Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Blood ; 138(6): 480-485, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34010413

RESUMEN

Congenital amegakaryocytic thrombocytopenia (CAMT) is a severe inherited thrombocytopenia due to loss-of-function mutations affecting the thrombopoietin (TPO) receptor, MPL. Here, we report a new homozygous MPL variant responsible for CAMT in 1 consanguineous family. The propositus and her sister presented with severe thrombocytopenia associated with mild anemia. Next-generation sequencing revealed the presence of a homozygous MPLR464G mutation resulting in a weak cell-surface expression of the receptor in platelets. In cell lines, we observed a defect in MPLR464G maturation associated with its retention in the endoplasmic reticulum. The low cell-surface expression of MPLR464G induced very limited signaling with TPO stimulation, leading to survival and reduced proliferation of cells. Overexpression of a myeloproliferative neoplasm-associated calreticulin (CALR) mutant did not rescue trafficking of MPLR464G to the cell surface and did not induce constitutive signaling. However, it unexpectedly restored a normal response to eltrombopag (ELT), but not to TPO. This effect was only partially mimicked by the purified recombinant CALR mutant protein. Finally, the endogenous CALR mutant was able to restore the megakaryocyte differentiation of patient CD34+ cells carrying MPLR464G in response to ELT.


Asunto(s)
Benzoatos/farmacología , Calreticulina , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Hidrazinas/farmacología , Mutación Missense , Pirazoles/farmacología , Receptores de Trombopoyetina , Trombocitopenia , Adulto , Sustitución de Aminoácidos , Calreticulina/genética , Calreticulina/metabolismo , Niño , Preescolar , Síndromes Congénitos de Insuficiencia de la Médula Ósea/tratamiento farmacológico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Femenino , Células HEK293 , Homocigoto , Humanos , Lactante , Masculino , Receptores de Trombopoyetina/genética , Receptores de Trombopoyetina/metabolismo , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patología
2.
Haematologica ; 108(8): 2130-2145, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36794499

RESUMEN

Sustained ANKRD26 expression associated with germline ANKRD26 mutations causes thrombocytopenia 2 (THC2), an inherited platelet disorder associated with a predisposition to leukemia. Some patients also present with erythrocytosis and/or leukocytosis. Using multiple human-relevant in vitro models (cell lines, primary patients' cells and patient-derived induced pluripotent stem cells) we demonstrate for the first time that ANKRD26 is expressed during the early steps of erythroid, megakaryocyte and granulocyte differentiation, and is necessary for progenitor cell proliferation. As differentiation progresses, ANKRD26 expression is progressively silenced, to complete the cellular maturation of the three myeloid lineages. In primary cells, abnormal ANKRD26 expression in committed progenitors directly affects the proliferation/differentiation balance for the three cell types. We show that ANKRD26 interacts with and crucially modulates the activity of MPL, EPOR and G-CSFR, three homodimeric type I cytokine receptors that regulate blood cell production. Higher than normal levels of ANKRD26 prevent the receptor internalization that leads to increased signaling and cytokine hypersensitivity. These findings afford evidence how ANKRD26 overexpression or the absence of its silencing during differentiation is responsible for myeloid blood cell abnormalities in patients with THC2.


Asunto(s)
Leucemia , Receptores de Citocinas , Humanos , Citocinas , Hematopoyesis , Leucemia/patología , Diferenciación Celular , Péptidos y Proteínas de Señalización Intercelular
3.
Blood ; 133(16): 1778-1788, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30602618

RESUMEN

Filamin A (FLNa) links the cell membrane with the cytoskeleton and is central in several cellular processes. Heterozygous mutations in the X-linked FLNA gene are associated with a large spectrum of conditions, including macrothrombocytopenia, called filaminopathies. Using an isogenic pluripotent stem cell model derived from patients, we show that the absence of the FLNa protein in megakaryocytes (MKs) leads to their incomplete maturation, particularly the inability to produce proplatelets. Reduction in proplatelet formation potential is associated with a defect in actomyosin contractility, which results from inappropriate RhoA activation. This dysregulated RhoA activation was observed when MKs were plated on fibrinogen but not on other matrices (fibronectin, vitronectin, collagen 1, and von Willebrand factor), strongly suggesting a role for FLNa/αIIbß3 interaction in the downregulation of RhoA activity. This was confirmed by experiments based on the overexpression of FLNa mutants deleted in the αIIbß3-binding domain and the RhoA-interacting domain, respectively. Finally, pharmacological inhibition of the RhoA-associated kinase ROCK1/2 restored a normal phenotype and proplatelet formation. Overall, this work suggests a new etiology for macrothrombocytopenia, in which increased RhoA activity is associated with disrupted FLNa/αIIbß3 interaction.


Asunto(s)
Filaminas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Trombocitopenia/etiología , Femenino , Fibrinógeno/metabolismo , Filaminas/genética , Humanos , Megacariocitos/química , Megacariocitos/patología , Mutación , Unión Proteica/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo
4.
Haematologica ; 104(6): 1244-1255, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30545930

RESUMEN

Germline RUNX1 mutations lead to thrombocytopenia and platelet dysfunction in familial platelet disorder with predisposition to acute myelogenous leukemia (AML). Multiple aspects of platelet function are impaired in these patients, associated with altered expression of genes regulated by RUNX1 We aimed to identify RUNX1-targets involved in platelet function by combining transcriptome analysis of patient and shRUNX1-transduced megakaryocytes (MK). Down-regulated genes included TREM-like transcript (TLT)-1 (TREML1) and the integrin subunit alpha (α)-2 (ITGA2) of collagen receptor α2-beta (ß)-1, which are involved in platelet aggregation and adhesion, respectively. RUNX1 binding to regions enriched for H3K27Ac marks was demonstrated for both genes using chromatin immunoprecipitation. Cloning of these regions upstream of the respective promoters in lentivirus allowing mCherry reporter expression showed that RUNX1 positively regulates TREML1 and ITGA2, and this regulation was abrogated after deletion of RUNX1 sites. TLT-1 content was reduced in patient MK and platelets. A blocking anti-TLT-1 antibody was able to block aggregation of normal but not patient platelets, whereas recombinant soluble TLT-1 potentiated fibrinogen binding to patient platelets, pointing to a role for TLT-1 deficiency in the platelet function defect. Low levels of α2 integrin subunit were demonstrated in patient platelets and MK, coupled with reduced platelet and MK adhesion to collagen, both under static and flow conditions. In conclusion, we show that gene expression profiling of RUNX1 knock-down or mutated MK provides a suitable approach to identify novel RUNX1 targets, among which downregulation of TREML1 and ITGA2 clearly contribute to the platelet phenotype of familial platelet disorder with predisposition to AML.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica , Integrina alfa2/genética , Leucemia Mieloide Aguda/etiología , Receptores Inmunológicos/genética , Trastornos de las Plaquetas Sanguíneas/sangre , Plaquetas/metabolismo , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Megacariocitos/metabolismo , Mutación , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Unión Proteica
5.
Blood ; 125(6): 930-40, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25490895

RESUMEN

To explore how RUNX1 mutations predispose to leukemia, we generated induced pluripotent stem cells (iPSCs) from 2 pedigrees with germline RUNX1 mutations. The first, carrying a missense R174Q mutation, which acts as a dominant-negative mutant, is associated with thrombocytopenia and leukemia, and the second, carrying a monoallelic gene deletion inducing a haploinsufficiency, presents only as thrombocytopenia. Hematopoietic differentiation of these iPSC clones demonstrated profound defects in erythropoiesis and megakaryopoiesis and deregulated expression of RUNX1 targets. iPSC clones from patients with the R174Q mutation specifically generated an increased amount of granulomonocytes, a phenotype reproduced by an 80% RUNX1 knockdown in the H9 human embryonic stem cell line, and a genomic instability. This phenotype, found only with a lower dosage of RUNX1, may account for development of leukemia in patients. Altogether, RUNX1 dosage could explain the differential phenotype according to RUNX1 mutations, with a haploinsufficiency leading to thrombocytopenia alone in a majority of cases whereas a more complete gene deletion predisposes to leukemia.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Hematopoyesis , Leucemia/genética , Mutación , Trombocitopenia/genética , Línea Celular , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Eliminación de Gen , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Leucemia/metabolismo , Leucemia/patología , Datos de Secuencia Molecular , Mutación Missense , Trombocitopenia/metabolismo , Trombocitopenia/patología
6.
Blood ; 120(13): 2708-18, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22898599

RESUMEN

FPD/AML is a familial platelet disorder characterized by platelet defects, predisposition to acute myelogenous leukemia (AML) and germ-line heterozygous RUNX1 alterations. Here we studied the in vitro megakaryopoiesis of 3 FPD/AML pedigrees. A 60% to 80% decrease in the output of megakaryocytes (MKs) from CD34(+) was observed. MK ploidy level was low and mature MKs displayed a major defect in proplatelet formation. To explain these defects, we focused on myosin II expression as RUNX1 has been shown to regulate MYL9 and MYH10 in an inverse way. In FPD/AML MKs, expression of MYL9 and MYH9 was decreased, whereas MYH10 expression was increased and the MYH10 protein was still present in the cytoplasm of mature MKs. Myosin II activity inhibition by blebbistatin rescued the ploidy defect of FPD/AML MKs. Finally, we demonstrate that MYH9 is a direct target of RUNX1 by chromatin immunoprecipitation and luciferase assays and we identified new RUNX1 binding sites in the MYL9 promoter region. Together, these results demonstrate that the defects in megakaryopoiesis observed in FPD/AML are, in part, related to a deregulation of myosin IIA and IIB expression leading to both a defect in ploidization and proplatelet formation.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Leucemia Mieloide Aguda/patología , Megacariocitos/patología , Mutación/genética , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Luciferasas/metabolismo , Masculino , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIB no Muscular/genética , Linaje , Ploidias , Pronóstico , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Clin Invest ; 124(2): 580-91, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24430186

RESUMEN

Point mutations in the 5' UTR of ankyrin repeat domain 26 (ANKRD26) are associated with familial thrombocytopenia 2 (THC2) and a predisposition to leukemia. Here, we identified underlying mechanisms of ANKRD26-associated thrombocytopenia. Using megakaryocytes (MK) isolated from THC2 patients and healthy subjects, we demonstrated that THC2-associated mutations in the 5' UTR of ANKRD26 resulted in loss of runt-related transcription factor 1 (RUNX1) and friend leukemia integration 1 transcription factor (FLI1) binding. RUNX1 and FLI1 binding at the 5' UTR from healthy subjects led to ANKRD26 silencing during the late stages of megakaryopoiesis and blood platelet development. We showed that persistent ANKRD26 expression in isolated MKs increased signaling via the thrombopoietin/myeloproliferative leukemia virus oncogene (MPL) pathway and impaired proplatelet formation by MKs. Importantly, we demonstrated that ERK inhibition completely rescued the in vitro proplatelet formation defect. Our data identify a mechanism for development of the familial thrombocytopenia THC2 that is related to abnormal MAPK signaling.


Asunto(s)
Trastornos de los Cromosomas/genética , Sistema de Señalización de MAP Quinasas , Mutación , Proteínas Nucleares/genética , Trombocitopenia/congénito , Regiones no Traducidas 5' , Adolescente , Adulto , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Rotura Cromosómica , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Activación Enzimática , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , Lactante , Péptidos y Proteínas de Señalización Intercelular , Masculino , Megacariocitos/citología , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Proteína Proto-Oncogénica c-fli-1/metabolismo , Receptores de Trombopoyetina/metabolismo , Transducción de Señal , Trombocitopenia/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA