Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Wound Care ; 22(3): 144-6, 148, 150-2 passim, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23665733

RESUMEN

OBJECTIVE: To compare the efficacy and safety of negatively-charged polystyrene microspheres (NCM)with controls (saline soaks) in the treatment of hard-to-heal wounds of various aetiologies. METHOD: Patients with one or more hard-to-heal wounds, defined as refractory to healing for at least 4 weeks, or those with exposed bone, tendon or ligament, were eligible for inclusion and were randomised to either NCM (PolyHeal; MediWound Ltd.) or controls, both applied twice daily for 4 weeks. Patients were monitored bi-weekly for an additional 8 weeks, while treated by standard wound care, at the investigators' discretion, and were re-evaluated 2 years after inclusion. The primary endpoint was defined as coverage of> 75% of the wound area by light-red granulation tissue after 4 weeks of treatment. RESULTS: Fifty-eight patients completed the study, 32 in the NCM group and 26 in the control group. The two most common wound types were those with primary etiologies of venous insufficiency and postoperative/post trauma. In the NCM group 47% of patients achieved > 75% light red granulation tissue after 4 weeks compared with 15% of patients in the control group (p=O.O I). The mean wound surface area in the NCM group was reduced by 39.0% after 4 weeks compared with 14.9% in the control group (p=0.02).The achievement of> 75% light red granulation tissue and reduction of mean wound surface area was also observed in the two main sub-groups (venous insufficiency and postoperative/post trauma), although it was not statistically significant, possibly due to the small sample size in each sub-group. CONCLUSION: This study demonstrates that compared to control treatment, NCM treatment of hard to-heal and chronic wounds improves formation of healthy granulation tissue and reduces wound size thus in fact 'kick-starting' the healing process and 'dechronifying' chronic wounds.


Asunto(s)
Aniones/uso terapéutico , Tejido de Granulación/crecimiento & desarrollo , Microesferas , Úlcera Cutánea/terapia , Cicatrización de Heridas , Adulto , Anciano , Enfermedad Crónica , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Poliestirenos , Estudios Prospectivos , Solución Salina Hipertónica , Electricidad Estática , Resultado del Tratamiento
3.
Haemophilia ; 14 Suppl 3: 202-10, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18510543

RESUMEN

During the haemostatic response, the formation of a primary platelet plug limits bleeding and provides a surface for clotting factors to assemble and become activated. The initial platelet plug is stabilized by fibrin monomers, covalently cross-linked by FXIII, forming a platelets-fibrin thrombus. Defects in platelets as well as inherited deficiencies of coagulation factors including fibrinogen, FII, FV, FV + FVIII, FVII, FX, FXI and FXIII deficiencies, generally lead to lifelong bleeding disorders, whose severity of bleeding symptoms is heterogeneous in platelets abnormalities but generally inversely proportional to the degree of the factor deficiency in rare bleeding disorders (RBDs). The prevalence of platelet defects among the general population has not been established, whereas for RBDs it ranges from approximately 1 in 2 million to 1 in 500,000, being higher in countries where consanguineous marriages are diffused. As a consequence of the rarity of these deficiencies, the type and severity of bleeding symptoms, the underlying molecular defects, and the actual management of bleeding episodes are not well established. In this review the main features, diagnosis, available treatment options and treatment complications of the platelet disorders, caused by abnormalities in platelet receptors for adhesive proteins, platelet receptors for soluble agonists, platelet granules, signal transduction pathways, or procoagulant phospholipids will be discussed by Dr Cattaneo, whereas fibrinogen deficiency and FXIII deficiency will be described by Dr Inbal and Dr de Moerloose, respectively. Finally, the update of the Rare Bleeding Disorders Database will be presented by Dr Spreafico.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/diagnóstico , Trastornos de las Plaquetas Sanguíneas/diagnóstico , Trastornos Hemorrágicos/diagnóstico , Trastornos de la Coagulación Sanguínea Heredados/genética , Trastornos de la Coagulación Sanguínea Heredados/terapia , Inhibidores de Factor de Coagulación Sanguínea/uso terapéutico , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/terapia , Bases de Datos Genéticas , Esquema de Medicación , Genotipo , Trastornos Hemorrágicos/genética , Trastornos Hemorrágicos/terapia , Humanos , Sistemas de Registros Médicos Computarizados , Fenotipo , Guías de Práctica Clínica como Asunto , Reino Unido
4.
J Clin Invest ; 78(4): 1112-9, 1986 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-3489737

RESUMEN

von Willebrand protein was found to promote the incorporation of platelets into evolving fibrin thrombi. Using formalin-treated or fresh platelets, both the initial rate and extent of platelet incorporation into polymerizing fibrin were dependent on von Willebrand protein. von Willebrand protein was incorporated into evolving fibrin thrombi in parallel with platelets. Soluble fibrin monomer covalently linked to acrylonitrile beads (Matrex 102) bound von Willebrand protein specifically and saturably with an apparent approximate dissociation constant (KD) of 15 micrograms/ml. Glycocalicin, the water-soluble proteolytic fragment of glycoprotein Ib, bound to fibrin monomer in this system specifically and saturably, as well, with an apparent approximate KD of 5 micrograms/ml, but only in the presence of saturating concentrations of von Willebrand protein. These data demonstrate that the initial rate and extent of platelet incorporation into evolving fibrin thrombi are dependent on von Willebrand protein; von Willebrand protein serves as a link between polymerizing fibrin and platelet surface glycoprotein Ib; and von Willebrand protein binds to fibrin monomer and is thereby able to bind to platelet surface glycoprotein Ib in the absence of ristocetin.


Asunto(s)
Plaquetas/metabolismo , Fibrina/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria , Factor de von Willebrand/farmacología , Plaquetas/efectos de los fármacos , Proteínas Sanguíneas/metabolismo , Humanos , Cinética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Polímeros/metabolismo , Solubilidad
5.
J Thromb Haemost ; 15(8): 1601-1606, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28581691

RESUMEN

Essentials Prophylaxis is the standard of care for congenital factor XIII-A (FXIII-A) deficiency. Six children with FXIII-A deficiency received once-monthly prophylaxis with recombinant FXIII-A. Prophylaxis was well tolerated and no anti-FXIII antibodies were detected. Prophylaxis was effective with an annualized bleeding rate of zero. SUMMARY: Background Factor XIII deficiency is a rare, severe congenital bleeding disorder. Monthly prophylaxis with recombinant FXIII A-Subunit (rFXIII) has demonstrated favorable safety and efficacy in patients aged ≥ 6 years, and may similarly benefit younger children. Objective To evaluate the long-term safety and efficacy of rFXIII in children aged < 6 years with congenital FXIII A-subunit deficiency. Patients/methods Six children, who had previously completed a single-dose pharmacokinetic trial of rFXIII, received 35 IU kg-1 rFXIII every 28 days (± 2 days) for a minimum of 52 weeks, and were evaluated for bleeding and adverse events. The Berichrom FXIII activity assay was used to monitor FXIII activity. Results The children, three girls and three boys, had an average age of 3.0 years (range: 1-4 years) at enrollment. The total treatment duration was 1.8-3.5 years, giving a total of 16.6 patient-years. No antibody development, thromboembolic events or allergic reactions occurred. There were 93 mild and seven moderate adverse events. Two adverse events (lymphopenia and gastroenteritis) were reported as probably or possibly related to rFXIII in two children. Two serious adverse events, unrelated to rFXIII, were reported in a single child, each related to head injury, and neither resulting in intracranial hemorrhage. The geometric mean FXIII activity trough was 0.19 IU mL-1 . No bleeding episodes requiring treatment with an FXIII-containing hemostatic agent occurred during the trial; thus, the annualized bleeding rate was 0. Conclusions Consistent with data from older age groups, prophylaxis with rFXIII appears to be safe and effective in young children with congenital FXIII A-subunit deficiency.


Asunto(s)
Coagulantes/administración & dosificación , Deficiencia del Factor XIII/tratamiento farmacológico , Factor XIII/administración & dosificación , Factores de Edad , Preescolar , Coagulantes/efectos adversos , Esquema de Medicación , Factor XIII/efectos adversos , Deficiencia del Factor XIII/sangre , Deficiencia del Factor XIII/diagnóstico , Deficiencia del Factor XIII/genética , Factor XIIIa/genética , Femenino , Humanos , Lactante , Masculino , Proteínas Recombinantes/administración & dosificación , Factores de Tiempo , Resultado del Tratamiento
6.
J Thromb Haemost ; 4(1): 19-25, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16129024

RESUMEN

Factor XIII is a plasma transglutaminase that participates in the final stage of the coagulation cascade. Thrombin-activated FXIII (FXIIIa) catalyzes the formation of covalent cross-links between gamma-glutamyl and epsilon-lysyl residues on adjacent fibrin chains in polymerized fibrin to yield the mature clot. In addition to its role in hemostasis, FXIII is known to participate in wound healing and embryo implantation, which are processes involving angiogenesis. In this review, we discuss the role of FXIII in angiogenesis and the molecular mechanisms underlying its proangiogenic effects. The FXIII role in tissue repair and remodeling may at least in part be attributed to its pro-angiogenic activity.


Asunto(s)
Factor XIII/fisiología , Neovascularización Fisiológica/fisiología , Animales , Implantación del Embrión , Homeostasis , Humanos , Cicatrización de Heridas
7.
Stroke ; 32(12): 2753-8, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11739968

RESUMEN

BACKGROUND AND PURPOSE: Although risk factors for carotid artery stenosis caused by atherosclerosis are known, it is unclear what triggers "activation" of the atherosclerotic plaques and the ensuing thromboembolic cerebral events. The aim of this study was to evaluate whether thrombophilic factors, platelet glycoprotein (GP) polymorphisms, and homocysteine are associated with a risk of ischemic events in patients with significant carotid stenosis. METHODS: Consecutive patients with >/=50% carotid stenosis, whether symptomatic (with ipsilateral ischemic events) or asymptomatic, who were evaluated and followed in a neurovascular clinic were tested for plasma levels of homocysteine, C677T mutation in methylenetetrahydrofolate reductase, G20210A mutation of factor II, factor V Leiden, antiphospholipid antibodies, and polymorphisms of platelet membrane GP: human platelet antigen (HPA)-1, GP Ia (C807T), and GP Ib (variable number of tandem repeats, Kozak, and HPA-2). RESULTS: Eighty-six asymptomatic and 67 symptomatic patients were evaluated. The former group was older (73.7+/-6.9 versus 69.5+/-9.1 years, P=0.02). Major risk factors for stroke were similar in both groups. In symptomatic patients versus asymptomatic patients, hyperhomocysteinemia was 3-fold more frequent (34.3% versus 12.8%, respectively; P=0.002) and HPA-1a/b was almost 2-fold more common (38.8% versus 20.9%, respectively; P=0.01). All other thrombophilic factors and platelet polymorphisms studied did not differ significantly between the 2 groups. Multivariate analysis revealed that hyperhomocysteinemia and the HPA-1a/b genotype conferred a significant risk of cerebral ischemic events, with odds ratios (95% CI) of 4.07 (1.7 to 9.7) and 3.4 (1.5 to 7.8), respectively. CONCLUSIONS: Hyperhomocysteinemia and HPA-1a/b are independent risk factors for ischemic events in patients with significant carotid stenosis.


Asunto(s)
Antígenos de Plaqueta Humana/genética , Estenosis Carotídea/sangre , Hiperhomocisteinemia/sangre , Polimorfismo Genético/genética , Accidente Cerebrovascular/sangre , Anciano , Sustitución de Aminoácidos/genética , Anticuerpos Antifosfolípidos/sangre , Estenosis Carotídea/diagnóstico , Estenosis Carotídea/epidemiología , Comorbilidad , Factor V/genética , Femenino , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/diagnóstico , Hiperhomocisteinemia/epidemiología , Integrina beta3 , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2) , Análisis Multivariante , Oportunidad Relativa , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Glicoproteínas de Membrana Plaquetaria/genética , Protrombina/genética , Medición de Riesgo , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/epidemiología
8.
J Thromb Haemost ; 1(3): 433-8, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12871446

RESUMEN

The effect of thromboprophylaxis with low molecular weight heparin (LMWH), on the subsequent live birth rate, in thrombophilic women with recurrent miscarriage has not been sufficiently assessed. The present study is a cohort study undertaken to assess the effect of enoxaparin on the subsequent live birth rate in women with hereditary thrombophila. Eighty-five patients with three or more consecutive pregnancy losses and a hereditary thrombophilia subsequently conceived. Thirty-seven were treated with daily subcutaneous injections of enoxaparin 40 mg and 48 were not treated. The outcome of the subsequent pregnancy was assessed in both groups of patients in terms of live births or repeat miscarriage. Forty-seven of the 85 patients were subsequently delivered, 38 have miscarried. Twenty-six of the 37 pregnancies in treated patients (70.2%) resulted in live births, compared with 21 of 48 (43.8%) in untreated patients (P < 0.02, OR 3.03, 95% CI 1.12-8.36). The beneficial effect was seen mainly in primary aborters, i.e. women with no previous live births (P < 0.008, OR 9.75, 95% CI 1.59-52.48). This benefit was also found in patients with a poor prognosis for a live birth (five or more miscarriages), where the live birth rate was increased from 18.2% to 61.6%. However, the benefit was not statistically significant, probably due to the small number of patients. If the beneficial effects of enoxaparin are confirmed by additional studies, thromboprophylaxis can be recommended for patients with hereditary thrombophilia and recurrent pregnancy loss.


Asunto(s)
Tasa de Natalidad , Trombofilia/complicaciones , Trombosis/prevención & control , Aborto Habitual/prevención & control , Adulto , Enoxaparina/uso terapéutico , Salud de la Familia , Femenino , Fibrinolíticos/uso terapéutico , Humanos , Cariotipificación , Embarazo , Complicaciones Hematológicas del Embarazo , Resultado del Embarazo , Trombofilia/tratamiento farmacológico , Trombofilia/genética , Trombosis/complicaciones
9.
J Thromb Haemost ; 2(10): 1790-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456491

RESUMEN

BACKGROUND: Hereditary factor (F)XIII deficiency is a rare bleeding disorder mostly due to mutations in FXIII A subunit. OBJECTIVES: We studied the molecular basis of FXIII deficiency in patients from 10 unrelated families originating from Israel, India and Tunisia. METHODS: Exons 2-15 of genomic DNA consisting of coding regions and intron/exon boundaries were amplified and sequenced. Structural analysis of the mutations was undertaken by computer modeling. RESULTS: Seven novel mutations were identified in the FXIIIA gene. The propositus from the Ethiopian-Jewish family was found to be a compound heterozygote for two novel mutations: a 10-bp deletion in exon 12 at nucleotides 1652-1661 (followed by 22 altered amino acids and termination codon) and Ala318Val mutation. The propositus of the Tunisian family was homozygous for C insertion after nucleotide 863 within a stretch of six cytosines of exon 7. This insertion results in generation of eight altered amino acids followed by a termination codon downstream. The propositus from Indian-Jewish origin was found to be homozygous for G to T substitution at IVS 11 [+1] resulting in skipping of exons 10 and 11. In addition to the Ala318Val mutation, three of the novel mutations identified are missense mutations: Arg260Leu, Thr398Asn and Gly210Arg each occurring in a homozygous state in an Israeli-Arab and two Indian families, respectively. CONCLUSIONS: Structure-function correlation analysis by computer modeling of the new missense mutations predicted that Gly210Arg will cause protein misfolding, Ala318Val and Thr398Asn will interfere with the catalytic process or protein stability, and Arg260Leu will impair dimerization.


Asunto(s)
Deficiencia del Factor XIII/genética , Factor XIII/genética , Mutación , Catálisis , Codón sin Sentido , Análisis Mutacional de ADN , Dimerización , Exones , Factor XIII/química , Salud de la Familia , Humanos , Modelos Moleculares , Mutación Missense , Pliegue de Proteína , Subunidades de Proteína/genética , Eliminación de Secuencia
10.
Thromb Haemost ; 77(6): 1062-7, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9241732

RESUMEN

In this report we describe the molecular basis of FXIII a-subunit deficiency in three unrelated Palestinian Arab families. In three patients representing each family two substitutions were identified in exon 14 on both alleles: C to G change resulting in a Gln651Glu substitution (a previously described polymorphism) and a T to C transition causing Leu660Pro substitution. The latter is a new mutation which creates a restriction site for FnuDII enzyme. Restriction analysis performed in members of the three families clearly distinguished between severely affected patients, obligate carriers and unaffected subjects. A population survey failed to detect the mutation among 250 Jewish individuals but did detect two heterozygotes among 300 Arabs suggesting a 0.0033 frequency for the Pro660 allele in this population. In two out of the three families the Pro660 allele was linked to allele 5 of the 5' short tandem repeat polymorphism within the FXIII a-subunit gene suggesting that the mutation might have occurred at least twice. cDNA obtained from mRNA isolated from patients' platelets and monocytes appeared similar in size to that of normal control indicating that the Leu660Pro mutation does not affect mRNA synthesis. Computer modeling based on cristallographic studies of the a-subunit of factor XIII predicted that the mutant protein is expected to misfold into a structure which is either unstable or susceptible to degradation.


Asunto(s)
Árabes , Deficiencia del Factor XIII/genética , Factor XIII/genética , Mutación , Deficiencia del Factor XIII/etnología , Femenino , Humanos , Leucina , Masculino
11.
Thromb Haemost ; 70(6): 1058-62, 1993 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-8165601

RESUMEN

Type IIB von Willebrand disease (vWD) is characterized by a selective loss of high molecular weight von Willebrand factor (vWF) multimers in plasma due to their abnormally enhanced reactivity with platelets. Several missense mutations in the platelet glycoprotein Ib (GPIb) binding domain of vWF were recently characterized that cause type IIB vWD. The effect of type IIB mutation Arg(545)Cys on vWF binding to platelet GPIb was studied using recombinant wild type (rvWFWT) and mutant rvWFR545C expressed in COS-7 cells. In the absence of ristocetin, 50% of rvWFR545C bound spontaneously to platelet GPIb and the binding increased to 70% in the presence of 0.2 mg/ml ristocetin; rvWFWT did not bind significantly under either condition. Botrocetin-induced binding of rvWFR545C was only slightly increased compared to rvWFWT. These data demonstrate that the Arg(545)Cys mutation increases the affinity of vWF for GPIb, resulting in the characteristics gain-of-function type IIB vWD phenotype.


Asunto(s)
Arginina/genética , Cisteína/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética , Secuencia de Bases , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Humanos , Datos de Secuencia Molecular , Mutación , Proteínas Recombinantes/genética , Ristocetina/farmacología
12.
Thromb Haemost ; 85(5): 806-9, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11372672

RESUMEN

Acquired von Willebrand syndrome (AVWS) has been associated mainly with monoclonal gammopathy of uncertain significance (MGUS), clonal lymphoproliferative or myeloproliferative disorders and autoimmunity. In the present work we studied 6 patients with AVWS: four with MGUS IgG (lambda or kappa), one with small lymphocytic lymphoma and one with agnogenic myeloid metaplasia (AMM). All the patients underwent a pharmacokinetic analysis at presentation in order to study potential differences in recovery, clearance (CL) or terminal half-life (THL) following administration of von Willebrand factor (VWF) concentrate. In all the patients with AVWS an increase in clearance and a decrease in THL was observed as compared to these parameters in patients with hereditary type 3 von Willebrand disease (VWD). No difference in recovery was observed among the groups. The increase in clearance and the decrease in THL were significantly more pronounced in the group of MGUS patients (57.93 +/- 25.6 ml/h/kg, and 1.39 +/- 0.5 h, respectively) as compared to these parameters in the AMM (8.06 ml/h/kg, and 6.96 h, respectively) or the lymphoma (4.76 ml/h/kg, and 6.76 h. respectively) patients (p = 0.03 for clearance and 0.001 for THL). These data indicate that the pharmacokinetic analysis can be a useful tool to distinguish between MGUS-related and other causes of AVWS, and to plan an appropriate treatment accordingly.


Asunto(s)
Factor VIII/farmacocinética , Enfermedades de von Willebrand/diagnóstico , Factor de von Willebrand/farmacocinética , Anciano , Tiempo de Sangría , Factor VIII/administración & dosificación , Femenino , Semivida , Humanos , Leucemia Linfocítica Crónica de Células B/complicaciones , Masculino , Tasa de Depuración Metabólica , Persona de Mediana Edad , Gammopatía Monoclonal de Relevancia Indeterminada/complicaciones , Mielofibrosis Primaria/complicaciones , Ristocetina , Enfermedades de von Willebrand/tratamiento farmacológico , Enfermedades de von Willebrand/etiología , Factor de von Willebrand/administración & dosificación
13.
Thromb Haemost ; 81(2): 229-33, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10063997

RESUMEN

We studied the safety and efficacy of treatment with continuous infusion of a von Willebrand factor (vWF) concentrate Haemate-P in patients with von Willebrand disease (vWD). Three patients with mild and 5 patients with severe forms of vWD, were treated with continuous infusion of Haemate-P by minipump. The indications for treatment were: to prevent bleeding during 9 surgical procedures or 1 vaginal delivery in 6 patients and to treat 2 bleeding episodes in 2 patients. The patients were monitored daily for factor VIII (FVIII:C) and ristocetin cofactor (vWF: RCo) levels and the infusion rate was adjusted to maintain the desired therapeutic level of vWF:RCo. The treatment was effective in preventing surgical bleeding and controlling bleeding episodes. All factor VIII:C and most of the vWF:RCo levels measured during the study period were above the target therapeutic levels. A significant decrease in clearance of FVIII:C and vWF:RCo was observed over the treatment period. Haemate-P consumption averaged 24.3+/-7.9 vWF:RCo U/kg/day which is approximately half the expected dose had intermittent bolus injections been used. We suggest that continuous Haemate-P infusion is superior to intermittent bolus injections for the treatment of vWD patients by virtue of its efficiency, simplicity and considerable savings.


Asunto(s)
Pérdida de Sangre Quirúrgica/prevención & control , Factor VIII/uso terapéutico , Enfermedades de von Willebrand/tratamiento farmacológico , Factor de von Willebrand/uso terapéutico , Adolescente , Adulto , Cesárea , Niño , Procedimientos Quirúrgicos Electivos , Factor VIII/administración & dosificación , Factor VIII/análisis , Femenino , Humanos , Bombas de Infusión , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Embarazo , Complicaciones Hematológicas del Embarazo/tratamiento farmacológico , Cuidados Preoperatorios , Seguridad , Resultado del Tratamiento , Factor de von Willebrand/administración & dosificación , Factor de von Willebrand/análisis
14.
Thromb Haemost ; 84(5): 912-7, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11127876

RESUMEN

Antiplatelet drugs are the mainstays of therapy for acute and chronic cardiovascular diseases. S-nitroso-AR545C - an S-nitrosoderivative of a recombinant von Willebrand factor fragment AR545C spanning Ala 444 to Asp 730 and containing an Arg 545 Cys mutation, was previously found to inhibit ristocetin- and ADP-induced platelet aggregation and the interaction of platelets with extracellular matrix (ECM). In the current study we tested the antithrombotic properties of S-nitroso-AR545C on guinea pig platelets and in a platelet-rich thrombosis model in the guinea pig. Preincubation of guinea pig platelets with 0.1 microM of S-nitroso-AR545C decreased ristocetin-induced agglutination by 40% (p = 0.009) and completely abolished ADP-induced aggregation (p <0.0001). At concentration of 1.0 microM, S-nitroso-AR545C completely inhibited platelet adhesion (represented by surface coverage - SC) and decreased aggregate formation (represented by average aggregate size - AS) by more than 50%. Treatment of guinea pigs with 1.0 mg/kg S-nitroso-AR545C resulted in a significantly delayed time to arterial occlusion (31.7+/-6.0 min vs. 13.9+/-3.2 min, p <0.02). Similarly, total patency time was longer in the group injected with S-nitroso-AR545C compared to the control group. However, the difference was not statistically significant (33.8+/-6.3 min vs. 20.2+/-3.3 min, p = 0.07). No change in platelet count, hematocrit and bleeding time was observed 60 min after injection compared to baseline. In contrast, a significant decrease in SC (p <0.0001) and AS (p <0.01) were observed 60 min after the injection of S-nitroso-AR545C, whereas no change in these parameters was observed in the control group. These observations indicate that S-nitroso-AR545C exhibits significant antiadhesive and antiaggregating effects in-vitro and inhibits clot formation in-vivo suggesting that this compound may have potential therapeutic advantages.


Asunto(s)
Trombosis de las Arterias Carótidas/tratamiento farmacológico , Fragmentos de Péptidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Factor de von Willebrand/farmacología , Animales , Modelos Animales de Enfermedad , Cobayas , Fragmentos de Péptidos/uso terapéutico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Factor de von Willebrand/uso terapéutico
15.
Thromb Haemost ; 77(6): 1086-9, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9241737

RESUMEN

Purpura fulminans is associated with homozygous protein C and homozygous protein S deficiency or may follow bacterial or viral infections. We present 2 children from 2 unrelated Arab families with purpura fulminans who were double heterozygotes for factor V Leiden inherited from their fathers and protein S deficiency inherited from their mothers. No previous thrombotic events have occurred in either patient or their respective family members. In one patient sepsis accompanied by disseminated intravascular coagulation appeared to be the trigger of purpura fulminans. In the other patient varicella infection preceded purpura fulminans and was also associated with disseminated intravascular coagulation. This report emphasizes the need for evaluation of hereditary defects in the inhibitory mechanisms of blood coagulation in patients with purpura fulminans at any age.


Asunto(s)
Enfermedades Transmisibles/complicaciones , Coagulación Intravascular Diseminada/genética , Factor V/genética , Vasculitis por IgA/genética , Deficiencia de Proteína S/genética , Preescolar , Coagulación Intravascular Diseminada/complicaciones , Coagulación Intravascular Diseminada/fisiopatología , Femenino , Heterocigoto , Humanos , Vasculitis por IgA/etiología , Vasculitis por IgA/fisiopatología , Masculino , Linaje , Deficiencia de Proteína S/complicaciones
16.
Thromb Haemost ; 67(6): 618-22, 1992 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-1324533

RESUMEN

Von Willebrand disease (vWD) type IIA is characterized by decreased ristocetin-induced platelet aggregation, and by the absence from plasma of high molecular weight multimers of von Willebrand factor (vWF). Most mutations causing vWD type IIA are clustered within the A2 domain of the mature vWF subunit that is encoded by exon 28. Using the polymerase chain reaction (PCR), the entire exon 28 from patients with vWD type IIA and normal controls was amplified and sequenced. Three missense mutations were detected that result in the amino acid substitutions were detected that result in the amino acid substitutions Arg(834)----Trp, Gly(742)----Glu, and Ser(743)----Leu. The first mutation occurred independently in three unrelated families; each of the latter mutations was found in one family. By restriction endonuclease analysis and allele-specific oligonucleotide (ASO) hybridization the mutations were confirmed in affected family members and excluded in unaffected members and 50 normal controls. The apparently high frequency of identical independent mutations among patients with vWD type IIA suggests that a precise diagnosis may be possible in a majority of patients using relatively simple recombinant DNA screening assays.


Asunto(s)
Enfermedades de von Willebrand/genética , Alelos , Aminoácidos/genética , Secuencia de Bases , Enzimas de Restricción del ADN , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Mutación/genética , Hibridación de Ácido Nucleico , Oligonucleótidos/genética , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa
17.
Am J Med Genet ; 55(3): 372-8, 1995 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-7726239

RESUMEN

We describe 2 sibs (brother and sister) with myopathy, sideroblastic anemia, lactic acidosis, mental retardation, microcephaly, high palate, high philtrum, distichiasis, and micrognathia. Very low levels of cytochromes a, b, and c were detected in the patients' muscle mitochondria. Deposition of iron within the mitochondria of bone marrow erythroblasts was observed on electron microscopy. Irregular and enlarged mitochondria with paracrystalline inclusions were also seen on electron microscopy of the patients' muscle specimen. Examination of DNA from the affected sibs showed no deletions in the mitochondrial DNA nor the mutations identified in the syndromes of mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes (MELAS) or myoclonus, and epilepsy associated with rugged-red fibers (MERRF). Since the parents were first cousins and 2 of 6 sibs (male and female) were affected, we suggest that the syndrome expressed by our patients represents a previously unknown autosomal recessive disorder that includes mitochondrial myopathy, lactic acidosis, and sideroblastic anemia.


Asunto(s)
Anomalías Múltiples , Acidosis Láctica , Anemia Sideroblástica , Discapacidad Intelectual , Miopatías Mitocondriales , 5-Aminolevulinato Sintetasa/sangre , Adulto , Consanguinidad , Citocromos/análisis , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Femenino , Humanos , Masculino , Mitocondrias Musculares/enzimología , Porfobilinógeno Sintasa/sangre , Síndrome
18.
Int J Oncol ; 19(5): 897-902, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11604985

RESUMEN

BAT is a monoclonal antibody produced against membranes of Daudi cells that induces anti-tumor activity in mice against a variety of solid murine and human tumors, mediated by its immune stimulatory properties on murine and human lymphocytes. The present study analyzes the effect of BAT on leukemia/lymphoma using the BCL1 model of leukemia/lymphoma in BALB/C mice. BAT antibody binds to BCL1 leukemia cells and recognizes a 48 kDa protein similar to the antigen on Daudi cells. Mice inoculated with leukemia cells were treated either by direct BAT injections or by adoptive transfer of lymphocytes from BAT-injected mice. Administration of BAT monoclonal antibody was either once, on day 14, or daily on days 10-13 post tumor inoculation. A single injection of BAT resulted in reduction of peripheral blood tumor cells, however additional injections further decreased the tumor cell number reaching a 95-fold reduction on day 20 post tumor inoculation. Anti-tumor effect was also obtained when animals were injected with splenocytes from BAT-treated donor mice. A significant prolongation of survival of BAT-treated mice was observed although with no cure. The results of this study indicate that BAT might be used for reducing the tumor burden in leukemia for immunotherapy and in combination with other treatment modalities.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfoma de Burkitt/inmunología , Leucemia Experimental/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Traslado Adoptivo , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo , Humanos , Leucemia Experimental/mortalidad , Leucemia Experimental/patología , Linfoma/mortalidad , Linfoma/patología , Ratones , Ratones Endogámicos BALB C , Peso Molecular , Proteínas de Neoplasias/inmunología , Bazo/citología , Bazo/inmunología , Tasa de Supervivencia , Células Tumorales Cultivadas
19.
Bone Marrow Transplant ; 34(5): 459-63, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15247931

RESUMEN

Antithymocyte globulin (ATG) is increasingly used in pre-allogeneic stem cell transplantation (allo-SCT) conditioning regimens to prevent graft rejection and graft-versus-host disease. However, ATG was also found to be associated with increased incidence of thrombosis during organ transplantation. In the present study, we tested the coagulation status of 21 patients with hematologic malignancies undergoing allo-SCT who received ATG-based (11 patients) or non-ATG-based (10) conditioning treatment. We assessed several thrombophilia markers as well as circulating total and endothelial microparticles (TMP/EMP) and soluble CD40 ligand (CD40L). No significant difference in the mean values of prothrombin time, partial thromboplastin time, fibrinogen, antithrombin, protein C, protein S, thrombin-antithrombin III complex, homocysteine levels, prevalence of genetic thrombophilia markers and levels of EMP, TMP or CD40L was observed between the ATG-treated and ATG-untreated patients, as well as before and after conditioning in each group separately. Platelet counts decreased significantly in ATG-treated patients; however, this decrease was not associated with clinical or laboratory evidence of disseminated intravascular coagulation. No patient developed thromboembolic event or veno-occlusive liver disease. Our results suggest that allo-SCT is not associated with increased hypercoagulability and addition of ATG to conditioning regimen has no significant procoagulant effect.


Asunto(s)
Suero Antilinfocítico/administración & dosificación , Coagulación Sanguínea , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Inmunosupresores/administración & dosificación , Acondicionamiento Pretrasplante , Adulto , Anciano , Ligando de CD40/sangre , Femenino , Neoplasias Hematológicas/epidemiología , Neoplasias Hematológicas/genética , Humanos , Incidencia , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Persona de Mediana Edad , Polimorfismo Genético , Prevalencia , Factores de Riesgo , Trombosis/sangre , Trombosis/epidemiología , Trombosis/genética , Trasplante Homólogo
20.
Hematol J ; 2(1): 38-41, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11920232

RESUMEN

Hyperhomocysteinemia is a defined risk factor for venous thromboembolism (VTE). Several polymorphisms of genes encoding for enzymes acting in the remethylation pathway of homocysteine metabolism, ie, methionine synthase (MS) A2756G, methylenetetrahydrofolate reductase (MTHFR) C677T and MTHFR A1298C, can cause increased homocysteine levels particularly in patients with deficiencies of folic acid, vitamin B6, or B12 and hence be potential risk factors for VTE. Indeed, homozygous MTHFR C677T was shown to be a mild risk factor for VTE by some, but not by all, investigators. In this study, we assessed the risk exerted by MS A2756G and MTHFR A1298C in a cohort of patients with idiopathic venous thromboembolism. Homozygosities for MS A2756G and MTHFR A1298C were not found to be statistically significant risk factors for VTE. In addition, no interactions were observed among MS A2756G, MTHFR A1298C and MTHFR C677T in conferring a risk of VTE.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Polimorfismo Genético , Tromboembolia/genética , Trombosis de la Vena/genética , Estudios de Casos y Controles , Factor V/genética , Pruebas Genéticas , Homocigoto , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2) , Mutación Puntual , Protrombina/genética , Factores de Riesgo , Tromboembolia/etiología , Trombosis de la Vena/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA