoatos/farmacología , Plaquetas/química , Plaquetas/efectos de los fármacos , Glicerofosfolípidos/sangre , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Humanos , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/sangre , Espectrometría de Masa por Ionización de Electrospray , Estrés Mecánico , Sulfonamidas/farmacología , Trombina/farmacología
17.
Blood ; 125(9): 1488-96, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25587039

RESUMEN

Activation of coagulation factor XI (FXI) may play a role in hemostasis. The primary substrate of activated FXI (FXIa) is FIX, leading to FX activation (FXa) and thrombin generation. However, recent studies suggest the hemostatic role of FXI may not be restricted to the activation of FIX. We explored whether FXI could interact with and inhibit the activity of tissue factor pathway inhibitor (TFPI). TFPI is an essential reversible inhibitor of activated factor X (FXa) and also inhibits the FVIIa-TF complex. We found that FXIa neutralized both endothelium- and platelet-derived TFPI by cleaving the protein between the Kunitz (K) 1 and K2 domains (Lys86/Thr87) and at the active sites of the K2 (Arg107/Gly108) and K3 (Arg199/Ala200) domains. Addition of FXIa to plasma was able to reverse the ability of TFPI to prolong TF-initiated clotting times in FXI- or FIX-deficient plasma, as well as FXa-initiated clotting times in FX-deficient plasma. Treatment of cultured endothelial cells with FXIa increased the generation of FXa and promoted TF-dependent fibrin formation in recalcified plasma. Together, these results suggest that the hemostatic role of FXIa may be attributed not only to activation of FIX but also to promoting the extrinsic pathway of thrombin generation through inactivation of TFPI.


Asunto(s)
Coagulación Sanguínea/fisiología , Plaquetas/metabolismo , Factor IX/metabolismo , Factor XIa/metabolismo , Factor Xa/metabolismo , Fibrina/metabolismo , Lipoproteínas/metabolismo , Plaquetas/citología , Western Blotting , Células Cultivadas , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Lipoproteínas/genética , Mutación/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
18.
ASAIO J ; 61(1): e1-4, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25251586

RESUMEN

All patients supported with continuous flow-left ventricular assist devices (CF-LVADs) develop acquired von Willebrand syndrome due to the loss of von Willebrand factor (vWF) high molecular weight (HMW) multimers and this phenomenon has been associated with clinical manifestation of bleeding diatheses. The precise timing of postoperative recovery of HMW multimers and correction of this condition after CF-LVAD explantation and heart transplantation is unknown. We sought to determine the specific timing of HMW multimer recovery by serially quantifying plasma vWF multimer ratios after CF-LVAD explant and orthotopic heart transplantation (OHT) in a patient implanted with a HeartWare ventricular assist device. Using densitometric analysis of multimer patterns, we demonstrated complete recovery of HMW multimers within the first few hours following CF-LVAD explant and OHT. These findings have critical implications in the context of perioperative bleeding diatheses in patients bridged to transplantation with a CF-LVAD.


Asunto(s)
Trasplante de Corazón , Corazón Auxiliar/efectos adversos , Enfermedades de von Willebrand/etiología , Humanos , Masculino , Persona de Mediana Edad , Peso Molecular , Proteínas de Resistencia a Mixovirus , Hemorragia Posoperatoria/sangre , Hemorragia Posoperatoria/etiología , Choque Cardiogénico/cirugía , Enfermedades de von Willebrand/sangre , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo
19.
PLoS One ; 9(11): e112753, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386896

RESUMEN

CD148 is a transmembrane tyrosine phosphatase that is expressed at cell junctions. Recent studies have shown that CD148 associates with the cadherin/catenin complex and p120 catenin (p120) may serve as a substrate. However, the role of CD148 in cadherin cell-cell adhesion remains unknown. Therefore, here we addressed this issue using a series of stable cells and cell-based assays. Wild-type (WT) and catalytically inactive (CS) CD148 were introduced to A431D (lacking classical cadherins), A431D/E-cadherin WT (expressing wild-type E-cadherin), and A431D/E-cadherin 764AAA (expressing p120-uncoupled E-cadherin mutant) cells. The effects of CD148 in cadherin adhesion were assessed by Ca2+ switch and cell aggregation assays. Phosphorylation of E-cadherin/catenin complex and Rho family GTPase activities were also examined. Although CD148 introduction did not alter the expression levels and complex formation of E-cadherin, p120, and ß-catenin, CD148 WT, but not CS, promoted cadherin contacts and strengthened cell-cell adhesion in A431D/E-cadherin WT cells. This effect was accompanied by an increase in Rac1, but not RhoA and Cdc42, activity and largely diminished by Rac1 inhibition. Further, we demonstrate that CD148 reduces the tyrosine phosphorylation of p120 and ß-catenin; causes the dephosphorylation of Y529 suppressive tyrosine residue in Src, a well-known CD148 site, increasing Src activity and enhancing the phosphorylation of Y228 (a Src kinase site) in p120, in E-cadherin contacts. Consistent with these findings, CD148 dephosphorylated both p120 and ß-catenin in vitro. The shRNA-mediated CD148 knockdown in A431 cells showed opposite effects. CD148 showed no effects in A431D and A431D/E-cadherin 764AAA cells. In aggregate, these findings provide the first evidence that CD148 promotes E-cadherin adhesion by regulating Rac1 activity concomitant with modulation of p120, ß-catenin, and Src tyrosine phosphorylation. This effect requires E-cadherin and p120 association.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Línea Celular , Proteínas de Drosophila/metabolismo , Humanos , Fosforilación , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Tirosina/metabolismo , beta Catenina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
20.
Blood ; 123(11): 1739-46, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24408325

RESUMEN

The plasma zymogens factor XII (fXII) and factor XI (fXI) contribute to thrombosis in a variety of mouse models. These proteins serve a limited role in hemostasis, suggesting that antithrombotic therapies targeting them may be associated with low bleeding risks. Although there is substantial epidemiologic evidence supporting a role for fXI in human thrombosis, the situation is not as clear for fXII. We generated monoclonal antibodies (9A2 and 15H8) against the human fXII heavy chain that interfere with fXII conversion to the protease factor XIIa (fXIIa). The anti-fXII antibodies were tested in models in which anti-fXI antibodies are known to have antithrombotic effects. Both anti-fXII antibodies reduced fibrin formation in human blood perfused through collagen-coated tubes. fXII-deficient mice are resistant to ferric chloride-induced arterial thrombosis, and this resistance can be reversed by infusion of human fXII. 9A2 partially blocks, and 15H8 completely blocks, the prothrombotic effect of fXII in this model. 15H8 prolonged the activated partial thromboplastin time of baboon and human plasmas. 15H8 reduced fibrin formation in collagen-coated vascular grafts inserted into arteriovenous shunts in baboons, and reduced fibrin and platelet accumulation downstream of the graft. These findings support a role for fXII in thrombus formation in primates.


Asunto(s)
Modelos Animales de Enfermedad , Deficiencia del Factor XII/complicaciones , Factor XII/antagonistas & inhibidores , Factor XII/fisiología , Trombina/metabolismo , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Coagulación Sanguínea , Factor XI/metabolismo , Factor XIIa/metabolismo , Fibrina/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Papio , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Tromboplastina/metabolismo , Trombosis/etiología , Trombosis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...