Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 128(23): 2729-2733, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27663674

RESUMEN

Affinity regulation of integrin αIIbß3 for fibrinogen by inside-out signaling plays a critical role in hemostasis. Calcium and diacylglycerol (DAG)-regulated guanine nucleotide exchange factor I (CalDAG-GEFI) was identified as a Rap1-activating molecule, and its role in inside-out αIIbß3 activation was established in CalDAG-GEFI-deficient mice. However, little information regarding CalDAG-GEFI in human platelets is available. Here, we report a 16-year-old girl with CalDAG-GEFI deficiency who has been suffering from severe bleeding tendency. Although talin and kindlin-3 were normally detected, CalDAG-GEFI was undetectable in her platelets by western blotting. Genetic analysis revealed compound heterozygous CalDAG-GEFI mutations, Lys309X and Leu360del, which were responsible for CalDAG-GEFI deficiency. The functional analysis demonstrated impaired αIIbß3 activation by various agonists except for phorbol myristate acetate, normal calcium mobilization, and impaired Rap1 activation, which were consistent with the phenotype of CalDAG-GEFI-deficient mice. Despite substantial αIIbß3 activation at high agonist concentrations, she had severe bleeding tendency. Further functional analysis demonstrated markedly delayed αIIbß3 activation velocity and decreased shear-induced thrombus formation. Contrary to CalDAG-GEFI-deficient mice, which showed integrin-dependent neutrophil functional abnormality, neutrophil ß2 integrin activation was not impaired in the patient. Our results demonstrate the critical role of CalDAG-GEFI in rapid αIIbß3 activation of human platelets.


Asunto(s)
Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Hemorragia , Mutación Missense , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Adolescente , Sustitución de Aminoácidos , Animales , Plaquetas/patología , Femenino , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Ratones , Ratones Noqueados , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética
2.
Blood ; 120(7): 1499-509, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22730538

RESUMEN

Platelet-associated (PA) IgG autoantibodies play an essential role in primary immune thrombocytopenia (ITP). However, little is known about the epitopes of these Abs. This study aimed to identify critical binding regions for PA anti-αIIbß3 Abs. Because PA anti-αIIbß3 Abs bound poorly to mouse αIIbß3, we created human-mouse chimera constructs. We first examined 76 platelet eluates obtained from patients with primary ITP. Of these, 26 harbored PA anti-αIIbß3 Abs (34%). Further analysis of 15 patients who provided sufficient materials showed that the epitopes of these Abs were mainly localized in the N-terminal half of the ß-propeller domain in αIIb (L1-W235). We could identify 3 main recognition sites in the region; 2 eluates recognized a conformation formed by the W1:1-2 and W2:3-4 loops, 5 recognized W1:2-3, and 4 recognized W3:4-1. The remaining 4 eluates could not be defined by the binding sites. Within these regions, we identified residues critical for binding, including S29 and R32 in W1:1-2; G44 and P45 in W1:2-3; and P135, E136, and R139 in W2:3-4. Of 11 eluates whose recognition sites were identified, 5 clearly showed restricted κ/λ-chain usage. These results suggested that PA anti-αIIbß3 Abs in primary ITP tended to recognize highly restricted regions of αIIb with clonality.


Asunto(s)
Autoanticuerpos/inmunología , Plaquetas/inmunología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/inmunología , Glicoproteína IIb de Membrana Plaquetaria/química , Glicoproteína IIb de Membrana Plaquetaria/inmunología , Trombocitopenia/inmunología , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Epítopos/inmunología , Femenino , Humanos , Cadenas Ligeras de Inmunoglobulina/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
3.
Blood ; 117(1): 250-8, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20940419

RESUMEN

Many different biochemical signaling pathways regulate integrin activation through the integrin cytoplasmic tail. Here, we describe a new role for α-actinin in inside-out integrin activation. In resting human platelets, α-actinin was associated with αIIbß3, whereas inside-out signaling (αIIbß3 activation signals) from protease-activated receptors (PARs) dephosphorylated and dissociated α-actinin from αIIbß3. We evaluated the time-dependent changes of the αIIbß3 activation state by measuring PAC-1 binding velocity. The initial velocity analysis clearly showed that PAR1-activating peptide stimulation induced only transient αIIbß3 activation, whereas PAR4-activating peptide induced long-lasting αIIbß3 activation. When αIIbß3 activation signaling dwindled, α-actinin became rephosphorylated and reassociated with αIIbß3. Compared with control platelets, the dissociation of α-actinin from αIIbß3 was only transient in PAR4-stimulated P2Y(12)-deficient platelets in which the sustained αIIbß3 activation was markedly impaired. Overexpression of wild-type α-actinin, but not the mutant Y12F α-actinin, increased its binding to αIIbß3 and inhibited PAR1-induced initial αIIbß3 activation in the human megakaryoblastic cell line, CMK. In contrast, knockdown of α-actinin augmented PAR-induced αIIbß3 activation in CMK. These observations suggest that α-actinin might play a potential role in setting integrins to a default low-affinity ligand-binding state in resting platelets and regulating αIIbß3 activation by inside-out signaling.


Asunto(s)
Actinina/metabolismo , Plaquetas/metabolismo , Leucemia Megacarioblástica Aguda/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Actinina/genética , Western Blotting , Citometría de Flujo , Humanos , Inmunoprecipitación , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/patología , Fosforilación , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , ARN Mensajero/genética , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Receptores Purinérgicos P2Y12/deficiencia , Receptores Purinérgicos P2Y12/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombastenia/genética , Trombastenia/metabolismo , Trombastenia/patología , Células Tumorales Cultivadas , Tirosina/metabolismo
4.
Blood ; 113(21): 5304-13, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19299337

RESUMEN

Platelet integrin alphaIIbbeta3 activation is tightly controlled by intracellular signaling pathways, and several molecules, including talin, have been identified as critical for alphaIIbbeta3 activation. However, the whole pathway associated with alphaIIbbeta3 activation remains to be determined. To address this issue, we established a Chinese hamster ovary cell line (parental cells) that expresses constitutively activated chimeric integrin alphaIIbalpha6Bbeta3, and then obtained mutant cells expressing inactivated alphaIIbalpha6Bbeta3 by genome-wide mutagenesis. We have performed expression cloning to isolate signaling molecules responsible for integrin activation in the mutant cells. We show that integrin-linked kinase (ILK) complements defective integrin activation in the mutant cells. ILK mRNAs in the mutant cells contained 2 nonsense mutations, R317X and W383X, in a compound heterozygous state, resulting in a complete loss of ILK expression. Moreover, the mutant cells showed partially impaired activation of endogenous beta1 integrins. Knockdown of ILK in parental cells significantly suppressed the activated state of alphaIIbalpha6Bbeta3. However, ILK overexpression did not rescue the impaired integrin activation in talin knocked-down parental cells, whereas overexpression of talin-F3, a subdomain of the talin head domain, restored the function. Our present data suggest that ILK contributes to inside-out integrin activation.


Asunto(s)
Integrinas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células CHO , Codón sin Sentido , Cricetinae , Cricetulus , Integrina beta1/metabolismo , Mutagénesis , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Transducción de Señal , Talina
5.
Biochem Biophys Res Commun ; 400(3): 323-8, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20728432

RESUMEN

Although vinculin is used frequently as a marker for integrin-mediated focal adhesion complexes, how it regulates the activation of integrin is mostly unknown. In this study, we examined whether vinculin would activate integrin in Chinese hamster ovary (CHO) cells expressing human integrin αIIbß3. Silencing of vinculin by lentiviral transduction with a short hairpin RNA sequence affected the binding of PAC-1 (an antibody recognizing activated human αIIbß3) to a constitutively active form of αIIbß3 (α6Bß3) expressed on CHO cells, while its inhibitory effects were much weaker than those of talin-1. Overexpression of an active form of vinculin without intramolecular interactions, but not the full length one, induced PAC-1 binding to native αIIbß3 expressed on CHO cells in a manner dependent on talin-1. On the other hand, silencing of talin-1, but not vinculin, failed to induce cell spreading of α6Bß3-CHO cells on fibrinogen, even in the presence of PT 25-2, a monoclonal antibody that activates αIIbß3. Thus, an active form of vinculin could induce αIIbß3 inside-out signaling through the actions of talin-1, while vinculin was dispensable for outside-in signaling.


Asunto(s)
Cadenas alfa de Integrinas/biosíntesis , Integrina beta3/biosíntesis , Vinculina/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Células CHO , Cricetinae , Cricetulus , Silenciador del Gen , Humanos , Cadenas alfa de Integrinas/inmunología , Integrina beta3/inmunología , Ratones , Talina/genética , Talina/metabolismo , Vinculina/genética
6.
Thromb Res ; 162: 44-52, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29289806

RESUMEN

BACKGROUND: Thrombin belongs to the most potent platelet agonists and activates human platelets through GPIbα and two protease activated receptors (PARs), PAR1 and PAR4. However, the details of thrombin receptor system, especially the role of PAR4 on human platelet activation is still not clear. OBJECTIVES: We found a significant difference in PAR4-activating peptide (PAR4-AP)-induced, but not PAR1-AP, platelet aggregation between healthy Japanese subjects. Sequencing analysis revealed a single nucleotide change in PAR4 gene F2RL3 (SNP rs773902) leading to Ala120Thr variant. To elucidate the role of PAR4 in human platelet activation, we examined if platelet activation induced by PAR4-AP may be associated with PAR4 genotype. METHODS: Platelets from 202 healthy Japanese volunteers were genetically analyzed and determined the genotype frequency of rs773902. Agonist induced platelet aggregation, integrin αIIbß3 activation, granule release, Ca2+ mobilization, and activation of ERK and MLC were evaluated. The specificity of effects observed in platelets was confirmed in 293T cells transfected PAR4-Thr120 or Ala120. RESULTS: The frequencies of PAR4 variant Thr/Thr120, Ala/Thr120, and Ala/Ala 120 were 5.9, 37.1, and 57.0%, respectively. Platelets with Thr/Thr120 showed significantly higher reactivity in PAR4-AP-induced platelet aggregation, αIIbß3 activation and granule release compared to platelets with Ala/Ala120. PAR4-AP induced higher Ca2+ mobilization and ERK activation in platelets with Thr/Thr120 than Ala/Ala120. Ca2+ mobilization and ERK activation were also increased in 293T cells transfected with PAR4-Thr120 compared to Ala120. CONCLUSION: Our data suggested that PAR4-AP-induced platelet reactivity between PAR4 rs773902 was associated with altered intensity of Ca2+ mobilization and ERK activation.


Asunto(s)
Plaquetas/fisiología , Calcio/sangre , Péptidos/farmacología , Activación Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Receptores de Trombina/agonistas , Receptores de Trombina/sangre , Plaquetas/efectos de los fármacos , Plaquetas/enzimología , Plaquetas/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/sangre , Humanos , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Trombina/genética , Receptores de Trombina/metabolismo , Transfección
7.
Thromb Haemost ; 98(4): 783-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17938802

RESUMEN

Inherited deficiency of protein S encoded by the PROS1 gene constitutes an important risk factor for deep vein thrombosis (DVT). Nevertheless, although more than 200 deleterious genetic variations in PROS1 have been identified, causative point mutations of PROS1 gene are not detected in approximately half of protein S-deficient families. The present study investigated whether there may exist a large deletion in PROS1 that constitutes a genetic risk factor for Japanese DVT patients. A multiplex ligation-dependent probe amplification analysis was employed to identify the deletions in PROS1 in 163 Japanese patients with DVT. A large gene deletion was identified in one patient who showed 16% protein S activity and did not carry point mutations in PROS1 by DNA sequencing and it was validated by the quantitative PCR method. The deletion spanned at least the whole PROS1 gene (107 kb) and at most from the centromere located downstream of PROS1, to before the D3S3619 marker, the first heterozygous marker in the upstream of PROS1 in chromosome 3. In conclusion, a large deletion in PROS1 was shown to partly account for DVT with protein S deficiency. Screening for large deletions in PROS1 might be warranted in PROS1 causative point mutation-negative DVT patients with protein S deficiency.


Asunto(s)
Proteínas Sanguíneas/genética , Eliminación de Gen , Deficiencia de Proteína S/genética , Trombosis de la Vena/genética , Adulto , Centrómero/ultraestructura , Femenino , Heterocigoto , Humanos , Japón , Masculino , Repeticiones de Microsatélite , Modelos Genéticos , Mutación Puntual , Proteína S , Factores de Riesgo , Trombosis de la Vena/etnología
8.
Thromb Res ; 120(2): 181-6, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17049586

RESUMEN

The dose required for the anticoagulant effect of warfarin exhibits large inter-individual variations. This study sought to determine the contribution of four genes, vitamin K epoxide reductase (VKORC1), gamma-glutamyl carboxylase (GGCX), calumenin (CALU), and cytochrome P450 2C9 (CYP2C9) to the warfarin maintenance dose required in Japanese patients following ischemic stroke. We recruited 93 patients on stable anticoagulation with a target International Normalized Ratio (INR) of 1.6-2.6. We genotyped eleven representative single nucleotide polymorphisms (SNPs) in the three genes involved in vitamin K cycle and the 42613A>C SNP in CYP2C9, known as CYP2C93, and then examined an association of these genotypes with warfarin maintenance doses (mean+/-SD=2.96+/-1.06 mg/day). We found an association of effective warfarin dose with the -1639G>A (p=0.004) and 3730G>A genotypes (p=0.006) in VKORC1, the 8016G>A genotype in GGCX (p=0.022), and the 42613A>C genotype in CYP2C9 (p=0.015). The model using the multiple regression analysis including age, sex, weight, and three genetic polymorphisms accounted for 33.3% of total variations in warfarin dose. The contribution to inter-individual variation in warfarin dose was 5.9% for VKORC1 -1639G>A, 5.2% for CYP2C9 42613A>C, and 4.6% for GGCX 8016G>A. In addition to polymorphisms in VKORC1 and CYP2C9, we identified GGCX 8016G>A, resulting in the missense mutation R325Q, as a genetic determinant of warfarin maintenance dose in Japanese patients.


Asunto(s)
Anticoagulantes/administración & dosificación , Hidrocarburo de Aril Hidroxilasas/genética , Ligasas de Carbono-Carbono/genética , Oxigenasas de Función Mixta/genética , Warfarina/administración & dosificación , Anciano , Citocromo P-450 CYP2C9 , Femenino , Genotipo , Humanos , Japón , Masculino , Persona de Mediana Edad , Modelos Biológicos , Farmacogenética , Polimorfismo de Nucleótido Simple , Análisis de Regresión , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/genética , Vitamina K Epóxido Reductasas
9.
Arterioscler Thromb Vasc Biol ; 26(1): 224-30, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16269667

RESUMEN

OBJECTIVE: Obesity is a common risk factor in insulin resistance and cardiovascular diseases. Although hypoadiponectinemia is associated with obesity-related metabolic and vascular diseases, the role of adiponectin in thrombosis remains elusive. METHODS AND RESULTS: We investigated platelet thrombus formation in adiponectin knockout (APN-KO) male mice (8 to 12 weeks old) fed on a normal diet. There was no significant difference in platelet counts or coagulation parameters between wild-type (WT) and APN-KO mice. However, APN-KO mice showed an accelerated thrombus formation on carotid arterial injury with a He-Ne laser (total thrombus volume: 13.36+/-4.25 x 10(7) arbitrary units for APN-KO and 6.74+/-2.87x10(7) arbitrary units for WT; n=10; P<0.01). Adenovirus-mediated supplementation of adiponectin attenuated the enhanced thrombus formation. In vitro thrombus formation on a type I collagen at a shear rate of 250 s(-1), as well as platelet aggregation induced by low concentrations of agonists, was enhanced in APN-KO mice, and recombinant adiponectin inhibited the enhanced platelet aggregation. In WT mice, adenovirus-mediated overexpression of adiponectin additionally attenuated thrombus formation. CONCLUSIONS: Adiponectin deficiency leads to enhanced thrombus formation and platelet aggregation. The present study reveals a new role of adiponectin as an endogenous antithrombotic factor.


Asunto(s)
Adiponectina/genética , Adiponectina/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Agregación Plaquetaria/fisiología , Trombosis/metabolismo , Adenoviridae/genética , Adiponectina/deficiencia , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Plaquetas/fisiología , Traumatismos de las Arterias Carótidas/genética , Colágeno , Integrina alfa2/metabolismo , Integrina beta3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/metabolismo , Recuento de Plaquetas , Flujo Pulsátil , Receptores de Adiponectina , Receptores de Superficie Celular/genética , Trombosis/genética
10.
Int J Hematol ; 84(5): 387-97, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17189218

RESUMEN

gamma-Glutamyl carboxylation, a reaction essential for the activity of vitamin K-dependent proteins, requires the concerted actions of gamma-glutamyl carboxylase (GGCX), vitamin K 2, 3-epoxide reductase complex 1 (VKORC1), and the chaperone calumenin (CALU). We evaluated the contribution of genetic polymorphisms in VKORC1, GGCX, and CALU to interindividual variation in the activities of plasma protein C and protein S. We sequenced these 3 genes in 96 Japanese individuals and geno-typed 9 representative single-nucleotide polymorphisms in 3655 Japanese individuals representative of the general population. The mean activity of protein C in women bearing the GG genotype of GGCX 8016G>A (130.8% +/- 1.5%, n = 156) was significantly greater (P = .002) than that of individuals with either the AG (126.8% +/- 0.7%, n = 728) or the AA (125.4% +/- 0.6%, n = 881) genotype, after adjusting for confounding factors. The GGCX 8016G>A change leads to the substitution of Gin for Arg at amino acid residue 325 (Arg 325 Gln). This effect was comparable to that of a previously defined polymorphism in the protein C promoter. Mean protein S activity was influenced by the VKORC1 3730G>A and CALU 20943T>A genotypes, after adjusting for confounding factors. Thus, polymorphisms in genes involved in the vitamin K-dependent gamma-carboxylation reaction influence interindividual variation in the activities of protein C and protein S in the general population.


Asunto(s)
Proteínas de Unión al Calcio/genética , Ligasas de Carbono-Carbono/genética , Oxigenasas de Función Mixta/genética , Polimorfismo de Nucleótido Simple , Proteína C/análisis , Proteína S/análisis , Anciano , Pueblo Asiatico , Femenino , Genética de Población , Humanos , Japón , Masculino , Persona de Mediana Edad , Proteína C/genética , Procesamiento Proteico-Postraduccional/genética , Proteína S/genética , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/genética , Vitamina K/genética , Vitamina K/metabolismo , Vitamina K Epóxido Reductasas
11.
Int J Hematol ; 103(3): 283-91, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26830967

RESUMEN

The congenital form of thrombotic thrombocytopenic purpura (TTP) is caused by genetic mutations in ADAMTS13. Some, but not all, congenital TTP patients manifest renal insufficiency in addition to microangiopathic hemolysis and thrombocytopenia. We included 32 congenital TTP patients in the present study, which was designed to assess whether congenital TTP patients with renal insufficiency have predisposing mutations in complement regulatory genes, as found in many patients with atypical hemolytic uremic syndrome (aHUS). In 13 patients with severe renal insufficiency, six candidate complement or complement regulatory genes were sequenced and 11 missense mutations were identified. One of these missense mutations, C3:p.K155Q mutation, is a rare mutation located in the macroglobulin-like 2 domain of C3, where other mutations predisposing for aHUS cluster. Several of the common missense mutations identified in our study have been reported to increase disease-risk for aHUS, but were not more common in patients with as compared to those without renal insufficiency. Taken together, our results show that the majority of the congenital TTP patients with renal insufficiency studied do not carry rare genetic mutations in complement or complement regulatory genes.


Asunto(s)
Complemento C3/genética , Estudios de Asociación Genética , Mutación Missense , Púrpura Trombocitopénica Trombótica/congénito , Púrpura Trombocitopénica Trombótica/genética , Insuficiencia Renal/etiología , Insuficiencia Renal/genética , Proteína ADAMTS13/genética , Adulto , Síndrome Hemolítico Urémico Atípico/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad
12.
J Thromb Haemost ; 3(4): 763-74, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15842360

RESUMEN

CD47 (integrin-associated protein) serves as a receptor for thrombospondin-1 (TSP-1) and Src homology 2 domain-containing protein tyrosine phosphatase substrate-1 (SHPS-1), and the TSP-1/CD47 interaction has been believed to augment integrin-mediated platelet function. Here, employing SHPS-1-immunoglobulin (Ig) as a ligand, we have newly demonstrated that CD47 acts as an inhibitory receptor for platelet function. The binding of SHPS-1-Ig was solely mediated by CD47, because CD47-deficient platelets failed to bind murine SHPS-1-Ig. The human SHPS-1/CD47 interaction inhibited the platelet aggregation induced by several kinds of agonists at a low concentration. Moreover, human SHPS-1 expressed on the cell surface as well as soluble SHPS-1-Ig markedly inhibited the platelet spreading on, but not initial adhesion to, immobilized fibrinogen. Again, neither murine SHPS-1 expressed on the cell surface nor murine SHPS-1-Ig inhibited the spreading of CD47-deficient platelets. We further investigated the tyrosine phosphorylation of signaling proteins during platelet spreading on immobilized fibrinogen. Unexpectedly, SHPS-1 inhibited alpha(IIb)beta(3)-mediated platelet spreading without disturbing focal adhesion kinase (FAK) tyrosine phosphorylation. Further examination revealed that SHPS-1 inhibited the tyrosine phosphorylation of alpha-actinin, a downstream effector of FAK, but not of cortactin. Thus, it is likely that the SHPS-1/CD47 interaction inhibits alpha(IIb)beta(3)-mediated outside-in signaling by interfering with the downstream pathway of FAK. Taken together, our data suggest that SHPS-1 negatively regulates platelet function via CD47, especially alpha(IIb)beta(3)-mediated outside-in signaling.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Diferenciación/fisiología , Regulación de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores Inmunológicos/fisiología , Actinina/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Plaquetas/metabolismo , Antígeno CD47 , Células CHO , Línea Celular , Membrana Celular/metabolismo , Cortactina , Cricetinae , Relación Dosis-Respuesta a Droga , Fibrinógeno/química , Fibrinógeno/metabolismo , Citometría de Flujo , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Immunoblotting , Inmunoprecipitación , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Fosforilación , Agregación Plaquetaria , Unión Proteica , Receptores Inmunológicos/metabolismo , Transducción de Señal , Tirosina/química
13.
Thromb Haemost ; 92(5): 1092-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15543338

RESUMEN

The crystal structure of alpha(v)beta(3) in complex with a cyclic RGD-containing ligand has recently been demonstrated. However, the functional significance of each residue within ligand binding loops has not been fully elucidated. Here, by employing alanine-scanning mutagenesis, we have examined the functional role of ligand contact residues in alpha(v). Tyr178 --> Ala substitution (Tyr178Ala) and Asp218Ala abolished a monovalent ligand, WOW-1 Fab binding as well as soluble fibrinogen binding, which is in perfect agreement with the crystallography. However, Asp150Ala showed no or only a modest inhibition of ligand binding. In contrast, Tyr substitution at Ala215 (Ala215Tyr) increased WOW-1 Fab binding, suggesting that the substitution increased the integrin affinity. The adhesion assay to immobilized fibrinogen showed essentially the same data as obtained using soluble ligands. Our present data indicate that Tyr178 and Asp218, but not Asp150 in alpha(v) is critically involved in ligand-binding and that Ala215 could regulate the affinity of alpha(v)beta(3).


Asunto(s)
Sustitución de Aminoácidos , Integrina alfaV/genética , Integrina alfaVbeta3/metabolismo , Animales , Sitios de Unión , Fibrinógeno/metabolismo , Fragmentos Fab de Inmunoglobulinas/metabolismo , Integrina alfaV/química , Integrina alfaVbeta3/química , Integrina alfaVbeta3/genética , Ratones , Mutagénesis , Fenotipo , Unión Proteica
14.
PLoS One ; 8(12): e85498, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24376884

RESUMEN

Integrin-linked kinase (ILK) is an important signaling regulator that assembles into the heteroternary complex with adaptor proteins PINCH and parvin (termed the IPP complex). We recently reported that ILK is important for integrin activation in a Chinese hamster ovary (CHO) cell system. We previously established parental CHO cells expressing a constitutively active chimeric integrin (αIIbα6Bß3) and mutant CHO cells expressing inactive αIIbα6Bß3 due to ILK deficiency. In this study, we further investigated the underlying mechanisms for ILK-dependent integrin activation. ILK-deficient mutant cells had trace levels of PINCH and α-parvin, and transfection of ILK cDNA into the mutant cells increased not only ILK but also PINCH and α-parvin, resulting in the restoration of αIIbα6Bß3 activation. In the parental cells expressing active αIIbα6Bß3, ILK, PINCH, and α-parvin were co-immunoprecipitated, indicating the formation of the IPP complex. Moreover, short interfering RNA (siRNA) experiments targeting PINCH-1 or both α- and ß-parvin mRNA in the parent cells impaired the αIIbα6Bß3 activation as well as the expression of the other components of the IPP complex. In addition, ILK mutants possessing defects in either PINCH or parvin binding failed to restore αIIbα6Bß3 activation in the mutant cells. Kindlin-2 siRNA in the parental cells impaired αIIbα6Bß3 activation without disturbing the expression of ILK. For CHO cells stably expressing wild-type αIIbß3 that is an inactive form, overexpression of a talin head domain (THD) induced αIIbß3 activation and the THD-induced αIIbß3 activation was impaired by ILK siRNA through a significant reduction in the expression of the IPP complex. In contrast, overexpression of all IPP components in the αIIbß3-expressing CHO cells further augmented THD-induced αIIbß3 activation, whereas they did not induce αIIbß3 activation without THD. These data suggest that the IPP complex rather than ILK plays an important role and supports integrin activation probably through stabilization of the active conformation.


Asunto(s)
Complejos Multiproteicos/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Citometría de Flujo , Immunoblotting , Inmunoprecipitación , Proteínas con Dominio LIM/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Exp Hematol ; 41(1): 79-90.e1, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23022222

RESUMEN

Platelet integrin α(IIb)ß(3) activation is regulated by inside-out signaling via agonist stimulation. However, when α(IIb)ß(3) was exogenously expressed in cell lines such as Chinese hamster ovarian cells, physiological agonists hardly induced α(IIb)ß(3) activation. To overcome this disadvantage, we characterized the functional regulation of endogenously expressed α(IIb)ß(3) in a megakaryoblastic cell line, CMK, employing an initial velocity assay for PAC-1 binding. We firstly demonstrated that protease-activated receptor 1-activating peptide induced robust, but transient α(IIb)ß(3) activation in CMK cells with high glycoprotein-Ib expression. Stable talin-1 or kindlin-3 knockdown cells confirmed that the protease-activated receptor 1-activating peptide-induced α(IIb)ß(3) activation was dependent on talin-1 and kindlin-3 expression. In sharp contrast to exogenously expressed α(IIb)ß(3) in Chinese hamster ovarian cells, transient overexpression of full-length talin (FL-talin) or talin-head domain (THD) alone did not activate α(IIb)ß(3) in CMK cells, but required agonist stimulation. Similarly, kindlin-3 overexpression alone did not induce α(IIb)ß(3) activation, but it significantly augmented agonist-induced α(IIb)ß(3) activation. Several mutants of FL-talin and THD suggested that the head-rod interaction was critical for autoinhibition of talin-1, and the interaction between the THD and the membrane-proximal region of the ß(3) cytoplasmic tail was essential for talin-mediated α(IIb)ß(3) activation. In addition, THD and kindlin-3 cooperatively augmented protease-activated receptor 1-induced α(IIb)ß(3) activation. We proposed that the CMK cell line is an attractive platform for investigating agonist-, talin-1-, and kindlin-3- dependent α(IIb)ß(3) activation.


Asunto(s)
Células Progenitoras de Megacariocitos/metabolismo , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Talina/fisiología , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Fosfatasa 2 de Especificidad Dual/fisiología , Humanos , Receptor PAR-1/fisiología
16.
Mol Immunol ; 54(2): 238-46, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23314101

RESUMEN

Hemolytic uremic syndrome (HUS) is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and renal impairment. Approximately 10% of cases are classified as atypical due to the absence of Shiga toxin-producing bacteria as a trigger. Uncontrolled activation of the complement system plays a role in the pathogenesis of atypical HUS (aHUS). Although many genetic studies on aHUS have been published in recent years, only limited data has been gathered in Asian countries. We analyzed the genetic variants of 6 candidate genes and the gene deletion in complement factor H (CFH) and CFH-related genes, examined the prevalence of CFH autoantibodies and evaluated the genotype-phenotype relationship in 10 Japanese patients with aHUS. We identified 7 causative or potentially causative mutations in CFH (p.R1215Q), C3 (p.R425C, p.S562L, and p.I1157T), membrane cofactor protein (p.Y189D and p.A359V) and thrombomodulin (p.T500M) in 8 out of 10 patients. All 7 of the mutations were heterozygous and four of them were novel. Two patients carried CFH p.R1215Q and 3 other patients carried C3 p.I1157T. One patient had 2 causative mutations in different genes. One patient was a compound heterozygote of the 2 MCP mutations. The patients carrying mutations in CFH or C3 had a high frequency of relapse and a worse prognosis. One patient had CFH autoantibodies. The present study identified the cause of aHUS in 9 out of 10 Japanese patients. Since the phenotype-genotype correlation of aHUS has clinical significance in predicting renal recovery and transplant outcome, a comprehensively accurate assessment of molecular variation would be necessary for the proper management of aHUS patients in Japan.


Asunto(s)
Pueblo Asiatico/genética , Predisposición Genética a la Enfermedad , Síndrome Hemolítico-Urémico/genética , Adolescente , Síndrome Hemolítico Urémico Atípico , Niño , Preescolar , Femenino , Humanos , Japón , Masculino , Mutación , Linaje , Polimorfismo de Longitud del Fragmento de Restricción , Adulto Joven
17.
Thromb Res ; 124(1): 14-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18954896

RESUMEN

INTRODUCTION: Genetic deficiencies of PROS1, PROC, and SERPINC1 (antithrombin) are risk factors for deep vein thrombosis (DVT). Diagnosis of the inherited deficiencies of these three genes is sometimes difficult because of the phenotypic variability. This study was undertaken to reveal the frequency of nonsynonymous mutations of these three genes in Japanese DVT patients. PATIENTS/METHODS: One hundred seventy-three DVT patients were registered by the Sub-group of Blood Coagulation Abnormality, from the Study Group of Research on Measures for Intractable Diseases. We sequenced the entire coding regions of the three genes in all DNA samples and identified the nonsynonymous mutations. RESULTS AND CONCLUSIONS: For PROS1 we identified 15 nonsynonymous mutations in 28 DVT patients; for PROC, 10 nonsynonymous mutations in 17 patients; and for SERPINC1, 13 nonsynonymous mutations in 14 patients. Five patients had two mutations in PROS1 and PROC, and all of them had PROS1 K196E mutation. We previously identified one patient with a large PROS1 gene deletion. Thus, 55 out of 173 patients (32%) carried at least one genetic defect in the three genes. The PROS1 K196E mutation found in 15 Japanese DVT patients was the most prevalent. Mutations of PROC K193del and V339M were the second, each found in four patients. Our data suggested that the PROC K193del mutation caused the loss of the anticoagulant activity but not the amidolytic activity. Our effort is the first DNA resequencing study to identify the genetic variations in DVT patients without any consideration of their plasma activities and antigens. To minimize selection bias in a future evaluation of the contribution of genetic deficiency to DVT, we must recruit patients consecutively.


Asunto(s)
Antitrombinas/genética , Pueblo Asiatico/genética , Mutación , Proteína C/genética , Proteína S/genética , Adulto , Factores de Edad , Alelos , Secuencia de Bases , Femenino , Frecuencia de los Genes , Heterocigoto , Homocigoto , Humanos , Japón/epidemiología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Prevalencia , Análisis de Secuencia de ADN , Factores Sexuales , Trombosis de la Vena/epidemiología , Trombosis de la Vena/genética
18.
Blood Coagul Fibrinolysis ; 20(1): 22-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20523161

RESUMEN

Tissue factor pathway inhibitor (TFPI) is an anticoagulant protease inhibitor that inhibits the tissue factor-initiated blood coagulation cascade reactions. Based on these anticoagulant functions of TFPI, we hypothesized that genetic variations in TFPI may alter the TFPI expression or impair the anticoagulant function and could predispose persons to deep vein thrombosis (DVT). This study was undertaken to examine whether the genetic polymorphisms in TFPI are associated with the plasma TFPI levels and risk for DVT. We sequenced the entire coding regions of TFPI in 175 Japanese DVT patients and identified 12 genetic variants, including one missense mutation, Asn221Ser. The missense mutation occurred at the site presumably attached to the glycosylphosphatidylinositol anchor in the TFPI-beta form. The allele frequency of the mutant Ser-coding allele of the Asn221Ser mutation was 8% in the Japanese general population consisting of 1684 individuals. The Asn221Ser mutation was significantly associated with the total TFPI levels (Asn/Asn, n = 108, total TFPI = 56.57 +/- 0.88 ng/ml (mean +/- SD) vs. Asn/Ser + Ser/Ser, n = 16, total TFPI = 63.44 +/- 2.28 ng/ml, P = 0.0058). The genotype was not associated with the free TFPI level. This Asn221Ser mutation was not associated with DVT. Thus, the Asn221Ser mutation occurring in the TFPI-beta form was associated with the total TFPI level, but not a risk for DVT. The absence of the putative glycosylphosphatidylinositol anchor in TFPI-beta under pathological conditions remains to be studied.


Asunto(s)
Lipoproteínas/sangre , Lipoproteínas/genética , Mutación Missense/genética , Alelos , Femenino , Variación Genética/genética , Genotipo , Humanos , Japón , Masculino , Polimorfismo de Nucleótido Simple/genética , Trombosis de la Vena/sangre , Trombosis de la Vena/genética
19.
Blood ; 107(8): 3161-6, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16368888

RESUMEN

ADAMTS13 is a plasma metalloproteinase that regulates platelet adhesion and aggregation through cleavage of von Willebrand factor (VWF) multimers. In humans, genetic or acquired deficiency in ADAMTS13 causes thrombotic thrombocytopenic purpura (TTP), a condition characterized by thrombocytopenia and hemolytic anemia with microvascular platelet thrombi. In this study, we report characterization of mice bearing a targeted disruption of the Adamts13 gene. ADAMTS13-deficient mice were born in the expected mendelian distribution; homozygous mice were viable and fertile. Hematologic and histologic analyses failed to detect any evidence of thrombocytopenia, hemolytic anemia, or microvascular thrombosis. However, unusually large VWF multimers were observed in plasma of homozygotes. Thrombus formation on immobilized collagen under flow was significantly elevated in homozygotes in comparison with wild-type mice. Thrombocytopenia was more severely induced in homozygotes than in wild-type mice after intravenous injection of a mixture of collagen and epinephrine. Thus, a complete lack of ADAMTS13 in mice was a prothrombotic state, but it alone was not sufficient to cause TTP-like symptoms. The phenotypic differences of ADAMTS13 deficiencies between humans and mice may reflect differences in hemostatic system functioning in these species. Alternatively, factors in addition to ADAMTS13 deficiency may be necessary for development of TTP.


Asunto(s)
Hemostasis/genética , Metaloendopeptidasas/deficiencia , Adhesividad Plaquetaria/genética , Púrpura Trombocitopénica Trombótica/genética , Proteína ADAMTS13 , Anemia Hemolítica/sangre , Anemia Hemolítica/genética , Anemia Hemolítica/patología , Animales , Predisposición Genética a la Enfermedad/genética , Homocigoto , Humanos , Metaloendopeptidasas/sangre , Ratones , Ratones Noqueados , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/patología , Especificidad de la Especie , Factor de von Willebrand/análisis
20.
Blood ; 99(3): 931-8, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11806996

RESUMEN

Alpha(IIb)beta(3) and alpha(v)beta(3) belong to the beta(3) integrin subfamily. Although the beta(3) subunit is a key regulator for the biosynthesis of beta(3) integrins, it remains obscure whether missense mutations in beta(3) may induce the same defects in both alpha(IIb)beta(3) and alpha(v)beta(3). In this study, it is revealed that thrombasthenic platelets with a His280Pro mutation in beta(3), which is prevalent in Japanese patients with Glanzmann thrombasthenia, did contain significant amounts of alpha(v)beta(3) (about 50% of control) using sensitive enzyme-linked immunosorbent assay. Expression studies showed that the His280Probeta(3) mutation impaired alpha(IIb)beta(3) expression but not alpha(v)beta(3) expression in 293 cells. To extend these findings, the effects of several beta(3) missense mutations leading to an impaired alpha(IIb)beta(3) expression on alpha(v)beta(3) function as well as expression was examined: Leu117Trp, Ser162Leu, Arg216Gln, Cys374Tyr, and a newly created Arg216Gln/Leu292Ser mutation. Leu117Trp and Cys374Tyr beta(3) mutations did impair alpha(v)beta(3) expression, while Ser162Leu, Arg216Gln, and Arg216Gln/Leu292Ser mutations did not. With regard to ligand binding function, Ser162Leu mutation induced especially distinct effects between 2 beta(3) integrins: it markedly impaired ligand binding to alpha(IIb)beta(3) but not to alpha(v)beta(3) at all. These data clearly demonstrate that the biosynthesis and the ligand binding function of alpha(IIb)beta(3) and those of alpha(v)beta(3) are regulated in part by different mechanisms. Present data would be a clue to elucidate the regulatory mechanism of expression and function of beta(3) integrins.


Asunto(s)
Antígenos CD/genética , Mutación Missense , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/genética , Receptores de Vitronectina/genética , Adulto , Secuencia de Aminoácidos , Antígenos CD/biosíntesis , Antígenos CD/metabolismo , Plaquetas/química , Plaquetas/metabolismo , Plaquetas/patología , Línea Celular , Análisis Mutacional de ADN , Femenino , Fibrinógeno/metabolismo , Expresión Génica , Humanos , Integrina beta3 , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación Missense/genética , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Receptores de Vitronectina/metabolismo , Trombastenia/sangre , Trombastenia/genética , Trombastenia/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA