Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 141(3): 260-270, 2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36219879

RESUMEN

Heterozygous defects in runt-related transcription factor 1 (RUNX1) are causative of a familial platelet disorder with associated myeloid malignancy (FPDMM). Because RUNX1-deficient animal models do not mimic bleeding disorder or leukemic risk associated with FPDMM, development of a proper model system is critical to understanding the underlying mechanisms of the observed phenotype and to identifying therapeutic interventions. We previously reported an in vitro megakaryopoiesis system comprising human CD34+ hematopoietic stem and progenitor cells that recapitulated the FPDMM quantitative megakaryocyte defect through a decrease in RUNX1 expression via a lentiviral short hairpin RNA strategy. We now show that shRX-megakaryocytes have a marked reduction in agonist responsiveness. We then infused shRX-megakaryocytes into immunocompromised NOD scid gamma (NSG) mice and demonstrated that these megakaryocytes released fewer platelets than megakaryocytes transfected with a nontargeting shRNA, and these platelets had a diminished half-life. The platelets were also poorly responsive to agonists, unable to correct thrombus formation in NSG mice homozygous for a R1326H mutation in von Willebrand Factor (VWFR1326H), which switches the species-binding specificity of the VWF from mouse to human glycoprotein Ibα. A small-molecule inhibitor RepSox, which blocks the transforming growth factor ß1 (TGFß1) pathway and rescued defective megakaryopoiesis in vitro, corrected the thrombopoietic defect, defects in thrombus formation and platelet half-life, and agonist response in NSG/VWFR1326H mice. Thus, this model recapitulates the defects in FPDMM megakaryocytes and platelets, identifies previously unrecognized defects in thrombopoiesis and platelet half-life, and demonstrates for the first time, reversal of RUNX1 deficiency-induced hemostatic defects by a drug.


Asunto(s)
Megacariocitos , Trombopoyesis , Humanos , Ratones , Animales , Megacariocitos/metabolismo , Trombopoyesis/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Semivida , Plaquetas/metabolismo
2.
Blood ; 132(7): 727-734, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-29914979

RESUMEN

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder initiated by antibodies to platelet factor 4 (PF4)/heparin complexes. PF4 released from platelets binds to surface glycosaminoglycans on hematopoietic and vascular cells that are heterogenous in composition and differ in affinity for PF4. PF4 binds to monocytes with higher affinity than to platelets, and depletion of monocytes exacerbates thrombocytopenia in a murine HIT model. Here we show that the expression of PF4 on platelets and development of thrombocytopenia are modulated by the (re)distribution of PF4 among hematopoietic and endothelial cell surfaces. Binding of PF4 to platelets in whole blood in vitro varies inversely with the white cell count, likely because of the greater affinity of monocytes for PF4. In mice, monocyte depletion increased binding of PF4 to platelets by two- to three-fold. Induction of HIT in mice caused a transient >80-fold increase in binding of HIT antibody to monocytes vs 3.5-fold increase to platelets and rapid transient monocytopenia. Normalization of monocyte counts preceded the return in platelet counts. Exposure of blood to endothelial cells also depletes PF4 from platelet surfaces. These studies demonstrate a dynamic interchange of surface-bound PF4 among hematopoetic and vascular cells that may limit thrombocytopenia at the expense of promoting prothrombotic processes in HIT.


Asunto(s)
Antígenos/biosíntesis , Plaquetas/metabolismo , Heparina/efectos adversos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Monocitos/metabolismo , Factor Plaquetario 4/biosíntesis , Trombocitopenia/metabolismo , Animales , Plaquetas/patología , Regulación de la Expresión Génica , Heparina/uso terapéutico , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Ratones , Monocitos/patología , Trombocitopenia/inducido químicamente , Trombocitopenia/patología
3.
Blood ; 127(4): 464-72, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26518435

RESUMEN

Heparin-induced thrombocytopenia (HIT) is characterized by a high incidence of thrombosis, unlike other antibody-mediated causes of thrombocytopenia. We have shown that monocytes complexed with surface-bound platelet factor 4 (PF4) activated by HIT antibodies contribute to the prothrombotic state in vivo, but the mechanism by which this occurs and the relationship to the requirement for platelet activation via fragment crystallizable (Fc)γRIIA is uncertain. Using a microfluidic model and human or murine blood, we confirmed that activation of monocytes contributes to the prothrombotic state in HIT and showed that HIT antibodies bind to monocyte FcγRIIA, which activates spleen tyrosine kinase and leads to the generation of tissue factor (TF) and thrombin. The combination of direct platelet activation by HIT immune complexes through FcγRIIA and transactivation by monocyte-derived thrombin markedly increases Annexin V and factor Xa binding to platelets, consistent with the formation of procoagulant coated platelets. These data provide a model of HIT wherein a combination of direct FcγRIIA-mediated platelet activation and monocyte-derived thrombin contributes to thrombosis in HIT and identifies potential new targets for lessening this risk.


Asunto(s)
Anticoagulantes/efectos adversos , Plaquetas/inmunología , Heparina/efectos adversos , Monocitos/inmunología , Trombocitopenia/inducido químicamente , Trombocitopenia/complicaciones , Trombosis/etiología , Animales , Anticoagulantes/inmunología , Células Cultivadas , Heparina/inmunología , Humanos , Ratones , Técnicas Analíticas Microfluídicas , Activación Plaquetaria , Factor Plaquetario 4/inmunología , Receptores de IgG/inmunología , Trombocitopenia/inmunología , Trombosis/inmunología
4.
J Cell Mol Med ; 18(6): 1028-34, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24708439

RESUMEN

Recently, we reported that human amniotic membrane-derived mesenchymal stem cells (AMMs) possess great angiogenic potential. In this study, we determined whether local injection of AMMs ameliorates peripheral neuropathy. AMMs were transplanted into injured sciatic nerves. AMM injection promoted significant recovery of motor nerve conduction velocity and voltage amplitude compared to human adipose-derived mesenchymal stem cells. AMM implantation also augmented blood perfusion and increased intraneural vascularity. Whole-mount fluorescent imaging analysis demonstrated that AMMs exhibited higher engraftment and endothelial incorporation abilities in the sciatic nerve. In addition, the higher expression of pro-angiogenic factors was detected in AMMs injected into the peripheral nerve. Therefore, these data provide novel therapeutic and mechanistic insights into stem cell biology, and AMM transplantation may represent an alternative therapeutic option for treating peripheral neuropathy.


Asunto(s)
Amnios/citología , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , Regeneración Nerviosa/fisiología , Nervio Ciático/fisiopatología , Tropismo , Inductores de la Angiogénesis/metabolismo , Animales , Conducta Animal , Western Blotting , Células Cultivadas , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monitorización Neurofisiológica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/lesiones
5.
Nat Commun ; 11(1): 398, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31964886

RESUMEN

A prevailing dogma is that inhibition of vascular thrombosis by antagonizing platelet integrin αIIbß3 cannot be achieved without compromising hemostasis, thus causing serious bleeding and increased morbidity and mortality. It is speculated that these adverse outcomes result from drug-induced activating conformational changes in αIIbß3 but direct proof is lacking. Here, we report the structure-guided design of peptide Hr10 and a modified form of the partial agonist drug tirofiban that act as "pure" antagonists of αIIbß3, i.e., they no longer induce the conformational changes in αIIbß3. Both agents inhibit human platelet aggregation but preserve clot retraction. Hr10 and modified tirofiban are as effective as partial agonist drugs in inhibiting vascular thrombosis in humanized mice, but neither causes serious bleeding, establishing a causal link between partial agonism and impaired hemostasis. Pure orthosteric inhibitors of αIIbß3 may thus provide safer alternatives for human therapy, and valuable tools to probe structure-activity relationships in integrins.


Asunto(s)
Diseño de Fármacos , Hemorragia/tratamiento farmacológico , Péptidos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/antagonistas & inhibidores , Trombosis/prevención & control , Animales , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Voluntarios Sanos , Humanos , Células K562 , Masculino , Ratones , Ratones Transgénicos , Péptidos/química , Péptidos/uso terapéutico , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/química , Inhibidores de Agregación Plaquetaria/uso terapéutico , Pruebas de Función Plaquetaria , Relación Estructura-Actividad , Tirofibán/química , Tirofibán/uso terapéutico , Factor de von Willebrand/genética
6.
Blood Adv ; 3(9): 1368-1378, 2019 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-31036722

RESUMEN

B-domainless factor VIII (FVIII) ectopically expressed in megakaryocytes (MKs) is stored in α granules of platelets (pFVIII) and is capable of restoring hemostasis in FVIIInull mice, even in the presence of circulating inhibitors. However, our prior studies have shown that this ectopically expressed pFVIII can injure developing MKs. Moreover, the known risks of prolonged thrombocytopenia after bone marrow transplantation are significant challenges to the use of this strategy to treat individuals with severe hemophilia A and particularly those with intractable clinically relevant inhibitors. Because of these limitations, we now propose the alternative therapeutic pFVIII strategy of infusing pFVIII-expressing MKs or platelets derived from induced pluripotent stem cells (iPSCs). pFVIII-expressing iPSC-derived MKs, termed iMKs, release platelets that can contribute to improved hemostasis in problematic inhibitor patients with hemophilia A. As proof of principle, we demonstrate that hemostasis can be achieved in vitro and in vivo with pFVIII-expressing platelets and show prolonged efficacy. Notably, pFVIII-expressing platelets are also effective in the presence of inhibitors, and their effect was enhanced with recombinant FVIIa. Human pFVIII-expressing iMKs improved hemostasis in vitro, and derived platelets from infused human pFVIII-expressing iMKs improved hemostasis in FVIIInull mice. These studies indicate the potential therapeutic use of recurrent pFVIII-expressing MK or platelet infusions with prolonged hemostatic coverage that may be additive with bypassing agents in hemophilia A patients with neutralizing inhibitors.


Asunto(s)
Factor VIII/genética , Hemofilia A/terapia , Megacariocitos/trasplante , Transfusión de Plaquetas , Animales , Área Bajo la Curva , Plaquetas/citología , Plaquetas/metabolismo , Factor VIII/análisis , Factor VIII/metabolismo , Factor VIIa/uso terapéutico , Hemofilia A/mortalidad , Humanos , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Curva ROC , Tasa de Supervivencia , Resultado del Tratamiento
7.
J Clin Invest ; 126(2): 483-94, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26690701

RESUMEN

The use of fibrinolytic agents to prevent new thrombus formation is limited by an increased risk of bleeding due to lysis of hemostatic clots that prevent hemorrhage in damaged blood vessels. We sought to develop an agent that provides thromboprophylaxis without carrying a significant risk of causing systemic fibrinolysis or disrupting hemostatic clots. We previously showed that platelet (PLT) α granule-delivered urokinase plasminogen activator (uPA) is highly effective in preventing thrombosis, while being associated with little systemic fibrinolysis or bleeding. Here, we generated a chimeric prodrug composed of a single-chain version of the variable region of an anti-αIIbß3 mAb fused to a thrombin-activatable, low-molecular-weight pro-uPA (PLT/uPA-T). PLT/uPA-T recognizes human αIIbß3 on both quiescent and activated platelets and is enzymatically activated specifically by thrombin. We found that this prodrug binds tightly to human platelets even after gel filtration, has a prolonged half-life in mice transgenic for human αIIb compared with that of uPA-T, and prevents clot formation in a microfluidic system. Importantly, in two murine injury models, PLT/uPA-T did not lyse preexisting clots, even when administration was delayed by as little as 10 minutes, while it concurrently prevented the development of nascent thrombi. Thus, PLT/uPA-T represents the prototype of a platelet-targeted thromboprophylactic agent that selectively targets nascent over preexisting thrombi.


Asunto(s)
Plaquetas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Profármacos/farmacología , Trombosis/tratamiento farmacológico , Activador de Plasminógeno de Tipo Uroquinasa/farmacología , Animales , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Profármacos/farmacocinética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología , Trombosis/sangre , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/farmacocinética
8.
Korean J Lab Med ; 30(1): 84-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20197728

RESUMEN

Deletions of chromosome 6q, particularly in the proximal region, are relatively rare. Here, we report on a de novo interstitial deletion of (6)(q13q16.2) in a girl with facial dysmorphism, congenital hip dislocation, porencephaly, and brain atrophy. Array comparative genomic hybridization analysis showed arr 6q13q16.2(73,378,824?99,824,130), demonstrating higher resolution than the conventional cytogenetic findings, del(6)(q12q15). The clinical data were analyzed and compared with those of similar patients previously reported in the literature.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 6 , Hibridación Genómica Comparativa/métodos , Femenino , Humanos , Recién Nacido , Cariotipificación , Análisis de Secuencia por Matrices de Oligonucleótidos
9.
Pediatr Allergy Immunol ; 18(1): 63-70, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17295801

RESUMEN

Food is closely associated with the pathogenesis of atopic dermatitis. Food allergy is usually mediated by IgE antibody to specific food proteins and determination of specific IgE antibody is the basis of the common diagnostic test for food allergy. IgG4 have been reported as blocking antibody and the protective effects of blocking antibody may be clear in inhalant allergy. However, the role of IgG4 in food allergy is still a matter of debate. In this study, the clinical significance of food allergen-specific IgE/IgG4 in atopic dermatitis was investigated and compared with that of IgE. A total of 97 patients who fulfilled the diagnostic criteria for atopic dermatitis participated in this study. Skin prick test and allergy patch test were performed. Specific IgE and IgG4 concentration were measured using allergy protein chip, 'AllergyChip'. Double blinded placebo controlled food challenge test (DBPCFC) was performed for the diagnosis of allergy to milk, egg white, wheat, and soybean. DBPCFCs for milk, egg white, soybean, and wheat were performed. The positive rates were 31.7% (19/60) in milk, 36.7% (18/49) in egg white, 30.4% (7/23) in soybean, and 34.8% (8/23) in wheat. Mean IgE/IgG4 levels in DBPCFC (+) subjects is higher than those in DBPCFC (-) subjects in all food items studied. Of them, there were significantly different between two groups in egg white and wheat (Egg white in DBPCFC (+) vs. (-): 0.4 +/- 0.3 vs. 0.2 +/- 0.2, wheat in DBPCFC (+) vs. (-): 1.2 +/- 1.2 vs. 0.3 +/- 0.3, p < 0.05). Allergen-specific IgE/IgG4 may provide one of the clues to understand the mechanism of food allergy in atopic dermatitis. The present study suggests that protein microarray can be one of the useful methods to assess ongoing status of allergic diseases.


Asunto(s)
Dermatitis Atópica/inmunología , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Análisis por Matrices de Proteínas/métodos , Adolescente , Adulto , Alérgenos/administración & dosificación , Alérgenos/inmunología , Niño , Preescolar , Dermatitis Atópica/diagnóstico , Dermatitis Atópica/etiología , Método Doble Ciego , Femenino , Hipersensibilidad a los Alimentos/diagnóstico , Hipersensibilidad a los Alimentos/etiología , Humanos , Masculino , Pruebas Cutáneas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA