Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Genet ; 6(2): 205-9, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8162076

RESUMEN

Hereditary haemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder that is characterized by frequent nosebleeds, mucocutaneous telangiectases and vascular malformations that cause recurrent haemorrhage and arteriovenous shunting. Linkage analyses in one kindred identified an HHT locus on the long arm of chromosome 9 (maximum multipoint lod score = 6.20 between D9S60 and D9S61). Analyses in two other unrelated HHT families demonstrated that the disease in one was not linked to the locus on chromosome 9q3. We conclude that HHT is a genetically heterogeneous disorder. Based on its map location (9q3) and expression in vascular tissues, type V collagen is a possible candidate gene for HHT.


Asunto(s)
Cromosomas Humanos Par 9 , Ligamiento Genético , Telangiectasia Hemorrágica Hereditaria/genética , Adulto , Niño , Mapeo Cromosómico , Europa (Continente) , Femenino , Marcadores Genéticos , Haplotipos , Humanos , Masculino , Linaje , Polimorfismo Genético , Secuencias Repetitivas de Ácidos Nucleicos
2.
Nat Genet ; 34(2): 220-5, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12717434

RESUMEN

Mutations in LMAN1 (also called ERGIC-53) result in combined deficiency of factor V and factor VIII (F5F8D), an autosomal recessive bleeding disorder characterized by coordinate reduction of both clotting proteins. LMAN1 is a mannose-binding type 1 transmembrane protein localized to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC; refs. 2,3), suggesting that F5F8D could result from a defect in secretion of factor V and factor VIII (ref. 4). Correctly folded proteins destined for secretion are packaged in the ER into COPII-coated vesicles, which subsequently fuse to form the ERGIC. Secretion of certain abundant proteins suggests a default pathway requiring no export signals (bulk flow; refs. 6,7). An alternative mechanism involves selective packaging of secreted proteins with the help of specific cargo receptors. The latter model would be consistent with mutations in LMAN1 causing a selective block to export of factor V and factor VIII. But approximately 30% of individuals with F5F8D have normal levels of LMAN1, suggesting that mutations in another gene may also be associated with F5F8D. Here we show that inactivating mutations in MCFD2 cause F5F8D with a phenotype indistinguishable from that caused by mutations in LMAN1. MCFD2 is localized to the ERGIC through a direct, calcium-dependent interaction with LMAN1. These findings suggest that the MCFD2-LMAN1 complex forms a specific cargo receptor for the ER-to-Golgi transport of selected proteins.


Asunto(s)
Proteínas Portadoras/genética , Deficiencia del Factor V/genética , Hemofilia A/genética , Hemorragia/genética , Lectinas de Unión a Manosa/genética , Proteínas de la Membrana/genética , Secuencia de Aminoácidos , Transporte Biológico Activo/genética , Retículo Endoplásmico/metabolismo , Deficiencia del Factor V/metabolismo , Femenino , Aparato de Golgi/metabolismo , Células HeLa , Hemofilia A/metabolismo , Hemorragia/etiología , Hemorragia/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Homología de Secuencia de Aminoácido , Transfección , Proteínas de Transporte Vesicular
3.
Haemophilia ; 16(4): 584-91, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20070385

RESUMEN

Bernard Soulier syndrome (BSS) is a rare disorder of platelets, inherited mainly as an autosomal recessive trait. It is characterised by qualitative and quantitative defects of the platelet membrane glycoprotein (GP) Ib-IX-V complex. The main clinical characteristics are thrombocytopenia, prolonged bleeding time and the presence of giant platelets. Data on the clinical course and outcome of pregnancy in women with Bernard Soulier syndrome is scattered in individual case reports. In this paper, we performed a systematic review of literature and identified 16 relevant articles; all case reports that included 30 pregnancies among 18 women. Primary postpartum haemorrhage was reported in 10 (33%) and secondary in 12 (40%) of pregnancies, requiring blood transfusion in 15 pregnancies. Two women had an emergency obstetric hysterectomy. Alloimmune thrombocytopenia was reported in 6 neonates, with one intrauterine death and one neonatal death. Bernard Soulier syndrome in pregnancy is associated with a high risk of serious bleeding for the mother and the neonate. A multidisciplinary team approach and individualised management plan for such women are required to minimise these risks. An international registry is recommended to obtain further knowledge in managing women with this rare disorder.


Asunto(s)
Síndrome de Bernard-Soulier/complicaciones , Complicaciones del Embarazo , Adulto , Transfusión Sanguínea/estadística & datos numéricos , Femenino , Humanos , Histerectomía/estadística & datos numéricos , Recién Nacido , Recuento de Plaquetas , Hemorragia Posparto/epidemiología , Embarazo , Resultado del Embarazo , Trombocitopenia Neonatal Aloinmune/epidemiología , Adulto Joven
4.
Haemophilia ; 15(2): 501-8, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19187194

RESUMEN

Patients with haemophilia complicated by inhibitors have a significant burden of joint disease, which is associated with a negative impact on their quality of life. Successful elective orthopaedic surgery can result in decreased bleed frequency into a new joint, less time spent in hospital, increased mobility and improved well being. This paper describes a new protocol for use of recombinant activated factor VII (rFVIIa) in elective orthopaedic surgery, based on a review of published data as well as the personal experience of a group of expert physicians. The protocol offers guidance on the planning of the surgery and preoperative testing as well as the bolus schedule for rFVIIa and advice on the concomitant use of antifibrinolytic agents and fibrin sealants. A total of 10 operations involving 13 procedures in eight patients in five comprehensive care centres have been undertaken until now using the protocol, which employs an initial bolus dose of rFVIIa in the range of 120-180 microg kg(-1) to cover surgery. The clinical experience reported here encompasses all cases of elective orthopaedic surgery using rFVIIa as initial treatment carried out in the UK and Republic of Ireland over the last 2 years. In all cases, there was good control of haemostasis during surgery and the final outcome was rated as 'excellent' or 'extremely satisfactory' by the reporting clinicians. Although the initial cost of product to cover surgery such as arthroplasty is high, it needs to be borne in mind that this may be offset in subsequent years by savings resulting from avoidance of bleeding episodes in the affected joint.


Asunto(s)
Conferencias de Consenso como Asunto , Factor VIIa/uso terapéutico , Hemofilia A/tratamiento farmacológico , Artropatías/cirugía , Hemorragia Posoperatoria/prevención & control , Proteínas Recombinantes/uso terapéutico , Adolescente , Adulto , Anciano , Pérdida de Sangre Quirúrgica/prevención & control , Niño , Preescolar , Protocolos Clínicos , Procedimientos Quirúrgicos Electivos , Hemofilia A/complicaciones , Humanos , Persona de Mediana Edad , Procedimientos Ortopédicos/efectos adversos , Resultado del Tratamiento , Adulto Joven
6.
J Thromb Haemost ; 4(10): 2191-8, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16803463

RESUMEN

BACKGROUND: Coagulation proteins promote neointimal hyperplasia and vascular remodelling after vessel injury, but the precise mechanisms by which they act in vivo remain undetermined. OBJECTIVES: This study, using an injury model in which the neointima is derived from bone marrow (BM)-derived cells, compared inhibition of tissue factor or thrombin on either BM-derived or existing vascular smooth muscle cells. METHODS: Two transgenic (Tg) mouse strains expressing membrane-tethered tissue factor pathway inhibitor (TFPI) or hirudin (Hir) fusion proteins driven by an alpha smooth muscle actin (SMA) promoter were generated (alpha-TFPI-Tg and alpha-Hir-Tg) and the phenotype after wire-induced endovascular injury was compared with that in wild-type (WT) controls. RESULTS: WT mice developed progressive neointimal expansion, whereas injury in either Tg was followed by repair back to a preinjured state. This was also seen when WT mice were reconstituted with BM from Tg mice but not when Tgs were reconstituted with WT BM, in which injury was followed by slowly progressive neointimal expansion. Injection of CD34+ cells from Tg mice into injured WT mice resulted in the accumulation of fusion protein-expressing cells from day 3 onwards and an absence of neointimal hyperplasia in those areas. CONCLUSIONS: Neointimal development after wire-induced endovascular injury in mice was completely inhibited when BM-derived cells infiltrating the damaged artery expressed membrane tethered anticoagulant fusion proteins under an alpha-SMA promoter. These findings enhance our understanding of the pathological role that coagulation proteins play in vascular inflammation.


Asunto(s)
Anticoagulantes/metabolismo , Antígenos CD34/biosíntesis , Células de la Médula Ósea/metabolismo , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Proteínas Recombinantes de Fusión/metabolismo , Células Madre/metabolismo , Animales , Aorta/metabolismo , Arteriosclerosis/terapia , Vasos Sanguíneos/patología , Arterias Carótidas/patología , Humanos , Inflamación , Ratones , Ratones Transgénicos , Músculo Liso/metabolismo , Fenotipo
7.
Cancer Res ; 58(19): 4461-7, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9766679

RESUMEN

Overexpression of tissue factor (TF) is characteristically observed in advanced pancreatic cancer and has been associated with invasion and metastasis. Functional responses of TF activation are here investigated using as a model system the human pancreatic cancer cell lines SW979 (which overexpresses TF) and MIAPaCa2 (which does not express detectable levels). After stimulation of these cell lines with factor VIIa (FVIIa), the only known TF ligand, expression of urokinase receptor (uPAR) gene was up-regulated in SW979 cells in a dose-dependent manner but not in MIAPaCa2 cells. Interestingly, urokinase (uPA) and its specific inhibitor PAI-1 were not up-regulated. Exposure to functionally inactivated FVIIa did not show any effect on uPAR expression on SW979 cells despite binding to TF with higher efficiency. The neutralizing anti-TF antibody 5G9 blocked the FVIIa-induced up-regulation of uPAR completely, whereas hirudin failed to block this up-regulation. Treatment of SW979 cells with Factor Xa did not up-regulate the expression of uPAR gene, whereas treatment with FVII induced the same level of enhanced uPAR gene expression as that with FVIIa. In the matrigel invasion assay, enhanced invasion of SW979 cell line induced by FVIIa was completely inhibited by anti-TF antibody and alpha2-antiplasmin. Moreover, the endogenous levels of uPAR gene expression were significantly correlated with the level of TF gene expression in 19 human cancer cell lines (P < 0.05). These data suggest that up-regulation of uPAR expression by tumor cells leading to tumor invasion is induced through the TF-FVIIa pathway rather than TF-initiated thrombin generation. This is the first report that TF may be one of the key receptors that can up-regulate expression of the plasminogen activator receptor in human cancer cells to enhance tumor invasion and metastasis.


Asunto(s)
Factor VIIa/fisiología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Receptores de Superficie Celular/genética , Tromboplastina/metabolismo , Neoplasias de la Mama , Movimiento Celular , Factor VIIa/farmacología , Factor Xa/farmacología , Factor Xa/fisiología , Femenino , Citometría de Flujo , Humanos , Neoplasias Pancreáticas/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Tromboplastina/genética , Transcripción Genética , Células Tumorales Cultivadas , Regulación hacia Arriba , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
8.
J Mol Biol ; 234(4): 1263-5, 1993 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-8263927

RESUMEN

The extracellular domain (residues 1 to 220) of human tissue factor has been cloned and expressed in Escherichia coli and purified to isoelectric homogeneity. Single crystals suitable for X-ray analysis have been obtained by vapour diffusion. They belong to the tetragonal space group P4(1)2(1)2 or P4(3)2(1)2 with a = b = 45.2 A, c = 231.5 A, contain one molecule per asymmetric unit and diffract to 2.6 A resolution. Native and derivative data sets have been collected to 3.6 and 3.9 A, respectively.


Asunto(s)
Tromboplastina/ultraestructura , Cristalografía por Rayos X , Espacio Extracelular/química , Humanos , Proteínas Recombinantes
9.
Hum Mutat ; 17(1): 3-17, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11139238

RESUMEN

Factor VII (FVII) is a zymogen for a vitamin K-dependent serine protease essential for the initiation of blood coagulation. It is synthesized primarily in the liver and circulates in plasma at a concentration of approximately 0.5 microg/ml (10 nmol/L). The FVII gene (F7) is located on chromosome 13 (13q34), consists of 9 exons, and spans approximately 12kb. It encodes a mature protein of 406 amino acids, which has an N-terminal domain (Gla) post-translationally modified by gamma-carboxylation of glutamic acid residues, two domains with homology to epidermal growth factor (EGF1 and 2), and a C-terminal serine protease domain. The single chain zymogen is activated by proteolytic cleavage at Arg152-Ile153. There are 238 individuals described in the world literature with mutations in their F7 genes (FVII mutation database; europium.csc. mrc.ac.uk). Complete absence of FVII activity in plasma is usually incompatible with life, and individuals die shortly after birth due to severe hemorrhage. The majority of individuals with mutations in their F7 gene(s), however, are either asymptomatic or the clinical phenotype is unknown. In general, a severe bleeding phenotype is only observed in individuals homozygous for a mutation in their F7 genes with FVII activities (FVII:C) below 2% of normal, however, a considerable proportion of individuals with a mild-moderate bleeding phenotype have similar FVII:C by in vitro assay. The failure of in vitro tests to differentiate between these groups may be due to lack of sensitivity in the assays to the very low amounts of FVII:C, which are sufficient to initiate coagulation in vivo. A number of polymorphisms have been identified in the F7 gene and some have been shown to influence plasma FVII antigen levels.


Asunto(s)
Bases de Datos Factuales , Deficiencia del Factor VII/genética , Factor VII/genética , Mutación/genética , Animales , Genes Letales/genética , Humanos
10.
Blood Rev ; 3(4): 251-62, 1989 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-2514817

RESUMEN

von Willebrand factor (vWF) is a plasma protein with multiple functions in haemostasis. The vWF gene, located on chromosome 12p2.1, encodes a primary gene product of 2813 amino acids. Post-translational modification, assembly and secretion of vWF are highly complex. The pro vWF promoter is covalently linked by intermolecular disulphide bonds to form a dimer of MW approximately 440 kDa. This then polymerises to form multimers ranging in MW from 1-20 x 10(6). Simultaneously the pro piece of vWF is cleaved, releasing a 741 amino acid peptide known as vW Ag II from the polymerised protomers. Two distinct secretion pathways are found in the endothelial cell, a regulated pathway with storage in Weibl-Palade bodies and a constitutive pathway. Platelets store vWF in their alpha-granules. Mature vWF participates in platelet adhesion, spreading and aggregation and is a carrier of factor VIII, protecting the latter from degradation. Disorders of vWF are highly diverse. At least 20 subtypes of von Willebrand's disease have been described to date, based on features of the vWF present in or absent from patients plasma and platelets. Some patients have reduced amounts of apparently normal vWF whilst others have clearly abnormal vWF with aberrant structure and function. Rare patients virtually or completely lack vWF. The genetic and structural basis of some of these abnormalities is just beginning to emerge. This article outlines the molecular biology and physiology of vWF, and reviews some recent progress on the molecular pathology and genetics of von Willebrand's disease.


Asunto(s)
Factor de von Willebrand/fisiología , Animales , Cromosomas Humanos Par 12 , ADN/genética , Desamino Arginina Vasopresina/uso terapéutico , Factor VIII/metabolismo , Regulación de la Expresión Génica , Genes , Hemostasis , Humanos , Adhesividad Plaquetaria , Procesamiento Proteico-Postraduccional , Porcinos , Enfermedades de los Porcinos/genética , Enfermedades de von Willebrand/clasificación , Enfermedades de von Willebrand/tratamiento farmacológico , Enfermedades de von Willebrand/genética , Enfermedades de von Willebrand/veterinaria , Factor de von Willebrand/biosíntesis , Factor de von Willebrand/genética
11.
Gene ; 139(2): 275-9, 1994 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-8112618

RESUMEN

Recombinant human proteins are generally recovered in low yields from mammalian tissue culture following transfection with commercially available vectors. We have constructed a novel vector containing both the neomycin-resistance-encoding gene (neo) as a dominant selectable marker, and the dihydrofolate reductase-encoding gene (DHFR) to enable amplification of transfected DNA followed by stable expression in mammalian cell lines. Levels of 5 micrograms/ml of the coagulation proteins, factor VII (FVII) and factor XI (FXI), have been achieved in serum-free media. N-terminal sequencing of the purified proteins, and of their separated chains after proteolytic activation, demonstrated correct processing of the recombinant products. In addition, the ratios of clotting activity to antigen for each are close to unity, and the recombinant and plasma-derived proteins had identical mobilities upon electrophoresis in the presence of SDS. The vector described will be of use for the synthesis of recombinant proteins, both wild-type and variants produced by site-directed mutagenesis, especially where complex post-translational modification of the protein makes it essential to use mammalian cells.


Asunto(s)
Factor VII/biosíntesis , Factor XI/biosíntesis , Vectores Genéticos/genética , Animales , Células CHO/efectos de los fármacos , Cricetinae , Medio de Cultivo Libre de Suero , Resistencia a Medicamentos/genética , Factor VII/genética , Factor XI/genética , Humanos , Metotrexato/farmacología , Datos de Secuencia Molecular , Neomicina , Plásmidos/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Tetrahidrofolato Deshidrogenasa/genética , Transcripción Genética/genética , Transfección/genética
12.
FEBS Lett ; 374(1): 141-6, 1995 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-7589502

RESUMEN

The four-domain structure of human factor VIIa and the two-domain structure of tissue factor form a tight complex to initiate blood coagulation. By solution scattering, the mean X-ray and neutron radii of gyration RG (which determine macro-molecular elongation) were found to be 3.25 nm, 2.13 nm and 3.14 nm (+/- 0.13 nm) for factor VIIa, the extracellular region of tissue factor and their complex in that order. The mean cross-sectional radii of gyration RXS were 1.33 nm, 0.56 nm and 1.42 nm (+/- 0.13 nm) in that order. The mean lengths were 10.3 nm, 7.7 nm and 10.2 nm in that order. The data show that, in solution, the free proteins have extended domain structures, and the complex is formed by a compact side-by-side alignment of the two proteins along their long axes. The high binding affinity of tissue factor for factor VIIa may thus be accounted for by the occurrence of many intermolecular contacts in the complex.


Asunto(s)
Factor VIIa/metabolismo , Tromboplastina/metabolismo , Animales , Células CHO , Cricetinae , Factor VIIa/química , Humanos , Neutrones , Conformación Proteica , Dispersión de Radiación , Tromboplastina/química , Rayos X
13.
J Thromb Haemost ; 1(7): 1487-94, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12871284

RESUMEN

In mammalian blood coagulation, five proteases (factor VII [FVII]; factor IX [FIX]; factor X [FX]; protein C [PC] and prothrombin [PT]) act with five cofactors (tissue factor [TF]; factor V [FV]; factor VIII [FVIII]; thrombomodulin and protein S) to control the generation of fibrin. Biochemical evidence, molecular cloning data and comparative sequence analysis support the existence of all components of this network in all jawed vertebrates, and strongly suggest that it evolved before the divergence of teleosts over 430 million years ago. Phylogenetic analysis of the amino acid sequences of the Gla-EGF1-EGF2-SP domain serine proteases (FVII, FIX, FX, PC) and the A domain-containing cofactors (FV and FVIII) strongly supports the evolution of the blood coagulation network through two rounds of gene duplication, and supports the hypothesis that vertebrate evolution benefited from two global genome duplications. The jawless vertebrates (hagfish and lamprey) that diverged over 450 million years ago have a blood coagulation network involving TF, PT and fibrinogen. Preliminary evidence indicates that they may have a smaller complement of Gla-EGF1-EGF2-SP domain proteins, suggesting the existence of a 'primitive' coagulation system in jawless vertebrates.


Asunto(s)
Hemostasis/fisiología , Animales , Evolución Biológica , Factor IX/química , Factor VII/química , Factor X/química , Fibrinógeno/química , Humanos , Modelos Biológicos , Modelos Genéticos , Filogenia , Proteína C/química , Estructura Terciaria de Proteína , Protrombina/química
14.
J Thromb Haemost ; 1(1): 139-46, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12871551

RESUMEN

Inhibitor antibody formation is a complication of factor VIII (FVIII) replacement therapy due to a failure to synthesize sufficient FVIII protein to induce immune tolerance. The incidence of nonsense mutations in inhibitor patients is high, however, this association is variable according to the position of the mutation. We have studied the effect of nonsense mutations on accumulation of FVIII mRNA, protein translation and secretion. Appropriately processed mRNA was detected in cells transfected with wild-type R1966X and R2116X expression constructs and no evidence of nonsense-mediated decay was observed. All constructs directed the translation of detectable intracellular FVIII antigen, however, secreted FVIII was detected only in conditioned media of cells transfected with wild-type cDNA. We have also analyzed ectopic FVIII mRNA transcripts in the lymphocytes of six hemophilia A patients with nonsense mutations (Q139X, R583X, R1941X, R1966X and two unrelated patients with R2116X). FVIII mRNA was detectable in every case. In R1941X and R1966X only normally spliced transcripts were present. In Q139X, R583X and R2116X aberrantly spliced transcripts were observed with two distinct patterns in two individuals with the R2116X mutation. No correlation between mutation, transcript pattern and incidence of inhibitor development was apparent.


Asunto(s)
Codón sin Sentido/genética , Codón sin Sentido/inmunología , Factor VIII/genética , Factor VIII/inmunología , Empalme Alternativo/genética , Animales , Anticuerpos/sangre , Anticuerpos/genética , Anticuerpos/inmunología , Antígenos/genética , Secuencia de Bases , Células CHO , Codón sin Sentido/metabolismo , Cricetinae , ADN Complementario/genética , ADN Complementario/metabolismo , Factor VIII/metabolismo , Hemofilia A/sangre , Hemofilia A/genética , Hemofilia A/inmunología , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo , Mutación Puntual , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Transfección
15.
J Thromb Haemost ; 1(9): 1935-44, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12941034

RESUMEN

UNLABELLED: Coagulation factors (F)VIIa, FXa and thrombin are implicated in cellular responses in vascular, mesenchymal and inflammatory cells. Fibroblasts are the most abundant cells in connective tissue, and damage to blood vessels places coagulation factors in contact with these and other cell types. OBJECTIVES: To investigate cellular responses of primary dermal fibroblasts to FVIIa, FXa and thrombin by following changes in expression of candidate proteins: monocyte chemotactic protein-1 (MCP-1), interleukin-8 (IL-8), interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF), and to determine the expression of receptors implicated in signaling by these coagulation factors. METHODS: Steady-state mRNA levels were quantified by RNase protection assay, and protein secretion by ELISA. PAR gene expression was assessed by ribonuclease protection assay and conventional and quantitative reverse-transcription-polymerase chain reaction. RESULTS: FVIIa did not induce the candidate genes. In contrast, FXa and thrombin induced MCP-1 mRNA and protein secretion strongly, IL-8 moderately, and IL-6 weakly. Neither FXa nor thrombin induced VEGF mRNA or protein secretion, although FXa induced VEGF protein secretion in lung fibroblasts. Comparison of the presence of candidate receptors in the two fibroblast subtypes demonstrated higher levels of PAR-1 and PAR-3 in lung fibroblasts relative to their dermal counterparts and the additional expression of PAR-2. CONCLUSIONS: FXa and thrombin induce expression of MCP-1, IL-8 and IL-6, and distribution and expression of PARs on dermal fibroblasts is reduced relative to their lung counterparts. Tissue origin may influence the cellular response of fibroblasts to coagulation proteases.


Asunto(s)
Factores de Coagulación Sanguínea/farmacología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Piel/citología , Células Cultivadas , Quimiocina CCL2/genética , Factor VIIa/farmacología , Factor X/farmacología , Fibroblastos/metabolismo , Humanos , Interleucina-6/genética , Interleucina-8/genética , Pulmón/citología , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos , Receptor PAR-1/genética , Receptor PAR-2/genética , Receptores de Trombina/genética , Trombina/farmacología , Factor A de Crecimiento Endotelial Vascular/genética
16.
Transplantation ; 68(6): 832-9, 1999 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-10515384

RESUMEN

BACKGROUND: Thrombotic vascular occlusion resulting in infarction occurs during hyperacute rejection of allografts transplanted into sensitized patients and remains a major problem in experimental xenotransplantation. A similar process is also found in disorders of diverse etiology including atherosclerosis, vasculitis, and disseminated intravascular coagulation. METHODS: We have previously constructed two membrane-tethered anticoagulant fusion proteins based on human tissue factor pathway inhibitor and the leech anticoagulant hirudin and demonstrated their functional efficacy in vitro. These constructs have now been modified by the addition of a P-selectin sequence to the cytoplasmic tail to localize them in Weibel-Palade bodies. They have been transfected into Weibel-Palade body-positive endothelial cells isolated from the inferior vena cava of normal pigs. RESULTS: In resting endothelial cells, fusion protein expression colocalized with P-selectin and was confined to Weibel-Palade bodies. These cells had a procoagulant phenotype in recalcified human plasma. However, after activation with phorbol ester the anticoagulant proteins were rapidly relocated to the cell surface where they specifically inhibited the clotting of human plasma. CONCLUSIONS: Novel anticoagulant molecules may prove useful therapeutic agents for gene therapy in thrombotic disease and postangioplasty or for transgenic expression in animals whose organs may be used for clinical xenotransplantation. Expression in vascular endothelial cells may be regulated by inclusion of P-selectin cytoplasmic sequence, to restrict cell surface expression to activated endothelium.


Asunto(s)
Coagulación Intravascular Diseminada/fisiopatología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Lipoproteínas/biosíntesis , Selectina-P/biosíntesis , Animales , Antígenos CD4/farmacología , Fibrinolíticos/farmacología , Hirudinas/biosíntesis , Proteínas de la Membrana/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Porcinos , Porcinos Enanos , Cuerpos de Weibel-Palade/metabolismo
17.
Transplantation ; 67(3): 467-74, 1999 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10030296

RESUMEN

BACKGROUND: Thrombotic vascular occlusion occurs in disorders of diverse etiology, including atherosclerosis, vasculitis, and disseminated intravascular coagulation. The same process results in hyperacute rejection of renal allografts transplanted into sensitized patients and remains a major problem in experimental xenotransplantation. METHODS: We have previously described the design and expression of several genetic constructs encoding novel fusion proteins with anticoagulant properties. They are based on two naturally occurring soluble anticoagulant proteins, human tissue factor pathway inhibitor (hTFPI) and the leech protein hirudin, which act early and late in the clotting cascade, respectively. We report the expression of human hTFPI-CD4 on the surface of immortalized porcine endothelial cells (IPEC), and show that it functions across the species divide as evidenced by the binding of membrane-expressed porcine tissue factor (pTF)-human factor VIIa complexes. RESULTS: Using a human plasma recalcification clotting assay, we distinguished between pTF-dependent and pTF-independent fibrin generation, and we have demonstrated that expression of hTFPI-CD4 on IPEC effectively prevented pTF-dependent clotting. Moreover, we show that when hTFPI-CD4 was co-expressed with the hirudin construct, the procoagulant properties of in vitro cultured, activated IPEC were almost completely abolished. CONCLUSIONS: These results suggest that these novel anticoagulant molecules may prove useful therapeutic agents for gene therapy or for transgenic expression in animals whose organs may be used for cliniCal xenotransplantation.


Asunto(s)
Anticoagulantes , Coagulación Sanguínea , Antígenos CD4/fisiología , Endotelio Vascular/fisiología , Factor VIIa/metabolismo , Factor Xa/metabolismo , Hirudinas/metabolismo , Lipoproteínas/metabolismo , Tromboplastina/fisiología , Animales , Antígenos CD4/genética , Células Cultivadas , Hirudinas/genética , Humanos , Cinética , Sanguijuelas , Lipoproteínas/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Porcinos , Transfección/métodos
18.
Thromb Haemost ; 80(4): 561-5, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9798969

RESUMEN

High FVIII:C levels have previously been shown to be an independent risk factor for thrombosis with 4.8 times higher potential risk of thrombosis in individuals with FVIII:C levels greater than 1.5 u/ml. Recently, we found that raised FVIII:C levels are largely attributable to elevated FVIII:Ag levels. The determinants of FVIII:Ag levels are unclear and might be partly genetic. The promoter of the F8 gene has recently been characterised we therefore investigated the promoter and the 3' terminus of the F8 gene for possible polymorphisms associated with raised FVIII:Ag levels in 62 selected individuals with a thrombotic tendency. We confirm previous reports that raised FVIII:C levels are largely attributable to an elevation in FVIII:Ag and this is also associated with elevation of vWF; non-O blood group: relatively short APTT and relatively low APC ratio. We screened 1140 bp of the proximal promoter including the protein binding sites identified by DNase I footprint analysis by SSCP, however no polymorphisms were identified. Direct DNA sequence analysis of the region -542 to +165 failed to identify any sequence polymorphisms. The recently described polymorphism in the polyadenylation cleavage site in the prothrombin gene associated with increased prothrombin activity prompted us to screen the region surrounding the 3' terminus of the F8 gene for polymorphisms but we found none.


Asunto(s)
Factor VIII/genética , Polimorfismo Genético , Trombosis de la Vena/genética , Secuencia de Bases , Coagulación Sanguínea/genética , Factor VIII/metabolismo , Humanos , Datos de Secuencia Molecular , Trombosis de la Vena/sangre
19.
Thromb Haemost ; 56(1): 6-8, 1986 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-3775691

RESUMEN

Twelve healthy subjects received ethamsylate or a placebo by mouth over a 48 h period in a randomized double-blind trial. The template bleeding time (including estimation of amount of blood loss), platelet aggregation studies, and plasma levels of plasminogen, alpha 2-antiplasmin and fibronectin were carried out before and during treatment. The effect of a single dose (600 mg) of aspirin, given 24 h after commencement of treatment, was also determined. Neither ethamsylate nor placebo caused a significant change in the basal values of any of the variables monitored but both the prolongation of the bleeding time and the amount of blood loss induced by aspirin were significantly less during ethamsylate treatment than with placebo. Ethamsylate failed to prevent the aspirin-induced elimination of the secondary wave of platelet aggregation. We conclude that ethamsylate may reduce the haemorrhagic symptoms associated with mild functional platelet defects.


Asunto(s)
Bencenosulfonatos/farmacología , Etamsilato/farmacología , Hemostasis/efectos de los fármacos , Adulto , Aspirina/farmacología , Tiempo de Sangría , Femenino , Fibronectinas/sangre , Humanos , Cinética , Masculino , Plasminógeno/metabolismo , Agregación Plaquetaria/efectos de los fármacos , alfa 2-Antiplasmina/metabolismo
20.
Thromb Haemost ; 77(2): 238-42, 1997 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9157573

RESUMEN

Molecular genetic studies have shown that development of antibodies to factor VIII (inhibitors) occurs most frequently in patients with severe haemophilia due to major gene lesions including inversions, stop codons and large deletions. Previous studies of HLA type were performed on inhibitor and non-inhibitor patients with diverse uncharacterized mutations which may have confounded detection of significant associations. We therefore selected a group of patients with a single mutation type, the prevalent intron 22 inversion, with or without inhibitors, to determine HLA genotype. Seventy-one such patients, 42 without and 29 with inhibitors (13 high, 9 low and 7 transient responders) were genotyped for MHC Class I HLA-A, -B, -C and Class II HLA-DQA, -DQB and -DRB loci. No strong correlation of any HLA-allele to inhibitor or non-inhibitor status was found. However, alleles of the haplotype HLA-A3, HLA-B7, HLA-C7, HLA-DQA0102, HLA-DQB0602, HLA-DR15 occurred more often in inhibitor patients. Since the alleles of this extended haplotype are common in the North European population only a very strong association would achieve statistical significance. Further studies of groups of patients similar to those studied here will be needed to confirm or exclude this association.


Asunto(s)
Inversión Cromosómica , Factor VIII/inmunología , Genes MHC Clase II , Antígenos HLA-D/genética , Hemofilia A/inmunología , Intrones/genética , Isoanticuerpos/inmunología , Alelos , Susceptibilidad a Enfermedades , Factor VIII/uso terapéutico , Genotipo , Alemania/epidemiología , Antígenos HLA-D/inmunología , Haplotipos , Hemofilia A/epidemiología , Hemofilia A/terapia , Humanos , Isoanticuerpos/genética , Masculino , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA