Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Allergy Clin Immunol ; 152(4): 961-971.e7, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37399947

RESUMEN

BACKGROUND: We examined how prekallikrein (PK) activation on human microvascular endothelial cells (HMVECs) is regulated by the ambient concentration of C1 inhibitor (C1INH) and prolylcarboxypeptidase (PRCP). OBJECTIVE: We sought to examine the specificity of PK activation on HMVECs by PRCP and the role of C1INH to regulate it, high-molecular-weight kininogen (HK) cleavage, and bradykinin (BK) liberation. METHODS: Investigations were performed on cultured HMVECs. Immunofluorescence, enzymatic activity assays, immunoblots, small interfering RNA knockdowns, and cell transfections were used to perform these studies. RESULTS: Cultured HMVECs constitutively coexpressed PK, HK, C1INH, and PRCP. PK activation on HMVECs was modulated by the ambient C1INH concentration. In the absence of C1INH, forming PKa on HMVECs cleaved 120-kDa HK completely to a 65-kDa H-chain and a 46-kDa L-chain in 60 minutes. In the presence of 2 µM C1INH, only 50% of the HK became cleaved. C1INH concentrations (0.0-2.5 µM) decreased but did not abolish BK liberated from HK by activated PK. Factor XII did not activate when incubated with HMVECs alone for 1 hour. However, if incubated in the presence of HK and PK, factor XII became activated. The specificity of PK activation on HMVECs by PRCP was shown by several inhibitors to each enzyme. Furthermore, PRCP small interfering RNA knockdowns magnified C1INH inhibitory activity on PK activation, and PRCP transfections reduced C1INH inhibition at any given concentration. CONCLUSIONS: These combined studies indicated that on HMVECs, PK activation and HK cleavage to liberate BK were modulated by the local concentrations of C1INH and PRCP.


Asunto(s)
Factor XII , Precalicreína , Humanos , Células Endoteliales , Bradiquinina/farmacología , Quininógeno de Alto Peso Molecular , ARN Interferente Pequeño/genética
2.
Arterioscler Thromb Vasc Biol ; 38(8): 1748-1760, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30354195

RESUMEN

Objective- Terminal complications of bacterial sepsis include development of disseminated intravascular consumptive coagulopathy. Bacterial constituents, including long-chain polyphosphates (polyP), have been shown to activate the contact pathway of coagulation in plasma. Recent work shows that activation of the contact pathway in flowing whole blood promotes thrombin generation and platelet activation and consumption distal to thrombus formation ex vivo and in vivo. Here, we sought to determine whether presence of long-chain polyP or bacteria in the bloodstream promotes platelet activation and consumption in a coagulation factor (F)XII-dependent manner. Approach and Results- Long-chain polyP promoted platelet P-selectin expression, microaggregate formation, and platelet consumption in flowing whole blood in a contact activation pathway-dependent manner. Moreover, long-chain polyP promoted local fibrin formation on collagen under shear flow in a FXI-dependent manner. Distal to the site of thrombus formation, platelet consumption was dramatically enhanced in the presence of long-chain polyP in the blood flow in a FXI- and FXII-dependent manner. In a murine model, long-chain polyP promoted platelet deposition and fibrin generation in lungs in a FXII-dependent manner. In a nonhuman primate model of bacterial sepsis, pre-treatment of animals with an antibody blocking FXI activation by FXIIa reduced lethal dose100 Staphylococcus aureus-induced platelet and fibrinogen consumption. Conclusions- This study demonstrates that bacterial-type long-chain polyP promotes platelet activation in a FXII-dependent manner in flowing blood, which may contribute to sepsis-associated thrombotic processes, consumptive coagulopathy, and thrombocytopenia.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Factor XII/metabolismo , Factor XIIa/metabolismo , Activación Plaquetaria/efectos de los fármacos , Polifosfatos/toxicidad , Trombosis/inducido químicamente , Animales , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor XII/genética , Factor XIIa/genética , Femenino , Fibrina/metabolismo , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Papio ursinus , Precalicreína/genética , Precalicreína/metabolismo , Embolia Pulmonar/sangre , Embolia Pulmonar/inducido químicamente , Embolia Pulmonar/genética , Sepsis/sangre , Sepsis/microbiología , Transducción de Señal/efectos de los fármacos , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/microbiología , Trombosis/sangre , Trombosis/genética , Calicreínas de Tejido/genética , Calicreínas de Tejido/metabolismo
3.
Blood ; 125(4): 710-9, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339356

RESUMEN

The precise mechanism for reduced thrombosis in prekallikrein null mice (Klkb1(-/-)) is unknown. Klkb1(-/-) mice have delayed carotid artery occlusion times on the rose bengal and ferric chloride thrombosis models. Klkb1(-/-) plasmas have long-activated partial thromboplastin times and defective contact activation-induced thrombin generation that partially corrects upon prolonged incubation. However, in contact activation-induced pulmonary thromboembolism by collagen/epinephrine or long-chain polyphosphate, Klkb1(-/-) mice, unlike F12(-/-) mice, do not have survival advantage. Klkb1(-/-) mice have reduced plasma BK levels and renal B2R mRNA. They also have increased expression of the renal receptor Mas and plasma prostacyclin. Increased prostacyclin is associated with elevated aortic vasculoprotective transcription factors Sirt1 and KLF4. Treatment of Klkb1(-/-) mice with the Mas antagonist A-779, COX-2 inhibitor nimesulide, or Sirt1 inhibitor splitomicin lowers plasma prostacyclin and normalizes arterial thrombosis times. Treatment of normal mice with the Mas agonist AVE0991 reduces thrombosis. Klkb1(-/-) mice have reduced aortic tissue factor (TF) mRNA, antigen, and activity. In sum, Klkb1(-/-) mice have a novel mechanism for thrombosis protection in addition to reduced contact activation. This pathway arises when bradykinin delivery to vasculature is compromised and mediated by increased receptor Mas, prostacyclin, Sirt1, and KLF4, leading to reduced vascular TF.


Asunto(s)
Trombosis de las Arterias Carótidas , Epoprostenol , Factores de Transcripción de Tipo Kruppel , Precalicreína , Proteínas Proto-Oncogénicas , Receptores Acoplados a Proteínas G , Tromboplastina , Angiotensina II/análogos & derivados , Angiotensina II/farmacología , Animales , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/genética , Trombosis de las Arterias Carótidas/metabolismo , Trombosis de las Arterias Carótidas/patología , Epoprostenol/biosíntesis , Epoprostenol/genética , Imidazoles/farmacología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Naftalenos/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Tiempo de Tromboplastina Parcial , Fragmentos de Péptidos/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pironas/farmacología , ARN Mensajero , Receptor de Bradiquinina B2/biosíntesis , Receptor de Bradiquinina B2/genética , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/biosíntesis , Sirtuina 1/genética , Sulfonamidas/farmacología , Sinaptotagminas/biosíntesis , Sinaptotagminas/genética , Tromboplastina/antagonistas & inhibidores , Tromboplastina/biosíntesis , Tromboplastina/genética
4.
Nature ; 467(7318): 972-6, 2010 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-20927103

RESUMEN

Reciprocity of inflammation, oxidative stress and neovascularization is emerging as an important mechanism underlying numerous processes from tissue healing and remodelling to cancer progression. Whereas the mechanism of hypoxia-driven angiogenesis is well understood, the link between inflammation-induced oxidation and de novo blood vessel growth remains obscure. Here we show that the end products of lipid oxidation, ω-(2-carboxyethyl)pyrrole (CEP) and other related pyrroles, are generated during inflammation and wound healing and accumulate at high levels in ageing tissues in mice and in highly vascularized tumours in both murine and human melanoma. The molecular patterns of carboxyalkylpyrroles are recognized by Toll-like receptor 2 (TLR2), but not TLR4 or scavenger receptors on endothelial cells, leading to an angiogenic response that is independent of vascular endothelial growth factor. CEP promoted angiogenesis in hindlimb ischaemia and wound healing models through MyD88-dependent TLR2 signalling. Neutralization of endogenous carboxyalkylpyrroles impaired wound healing and tissue revascularization and diminished tumour angiogenesis. Both TLR2 and MyD88 are required for CEP-induced stimulation of Rac1 and endothelial migration. Taken together, these findings establish a new function of TLR2 as a sensor of oxidation-associated molecular patterns, providing a key link connecting inflammation, oxidative stress, innate immunity and angiogenesis.


Asunto(s)
Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , Estrés Oxidativo/fisiología , Pirroles/metabolismo , Receptor Toll-Like 2/metabolismo , Envejecimiento/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Línea Celular , Movimiento Celular , Células Endoteliales/metabolismo , Miembro Posterior/metabolismo , Humanos , Inmunidad Innata/inmunología , Inflamación/metabolismo , Isquemia/metabolismo , Ligandos , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Oxidación-Reducción , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Propionatos , Pirroles/química , Pirroles/farmacología , Receptores Depuradores/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Proteína de Unión al GTP rac1/metabolismo
5.
Blood ; 122(8): 1522-31, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23744584

RESUMEN

Prolylcarboxypeptidase (PRCP) is associated with leanness, hypertension, and thrombosis. PRCP-depleted mice have injured vessels with reduced Kruppel-like factor (KLF)2, KLF4, endothelial nitric oxide synthase (eNOS), and thrombomodulin. Does PRCP influence vessel growth, angiogenesis, and injury repair? PRCP depletion reduced endothelial cell growth, whereas transfection of hPRCP cDNA enhanced cell proliferation. Transfection of hPRCP cDNA, or an active site mutant (hPRCPmut) rescued reduced cell growth after PRCP siRNA knockdown. PRCP-depleted cells migrated less on scratch assay and murine PRCP(gt/gt) aortic segments had reduced sprouting. Matrigel plugs in PRCP(gt/gt) mice had reduced hemoglobin content and angiogenic capillaries by platelet endothelial cell adhesion molecule (PECAM) and NG2 immunohistochemistry. Skin wounds on PRCP(gt/gt) mice had delayed closure and reepithelialization with reduced PECAM staining, but increased macrophage infiltration. After limb ischemia, PRCP(gt/gt) mice also had reduced reperfusion of the femoral artery and angiogenesis. On femoral artery wire injury, PRCP(gt/gt) mice had increased neointimal formation, CD45 staining, and Ki-67 expression. Alternatively, combined PRCP(gt/gt) and MRP-14(-/-) mice were protected from wire injury with less neointimal thickening, leukocyte infiltration, and cellular proliferation. PRCP regulates cell growth, angiogenesis, and the response to vascular injury. Combined with its known roles in blood pressure and thrombosis control, PRCP is positioned as a key regulator of vascular homeostasis.


Asunto(s)
Carboxipeptidasas/fisiología , Células Endoteliales/enzimología , Neovascularización Patológica , Neovascularización Fisiológica , Animales , Aorta/metabolismo , Apoptosis , Calgranulina B/metabolismo , Bovinos , Movimiento Celular , Proliferación Celular , Células Cultivadas , Arteria Femoral/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isquemia/patología , Antígeno Ki-67/metabolismo , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Cicatrización de Heridas
6.
Blood ; 119(9): 2149-58, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22134168

RESUMEN

Vascular development and angiogenesis initially depend on endothelial tip cell invasion, which is followed by a series of maturation steps, including lumen formation and recruitment of perivascular cells. Notch ligands expressed on the endothelium and their cognate receptors expressed on perivascular cells are involved in blood vessel maturation, though little is known regarding the Notch-dependent effectors that facilitate perivascular coverage of nascent vessels. Here, we report that vascular smooth muscle cell (VSMC) recognition of the Notch ligand Jagged1 on endothelial cells leads to expression of integrin αvß3 on VSMCs. Once expressed, integrin αvß3 facilitates VSMC adhesion to VWF in the endothelial basement membrane of developing retinal arteries, leading to vessel maturation. Genetic or pharmacologic disruption of Jagged1, Notch, αvß3, or VWF suppresses VSMC coverage of nascent vessels and arterial maturation during vascular development. Therefore, we define a Notch-mediated interaction between the developing endothelium and VSMCs leading to adhesion of VSMCs to the endothelial basement membrane and arterial maturation.


Asunto(s)
Membrana Basal/metabolismo , Adhesión Celular/fisiología , Endotelio Vascular/metabolismo , Integrinas/metabolismo , Músculo Liso Vascular/metabolismo , Receptores Notch/metabolismo , Animales , Arterias/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Humanos , Integrinas/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica/genética , Unión Proteica , ARN Mensajero/metabolismo , Receptores Notch/genética , Proteínas Serrate-Jagged , Transducción de Señal/fisiología , Factor de von Willebrand/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 305(3): H305-20, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23709605

RESUMEN

How single-chain urokinase (ScuPA) mediates angiogenesis is incompletely understood. ScuPA (≥4 nM) induces phosphorylated (p)ERK1/2 (MAPK44 and MAPK42) and pAkt (Ser(473)) in umbilical vein and dermal microvascular endothelial cells. Activation of pERK1/2 by ScuPA is blocked by PD-98059 or U-0126, and pAkt (Ser(473)) activation is inhibited by wortmannin or LY-294002. ScuPA (32 nM) or protease-inhibited two-chain urokinase stimulates pERK1/2 to the same extent, indicating that signaling is not dependent on enzymatic activity. ScuPA induces pERK1/2, but not pAkt (Ser(473)), in SIN1(-/-) cells, indicating that the two pathways are not identical. Peptides from domain 2 of the urokinase plasminogen activator receptor (uPAR) or domain 5 of high-molecular-weight kininogen compete with ScuPA for the induction of pERK1/2 and pAkt (Ser(473)). A peptide of the integrin-binding site on uPAR, a ß1-integrin peptide that binds uPAR, antibody 6S6 to ß1-integrin, tyrosine kinase inhibitors AG-1478 or PP3, and small interfering RNA knockdown of VEFG receptor 2, but not HER1-HER4, blocked ScuPA-induced pERK1/2 and pAkt (Ser(473)). ScuPA-induced endothelial cell proliferation was blocked by inhibitors of pERK1/2 and pAkt (Ser(473)), antibody 6S6, and uPAR or kininogen peptides. ScuPA initiated aortic sprouts and Matrigel plug angiogenesis in normal, but not uPAR-deficient, mouse aortae or mice, respectively, but these were blocked by PD-98059, LY-294002, AG-1478, or cleaved high-molecular-weight kininogen. In summary, this investigation indicates a novel, a nonproteolytic signaling pathway initiated by zymogen ScuPA and mediated by domain 2 of uPAR, ß1-integrins, and VEGF receptor 2 leading to angiogenesis. Kininogens or peptides from it downregulate this pathway.


Asunto(s)
Células Endoteliales/enzimología , Integrina beta1/metabolismo , Neovascularización Fisiológica , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proliferación Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Quininógeno de Alto Peso Molecular/metabolismo , Ratones , Ratones Noqueados , Modelos Moleculares , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Receptores del Activador de Plasminógeno Tipo Uroquinasa/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Transducción de Señal , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transfección , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
8.
Blood Adv ; 7(8): 1446-1459, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36006440

RESUMEN

Elevated neutrophil-to-lymphocyte ratio (NLR) in patients who undergo elective vascular surgery (EVS) have increased mortality independent of perioperative surgical outcome. To understand why high NLR is associated with higher mortality, we investigated neutrophil and lymphocyte transcriptome expression in patients undergoing EVS. Blood samples were collected from patients undergoing EVS and healthy donors for NLR calculation. RNA samples were isolated from patients' neutrophils and lymphocytes and divided into NLR_Low (<3) and NLR_High (≥3) groups (n = 6 each). Paired samples with the highest RNA integrity number (mean = 9.8 ± 0.4) were sequenced and analyzed for differential expression. Normalized data were inputted for downstream analysis using iPathwayGuide (AdvaitaBio) and gene set enrichment analysis using GenePattern and MSigDB (Broad Institute). There was no clinical difference between the patient groups with regard to clinical diagnosis, age, sex, history of hypertension, lipid abnormalities, diabetes mellitus, smoking, or statin use. The mean NLR was 4.37 ± 0.27 SEM in the NLR_High and 1.88 ± 0.16 for the NLR_Low groups. Significantly differentially expressed gene sets identified in the RNA sequence data were enriched highly (P = 1E-24) in the humoral immunity and complement systems. Neutrophils from NLR_High patients downregulated complement genes (C1QA, C1QB, C1QC, C1S, C2, CR2, C3AR1, C3, C8G, and C9 and complement regulatory genes CD59, SERPING1, C4BPA, CFH, and CFI). Downregulation of gene expressions of humoral immunity and complement within the neutrophils are associated with elevated NLR. It remains to be determined whether and how these changes contribute to increased late mortality previously observed in patients undergoing EVS.


Asunto(s)
Linfocitos , Neutrófilos , Humanos , Procedimientos Quirúrgicos Vasculares , Procedimientos Quirúrgicos Electivos
9.
bioRxiv ; 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37662286

RESUMEN

Background: Antibodies to ß2-glycoprotein I (ß2GPI) cause thrombosis in antiphospholipid syndrome, however the role of ß2GPI itself in regulation of coagulation pathways in vivo is not well understood. Methods: We developed ß2GPI-deficient mice (Apoh -/- ) by deleting exon 2 and 3 of Apoh using CRISPR/Cas9 and compared the propensity of wild-type (WT) and Apoh -/- mice to develop thrombosis using rose bengal and FeCl 3 -induced carotid thrombosis, laser-induced cremaster arteriolar injury, and inferior vena cava (IVC) stasis models. We also compared tail bleeding times and assessed platelet activation in WT and Apoh -/- mice in the absence and presence of exogenous ß2GPI. Results: Compared to WT littermates, Apoh -/- mice demonstrated a prolonged time to occlusion of the carotid artery after exposure to rose bengal or FeCl 3 , and reduced platelet and fibrin accumulation in cremasteric arterioles after laser injury. Similarly, significantly smaller thrombi were retrieved from the IVC of Apoh -/- mice 48 hours after IVC occlusion. The activated partial thromboplastin time (aPTT) and prothrombin time, as well as aPTT reagent- and tissue factor-induced thrombin generation times using plasma from Apoh -/- and WT mice revealed no differences. However, we observed significant prolongation of tail bleeding in Apoh -/- mice, and reduced P-selectin expression and binding of fibrinogen to the activated α2bß3 integrin on platelets from these mice after stimulation with low thrombin concentrations; these changes were reversed by exogenous ß2GPI. An antibody to PAR3 blocked thrombin-induced activation of WT, but not Apoh -/- platelets, as well as the ability of ß2GPI to restore the activation response of Apoh -/- platelets to thrombin. ß2GPI deficiency did not affect platelet activation by a PAR4-activator peptide, or ADP. Conclusions: In mice, ß2GPI may mediate procoagulant activity by enhancing the ability of PAR3 to present thrombin to PAR4, promoting platelet activation at low thrombin concentrations. Key Points: ß2GPI deficient mice are protected from experimental arterial, venous, and microvascular thrombosis.ß2GPI deficient mice display prolonged tail bleeding times and reduced PAR3-facilitated platelet activation by low concentrations of thrombin.

10.
Blood Adv ; 5(22): 4741-4751, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34597365

RESUMEN

Extracellular vesicles (EV) have been implicated in diverse biological processes, including intracellular communication, transport of nucleic acids, and regulation of vascular function. Levels of EVs are elevated in cancer, and studies suggest that EV may stimulate thrombosis in patients with cancer through expression of tissue factor. However, limited data also implicate EV in the activation of the contact pathway of coagulation through activation of factor XII (FXII) to FXIIa. To better define the ability of EV to initiate contact activation, we compared the ability of EV derived from different cancer cell lines to activate FXII. EV from all cell lines activated FXII, with those derived from pancreatic and lung cancer cell lines demonstrating the most potent activity. Concordant with the activation of FXII, EV induced the cleavage of high molecular weight kininogen (HK) to cleaved kininogen. We also observed that EVs from patients with cancer stimulated FXII activation and HK cleavage. To define the mechanisms of FXII activation by EV, EV were treated with calf intestinal alkaline phosphatase or Escherichia coli exopolyphosphatase to degrade polyphosphate; this treatment blocked binding of FXII to EVs and the ability of EV to mediate FXII activation. In vivo, EV induced pulmonary thrombosis in wild-type mice, with protection conferred by a deficiency in FXII, HK, or prekallikrein. Moreover, pretreatment of EVs with calf intestinal alkaline phosphatase inhibited their prothrombotic effect. These results indicate that polyphosphate mediates the binding of contact factors to EV and that EV-associated polyphosphate may contribute to the prothrombotic effects of EV in cancer.


Asunto(s)
Vesículas Extracelulares , Neoplasias , Animales , Factor XII , Factor XIIa , Humanos , Ratones , Polifosfatos , Precalicreína
11.
Front Med (Lausanne) ; 7: 591546, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330551

RESUMEN

A previously hemostatically asymptomatic patient with common variable hypogammaglobulinemia was given everolimus to prevent growth of her liver. Within several months, the patient developed a severe bleeding disorder. The bleeding was due to fibrin polymerization defect that upon sequencing was shown to be dysfibrinogenemia Krakow III. Elimination of the mTor inhibitor ameliorated the clinical bleeding state.

12.
Cancer Epidemiol Biomarkers Prev ; 17(2): 339-42, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18268117

RESUMEN

Three recent studies identified common variants on 8q24 that confer modestly increased susceptibility to colorectal cancer. Here, we replicate the association in a population-based case-control study of colon cancer, including 561 cases and 721 unrelated controls. The rs6983267 marker was significantly associated with colon cancer risk. Compared with those homozygous for the T allele, the heterozygous and homozygous carriers for the G allele had an age-adjusted odds ratio of 1.39 (95% confidence interval, 1.03-1.88) and 1.68 (95% confidence interval, 1.21-2.33), respectively. An additive model showed strong evidence for a gene-dose response relationship (P(trend) = 0.0022). The association remained statistically significant when restricted to Caucasians only (527 cases and 679 controls; P(trend) = 0.0056). Further adjustment for other known risk factors did not alter the results. Stratified analysis revealed no evidence for effect modification by family history of colorectal cancer, age, or gender. These data replicate the association identified from recent studies, providing additional evidence supporting the rs6983267 genetic polymorphism as a marker predisposing to colon cancer.


Asunto(s)
Cromosomas Humanos Par 8/genética , Neoplasias del Colon/genética , Alelos , Estudios de Casos y Controles , Neoplasias del Colon/epidemiología , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Incidencia , Kentucky/epidemiología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Programa de VERF
13.
J Clin Invest ; 128(3): 944-959, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29376892

RESUMEN

Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMß2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.


Asunto(s)
Factor XII/metabolismo , Neutrófilos/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Cicatrización de Heridas , Animales , Calcio/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Trampas Extracelulares , Femenino , Humanos , Inflamación , Leucocitos/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal
14.
Front Med (Lausanne) ; 3: 17, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27200353

RESUMEN

Plasma kallikrein formed from prekallikrein (PK) produces bradykinin from kininogens and activates factor XII. Plasma PK is activated by factors αXIIa, ßXIIa, or prolylcarboxypeptidase (PRCP). A cross-sectional investigation determined if there is an association of PRCP and KLKB1 polymorphisms with cardiovascular disease (CVD). DNA was obtained from 2243 individuals from the Prevention of Events with Angiotensin Converting Enzyme trial. Two PRCP SNPs, rs7104980 and rs2298668, and two KLKB1 SNPs, rs3733402 and rs3087505, were genotyped. Logistic regression models were performed for history of diabetes, myocardial infarction, stroke, angina, angiographic coronary disease, CABG, intermittent claudication, percutaneous transluminal coronary angioplasty (PTCA), and transient ischemic attack. The PRCP SNP rs7104980 increased the odds of having a history of PTCA by 21% [odds ratio (OR) = 1.211; 95% confidence intervals (CI) = (1.008, 1.454)]; P = 0.041, but was non-significant after Bonferroni correction. Alternatively, having the G allele for rs3733402 (KLKB1 gene) decreased the odds of having a history of angiographic coronary disease by 24% [OR = 0.759; 95% CI = (0.622, 0.927)]; P = 0.007 that was statistically significant (P < 0.01) after Bonferroni correction for multiple hypothesis testing. When the best-fit model based on the Akaike information criterion controlled for age, weight, gender, hypertension, and history of angina, the G allele of KLKB1 rs3733402 that is associated with less plasma kallikrein activity correlated with reduced history of CVD.

17.
World J Gastroenterol ; 15(18): 2240-4, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19437564

RESUMEN

AIM: To investigate the association of variations in the cyclooxygenase-2 (COX2) and uridine diphosphate glucuronosyltransferase 1A6 (UGT1A6) genes and non-steroidal anti-inflammatory drugs (NSAIDs) use with risk of colon cancer. METHODS: NSAIDs, which are known to reduce the risk of colon cancer, act directly on COX2 and reduce its activity. Epidemiological studies have associated variations in the COX2 gene with colon cancer risk, but others were unable to replicate this finding. Similarly, enzymes in the UGT1A6 gene have been demonstrated to modify the therapeutic effect of NSAIDs on colon adenomas. Polymorphisms in the UGT1A6 gene have been statistically shown to interact with NSAID intake to influence risk of developing colon adenomas, but not colon cancer. Here we examined the association of tagging single nucleotide polymorphisms (SNPs) in the COX2 and UGT1A6 genes, and their interaction with NSAID consumption, on risk of colon cancer in a population of 422 colon cancer cases and 481 population controls. RESULTS: No SNP in either gene was individually statistically significantly associated with colon cancer, nor did they statistically significantly change the protective effect of NSAID consumption in our sample. Like others, we were unable to replicate the association of variants in the COX2 gene with colon cancer risk (P > 0.05), and we did not observe that these variants modify the protective effect of NSAIDs (P > 0.05). We were able to confirm the lack of association of variants in UGT1A6 with colon cancer risk, although further studies will have to be conducted to confirm the association of these variants with colon adenomas. CONCLUSION: Our study does not support a role of COX2 and UGT1A6 genetic variations in the development of colon cancer.


Asunto(s)
Neoplasias del Colon/genética , Ciclooxigenasa 2/genética , Predisposición Genética a la Enfermedad , Glucuronosiltransferasa/genética , Polimorfismo de Nucleótido Simple , Anciano , Antiinflamatorios no Esteroideos/efectos adversos , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo
18.
World J Gastroenterol ; 15(30): 3771-5, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19673018

RESUMEN

AIM: To investigate the association between single nucleotide polymorphisms (SNPs) in the phosphatase and tensin homolog (PTEN) tumor suppressor gene and risk of colon cancer. METHODS: We utilized a population-based case-control study of incident colon cancer individuals (n = 421) and controls (n = 483) aged > or = 30 years to conduct a comprehensive tagSNP association analysis of the PTEN gene. RESULTS: None of the PTEN SNPs were statistically significantly associated with colon cancer when controlled for age, gender, and race, or when additionally adjusted for other known risk factors (P > 0.05). Haplotype analyses similarly showed no association between the PTEN gene and colon cancer. CONCLUSION: Our study does not support PTEN as a colon cancer susceptibility gene.


Asunto(s)
Neoplasias del Colon/genética , Predisposición Genética a la Enfermedad , Fosfohidrolasa PTEN/genética , Polimorfismo Genético , Humanos , Fosfohidrolasa PTEN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA