Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(7): e270-e278, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37128917

RESUMEN

BACKGROUND: Autosomal dominant hypercholesterolemia (ADH) is due to deleterious variants in LDLR, APOB, or PCSK9 genes. Double heterozygote for these genes induces a more severe phenotype. More recently, a new causative variant of heterozygous ADH was identified in APOE. Here we study the phenotype of 21 adult patients, double heterozygotes for rare LDLR and rare APOE variants (LDLR+APOE) in a national wide French cohort. METHODS: LDLR, APOB, PCSK9, and APOE genes were sequenced in 5743 probands addressed for ADH genotyping. The lipid profile and occurrence of premature atherosclerotic cardiovascular diseases were compared between the LDLR+APOE carriers (n=21) and the carriers of the same LDLR causative variants alone (n=22). RESULTS: The prevalence of LDLR+APOE carriers in this French ADH cohort is 0.4%. Overall, LDL (low-density lipoprotein)-cholesterol concentrations were 23% higher in LDLR+APOE patients than in LDLR patients (9.14±2.51 versus 7.43±1.59 mmol/L, P=0.0221). When only deleterious or probably deleterious variants were considered, the LDL-cholesterol concentrations were 46% higher in LDLR+APOE carriers than in LDLR carriers (10.83±3.45 versus 7.43±1.59 mmol/L, P=0.0270). Two patients exhibited a homozygous familial hypercholesterolemia phenotype (LDL-cholesterol >13 mmol/L). Premature atherosclerotic cardiovascular disease was more common in LDLR+APOE patients than in LDLR carriers (70% versus 30%, P=0.026). CONCLUSIONS: Although an incomplete penetrance should be taken into account for APOE variant classification, these results suggest an additive effect of deleterious APOE variants on ADH phenotype highlighting the relevance of APOE sequencing.


Asunto(s)
Aterosclerosis , Hiperlipoproteinemia Tipo II , Humanos , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/genética , LDL-Colesterol , Fenotipo , Aterosclerosis/epidemiología , Aterosclerosis/genética , Apolipoproteínas B/genética , Apolipoproteínas E/genética , Mutación , Heterocigoto
2.
Cell Stem Cell ; 30(2): 153-170.e9, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36736290

RESUMEN

Fanconi anemia (FA) patients experience chromosome instability, yielding hematopoietic stem/progenitor cell (HSPC) exhaustion and predisposition to poor-prognosis myeloid leukemia. Based on a longitudinal cohort of 335 patients, we performed clinical, genomic, and functional studies in 62 patients with clonal evolution. We found a unique pattern of somatic structural variants and mutations that shares features of BRCA-related cancers, the FA-hallmark being unbalanced, microhomology-mediated translocations driving copy-number alterations. Half the patients developed chromosome 1q gain, driving clonal hematopoiesis through MDM4 trisomy downmodulating p53 signaling later followed by secondary acute myeloid lukemia genomic alterations. Functionally, MDM4 triplication conferred greater fitness to murine and human primary FA HSPCs, rescued inflammation-mediated bone marrow failure, and drove clonal dominance in FA mouse models, while targeting MDM4 impaired leukemia cells in vitro and in vivo. Our results identify a linear route toward secondary leukemogenesis whereby early MDM4-driven downregulation of basal p53 activation plays a pivotal role, opening monitoring and therapeutic prospects.


Asunto(s)
Anemia de Fanconi , Leucemia , Humanos , Ratones , Animales , Anemia de Fanconi/genética , Hematopoyesis Clonal , Trisomía/genética , Proteína p53 Supresora de Tumor/genética , Leucemia/genética , Cromosomas , Hematopoyesis/genética , Proteínas Proto-Oncogénicas/genética , Proteínas de Ciclo Celular/genética
3.
Int J Mol Sci ; 23(10)2022 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-35628605

RESUMEN

Primary hypercholesterolemia is characterized by elevated LDL-cholesterol (LDL-C) levels isolated in autosomal dominant hypercholesterolemia (ADH) or associated with elevated triglyceride levels in familial combined hyperlipidemia (FCHL). Rare APOE variants are known in ADH and FCHL. We explored the APOE molecular spectrum in a French ADH/FCHL cohort of 5743 unrelated probands. The sequencing of LDLR, PCSK9, APOB, and APOE revealed 76 carriers of a rare APOE variant, with no mutation in LDLR, PCSK9, or APOB. Among the 31 APOE variants identified here, 15 are described in ADH, 10 in FCHL, and 6 in both probands. Five were previously reported with dyslipidemia and 26 are novel, including 12 missense, 5 synonymous, 2 intronic, and 7 variants in regulatory regions. Sixteen variants were predicted as pathogenic or likely pathogenic, and their carriers had significantly lower polygenic risk scores (wPRS) than carriers of predicted benign variants. We observed no correlation between LDL-C levels and wPRS, suggesting a major effect of APOE variants. Carriers of p.Leu167del were associated with a severe phenotype. The analysis of 11 probands suggests that carriers of an APOE variant respond better to statins than carriers of a LDLR mutation. Altogether, we show that the APOE variants account for a significant contribution to ADH and FCHL.


Asunto(s)
Apolipoproteínas E , Hiperlipoproteinemia Tipo II , Proproteína Convertasa 9 , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo
4.
Oncogene ; 39(31): 5323-5337, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32572159

RESUMEN

Mutations of calreticulin (CALRm) define a subtype of myeloproliferative neoplasms (MPN). We studied the biological and genetic features of CALR-mutated essential thrombocythemia and myelofibrosis patients. In most cases, CALRm were found in granulocytes, monocytes, B and NK cells, but also in T cells. However, the type 1 CALRm spreads more easily than the type 2 CALRm in lymphoid cells. The CALRm were also associated with an early clonal dominance at the level of hematopoietic stem and progenitor cells (HSPC) with no significant increase during granulo/monocytic differentiation in most cases. Moreover, we found that half of type 2 CALRm patients harbors some homozygous progenitors. Those patients were associated with a higher clonal dominance during granulo/monocytic differentiation than patients with only heterozygous type 2 CALRm progenitors. When associated mutations were present, CALRm were the first genetic event suggesting that they are both the initiating and phenotypic event. In blood, type 1 CALRm led to a greater increased number of all types of progenitors compared with the type 2 CALRm. However, both types of CALRm induced an increase in megakaryocytic progenitors associated with a ruxolitinib-sensitive independent growth and with a mild constitutive signaling in megakaryocytes. At the transcriptional level, type 1 CALRm seems to deregulate more pathways than the type 2 CALRm in megakaryocytes. Altogether, our results show that CALRm modify both the HSPC and megakaryocyte biology with a stronger effect for type 1 than for type 2 CALRm.


Asunto(s)
Calreticulina/efectos adversos , Hematopoyesis/efectos de los fármacos , Trastornos Mieloproliferativos/genética , Femenino , Humanos , Masculino , Mutación
5.
Nat Genet ; 51(10): 1518-1529, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31570891

RESUMEN

RNA modifications are emerging as key determinants of gene expression. However, compelling genetic demonstrations of their relevance to human disease are lacking. Here, we link ribosomal RNA 2'-O-methylation (2'-O-Me) to the etiology of dyskeratosis congenita. We identify nucleophosmin (NPM1) as an essential regulator of 2'-O-Me on rRNA by directly binding C/D box small nucleolar RNAs, thereby modulating translation. We demonstrate the importance of 2'-O-Me-regulated translation for cellular growth, differentiation and hematopoietic stem cell maintenance, and show that Npm1 inactivation in adult hematopoietic stem cells results in bone marrow failure. We identify NPM1 germline mutations in patients with dyskeratosis congenita presenting with bone marrow failure and demonstrate that they are deficient in small nucleolar RNA binding. Mice harboring a dyskeratosis congenita germline Npm1 mutation recapitulate both hematological and nonhematological features of dyskeratosis congenita. Thus, our findings indicate that impaired 2'-O-Me can be etiological to human disease.


Asunto(s)
Disqueratosis Congénita/genética , Epigenómica/métodos , Mutación de Línea Germinal , Proteínas Nucleares/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , ARN Ribosómico/genética , Animales , Disqueratosis Congénita/patología , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/química , Nucleofosmina , ARN Nucleolar Pequeño , Transcriptoma
6.
Oncogene ; 38(10): 1651-1660, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30846848

RESUMEN

Frameshift mutations in the calreticulin (CALR) gene are present in 30% of essential thrombocythemia and myelofibrosis patients. The two most frequent mutations are CALR del52 (type 1, approximately 60%) and CALR ins5 (type 2, around 30%), but many other rarer mutations exist accounting each for less than 2% of all CALR mutations. Most of them are structurally classified as type 1-like and type 2-like CALR mutations according to the absence or presence of a residual wild-type calcium-binding motif and the modification of the alpha-helix structure. Yet, several key questions remain unanswered, especially the reason of such low frequencies of these other mutations. In an attempt to investigate specific pathogenic differences between type 1-like and type 2-like CALR mutations and del52 and ins5, we modeled two type 1-like (del34 and del46) and one type 2-like (del19) mutations in cell lines and in mice. All CALR mutants constitutively activate JAK2 and STAT5/3/1 in a similar way in the presence of the thrombopoietin receptor (MPL) and induced cytokine-independent cell growth but to a lesser extent with rare mutants over time. This correlates with reduced expression levels of rare CALR mutants compared to del52 and ins5. Lethally irradiated mice that were engrafted with bone marrow transduced with the different CALR mutations developed thrombocytosis, but to a much lesser extent with ins5 and the type 2-like CALR mutation. In contrast to type 2-like mice, type 1-like mice developed marked myelofibrosis and splenomegaly 10 months after engraftment. Similar to del52, type 1-like CALR mutations induced an expansion at an early stage of hematopoiesis compared to ins5 and type 2-like mutation. Thus, type 1-like and type 2-like CALR mutants structurally and functionally resemble del52 and ins5 mutants, respectively.


Asunto(s)
Calreticulina/genética , Calreticulina/metabolismo , Mutación , Trastornos Mieloproliferativos/genética , Animales , Calreticulina/química , Línea Celular , Modelos Animales de Enfermedad , Humanos , Janus Quinasa 2/genética , Ratones , Trastornos Mieloproliferativos/metabolismo , Receptores de Trombopoyetina/metabolismo , Factores de Transcripción STAT/genética , Activación Transcripcional
7.
Blood ; 131(7): 717-732, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29146883

RESUMEN

Bone marrow (BM) failure (BMF) in children and young adults is often suspected to be inherited, but in many cases diagnosis remains uncertain. We studied a cohort of 179 patients (from 173 families) with BMF of suspected inherited origin but unresolved diagnosis after medical evaluation and Fanconi anemia exclusion. All patients had cytopenias, and 12.0% presented ≥5% BM blast cells. Median age at genetic evaluation was 11 years; 20.7% of patients were aged ≤2 years and 36.9% were ≥18 years. We analyzed genomic DNA from skin fibroblasts using whole-exome sequencing, and were able to assign a causal or likely causal germ line mutation in 86 patients (48.0%), involving a total of 28 genes. These included genes in familial hematopoietic disorders (GATA2, RUNX1), telomeropathies (TERC, TERT, RTEL1), ribosome disorders (SBDS, DNAJC21, RPL5), and DNA repair deficiency (LIG4). Many patients had an atypical presentation, and the mutated gene was often not clinically suspected. We also found mutations in genes seldom reported in inherited BMF (IBMF), such as SAMD9 and SAMD9L (N = 16 of the 86 patients, 18.6%), MECOM/EVI1 (N = 6, 7.0%), and ERCC6L2 (N = 7, 8.1%), each of which was associated with a distinct natural history; SAMD9 and SAMD9L patients often experienced transient aplasia and monosomy 7, whereas MECOM patients presented early-onset severe aplastic anemia, and ERCC6L2 patients, mild pancytopenia with myelodysplasia. This study broadens the molecular and clinical portrait of IBMF syndromes and sheds light on newly recognized disease entities. Using a high-throughput sequencing screen to implement precision medicine at diagnosis can improve patient management and family counseling.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Mutación de Línea Germinal , Adolescente , Enfermedades de la Médula Ósea/epidemiología , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Síndromes Mielodisplásicos/epidemiología , Síndromes Mielodisplásicos/genética , Secuenciación del Exoma
8.
Nat Commun ; 8(1): 1786, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29176689

RESUMEN

Thrombocytopenia is a major side effect of a new class of anticancer agents that target histone deacetylase (HDAC). Their mechanism is poorly understood. Here, we show that HDAC6 inhibition and genetic knockdown lead to a strong decrease in human proplatelet formation (PPF). Unexpectedly, HDAC6 inhibition-induced tubulin hyperacetylation has no effect on PPF. The PPF decrease induced by HDAC6 inhibition is related to cortactin (CTTN) hyperacetylation associated with actin disorganization inducing important changes in the distribution of megakaryocyte (MK) organelles. CTTN silencing in human MKs phenocopies HDAC6 inactivation and knockdown leads to a strong PPF defect. This is rescued by forced expression of a deacetylated CTTN mimetic. Unexpectedly, unlike human-derived MKs, HDAC6 and CTTN are shown to be dispensable for mouse PPF in vitro and platelet production in vivo. Our results highlight an unexpected function of HDAC6-CTTN axis as a positive regulator of human but not mouse MK maturation.


Asunto(s)
Cortactina/metabolismo , Histona Desacetilasa 6/metabolismo , Megacariocitos/metabolismo , Trombocitopenia/metabolismo , Acetilación/efectos de los fármacos , Animales , Plaquetas/citología , Plaquetas/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Cortactina/genética , Histona Desacetilasa 6/antagonistas & inhibidores , Histona Desacetilasa 6/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Megacariocitos/citología , Ratones Noqueados , Pirimidinas/farmacología , Interferencia de ARN , Trombocitopenia/genética
10.
Blood ; 128(26): 3146-3158, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-28034873

RESUMEN

The mechanisms behind the hereditary thrombocytosis induced by the thrombopoietin (THPO) receptor MPL P106L mutant remain unknown. A complete trafficking defect to the cell surface has been reported, suggesting either weak constitutive activity or nonconventional THPO-dependent mechanisms. Here, we report that the thrombocytosis phenotype induced by MPL P106L belongs to the paradoxical group, where low MPL levels on platelets and mature megakaryocytes (MKs) lead to high serum THPO levels, whereas weak but not absent MPL cell-surface localization in earlier MK progenitors allows response to THPO by signaling and amplification of the platelet lineage. MK progenitors from patients showed no spontaneous growth and responded to THPO, and MKs expressed MPL on their cell surface at low levels, whereas their platelets did not respond to THPO. Transduction of MPL P106L in CD34+ cells showed that this receptor was more efficiently localized at the cell surface on immature than on mature MKs, explaining a proliferative response to THPO of immature cells and a defect in THPO clearance in mature cells. In a retroviral mouse model performed in Mpl-/- mice, MPL P106L could induce a thrombocytosis phenotype with high circulating THPO levels. Furthermore, we could select THPO-dependent cell lines with more cell-surface MPL P106L localization that was detected by flow cytometry and [125I]-THPO binding. Altogether, these results demonstrate that MPL P106L is a receptor with an incomplete defect in trafficking, which induces a low but not absent localization of the receptor on cell surface and a response to THPO in immature MK cells.


Asunto(s)
Membrana Celular/metabolismo , Mutación/genética , Receptores de Trombopoyetina/genética , Trombocitosis/genética , Trombocitosis/patología , Animales , Línea Celular , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Familia , Femenino , Humanos , Masculino , Megacariocitos/metabolismo , Ratones , Linaje , Transporte de Proteínas , Receptores de Trombopoyetina/metabolismo , Retroviridae/metabolismo , Transducción Genética
11.
J Clin Invest ; 126(9): 3580-4, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27500492

RESUMEN

Fanconi anemia (FA) is a recessive genetic disease characterized by congenital abnormalities, chromosome instability, progressive bone marrow failure (BMF), and a strong predisposition to cancer. Twenty FA genes have been identified, and the FANC proteins they encode cooperate in a common pathway that regulates DNA crosslink repair and replication fork stability. We identified a child with severe BMF who harbored biallelic inactivating mutations of the translesion DNA synthesis (TLS) gene REV7 (also known as MAD2L2), which encodes the mutant REV7 protein REV7-V85E. Patient-derived cells demonstrated an extended FA phenotype, which included increased chromosome breaks and G2/M accumulation upon exposure to DNA crosslinking agents, γH2AX and 53BP1 foci accumulation, and enhanced p53/p21 activation relative to cells derived from healthy patients. Expression of WT REV7 restored normal cellular and functional phenotypes in the patient's cells, and CRISPR/Cas9 inactivation of REV7 in a non-FA human cell line produced an FA phenotype. Finally, silencing Rev7 in primary hematopoietic cells impaired progenitor function, suggesting that the DNA repair defect underlies the development of BMF in FA. Taken together, our genetic and functional analyses identified REV7 as a previously undescribed FA gene, which we term FANCV.


Asunto(s)
Anemia de Fanconi/genética , Proteínas Mad2/genética , Mutación , Alelos , Animales , Ciclo Celular , Línea Celular Tumoral , Niño , Inestabilidad Cromosómica , Rotura Cromosómica , Estudios de Cohortes , Reactivos de Enlaces Cruzados/química , Daño del ADN , Reparación del ADN , Femenino , Fibroblastos/metabolismo , Silenciador del Gen , Prueba de Complementación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Células Madre Hematopoyéticas/citología , Humanos , Lentivirus , Proteínas Mad2/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitosis , Fenotipo
12.
Blood ; 127(3): 333-42, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26450985

RESUMEN

Mutations in signaling molecules of the cytokine receptor axis play a central role in myeloproliferative neoplasm (MPN) pathogenesis. Polycythemia vera is mainly related to JAK2 mutations, whereas a wider mutational spectrum is detected in essential thrombocythemia (ET) with mutations in JAK2, the thrombopoietin (TPO) receptor (MPL), and the calreticulin (CALR) genes. Here, we studied the mutational profile of 17 ET patients negative for JAK2V617F, MPLW515K/L, and CALR mutations, using whole-exome sequencing and next-generation sequencing (NGS) targeted on JAK2 and MPL. We found several signaling mutations including JAK2V617F at very low allele frequency, 1 homozygous SH2B3 mutation, 1 MPLS505N, 1 MPLW515R, and 2 MPLS204P mutations. In the remaining patients, 4 presented a clonal and 7 a polyclonal hematopoiesis, suggesting that certain triple-negative ETs are not MPNs. NGS on 26 additional triple-negative ETs detected only 1 MPLY591N mutation. Functional studies on MPLS204P and MPLY591N revealed that they are weak gain-of-function mutants increasing MPL signaling and conferring either TPO hypersensitivity or independence to expressing cells, but with a low efficiency. Further studies should be performed to precisely determine the frequency of MPLS204 and MPLY591 mutants in a bigger cohort of MPN.


Asunto(s)
Mutación , Receptores de Trombopoyetina/genética , Trombocitemia Esencial/genética , Sustitución de Aminoácidos , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Codón , Estudios de Cohortes , Hibridación Genómica Comparativa , Citocinas/farmacología , Análisis Mutacional de ADN , Exoma , Genotipo , Granulocitos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Janus Quinasa 2/genética , Transporte de Proteínas , Receptores de Trombopoyetina/metabolismo , Trombocitemia Esencial/metabolismo
13.
Stem Cell Reports ; 3(6): 1085-102, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25458892

RESUMEN

Hematopoietic stem cells (HSCs) are characterized by the capacity for self-renewal and the ability to reconstitute the entire hematopoietic compartment. Thrombopoietin maintains adult HSCs in a quiescent state through the induction of cell cycle inhibitors p57(Kip2) and p19(INK4d). Using the p19(INK4d-/-) mouse model, we investigated the role of p19(INK4d) in basal and stress-induced hematopoiesis. We demonstrate that p19(INK4d) is involved in the regulation of HSC quiescence by inhibition of the G0/G1 cell cycle transition. Under genotoxic stress conditions, the absence of p19(INK4d) in HSCs leads to accelerated cell cycle exit, accumulation of DNA double-strand breaks, and apoptosis when cells progress to the S/G2-M stages of the cell cycle. Moreover, p19(INK4d) controls the HSC microenvironment through negative regulation of megakaryopoiesis. Deletion of p19(INK4d) results in megakaryocyte hyperproliferation and increased transforming growth factor ß1 secretion. This leads to fibrosis in the bone marrow and spleen, followed by loss of HSCs during aging.


Asunto(s)
Senescencia Celular/genética , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Nicho de Células Madre/genética , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Recuento de Células , Diferenciación Celular/genética , Linaje de la Célula/genética , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/metabolismo , Hematopoyesis , Ratones , Ratones Noqueados , Osteosclerosis/genética , Osteosclerosis/patología , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Fase de Descanso del Ciclo Celular/genética , Células del Estroma/metabolismo
14.
Blood ; 121(13): 2440-51, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23335373

RESUMEN

The transcription factor Ikaros regulates the development of hematopoietic cells. Ikaros-deficient animals fail to develop B cells and display a T-cell malignancy, which is correlated with altered Notch signaling. Recently, loss of Ikaros was associated with progression of myeloproliferative neoplasms to acute myeloid leukemia and increasing evidence shows that Ikaros is also critical for the regulation of myeloid development. Previous studies showed that Ikaros-deficient mice have increased megakaryopoiesis, but the molecular mechanism of this phenomenon remains unknown. Here, we show that Ikaros overexpression decreases NOTCH-induced megakaryocytic specification, and represses expression of several megakaryocytic genes including GATA-1 to block differentiation and terminal maturation. We also demonstrate that Ikaros expression is differentially regulated by GATA-2 and GATA-1 during megakaryocytic differentiation and reveal that the combined loss of Ikzf1 and Gata1 leads to synthetic lethality in vivo associated with prominent defects in erythroid cells and an expansion of megakaryocyte progenitors. Taken together, our observations demonstrate an important functional interplay between Ikaros, GATA factors, and the NOTCH signaling pathway in specification and homeostasis of the megakaryocyte lineage.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Factor de Transcripción Ikaros/fisiología , Receptores Notch/metabolismo , Trombopoyesis/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo/genética , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Noqueados , Modelos Biológicos , Unión Proteica/genética , Unión Proteica/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
15.
J Exp Med ; 209(11): 2017-31, 2012 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-23045605

RESUMEN

Acute megakaryoblastic leukemia (AMKL) is a heterogeneous disease generally associated with poor prognosis. Gene expression profiles indicate the existence of distinct molecular subgroups, and several genetic alterations have been characterized in the past years, including the t(1;22)(p13;q13) and the trisomy 21 associated with GATA1 mutations. However, the majority of patients do not present with known mutations, and the limited access to primary patient leukemic cells impedes the efficient development of novel therapeutic strategies. In this study, using a xenotransplantation approach, we have modeled human pediatric AMKL in immunodeficient mice. Analysis of high-throughput RNA sequencing identified recurrent fusion genes defining new molecular subgroups. One subgroup of patients presented with MLL or NUP98 fusion genes leading to up-regulation of the HOX A cluster genes. A novel CBFA2T3-GLIS2 fusion gene resulting from a cryptic inversion of chromosome 16 was identified in another subgroup of 31% of non-Down syndrome AMKL and strongly associated with a gene expression signature of Hedgehog pathway activation. These molecular data provide useful markers for the diagnosis and follow up of patients. Finally, we show that AMKL xenograft models constitute a relevant in vivo preclinical screening platform to validate the efficacy of novel therapies such as Aurora A kinase inhibitors.


Asunto(s)
Genómica/métodos , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Leucemia Megacarioblástica Aguda/genética , Ensayos Antitumor por Modelo de Xenoinjerto , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Anciano , Secuencia de Aminoácidos , Animales , Aurora Quinasa A , Aurora Quinasas , Azepinas/farmacología , Secuencia de Bases , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Lactante , Estimación de Kaplan-Meier , Factores de Transcripción de Tipo Kruppel/genética , Leucemia Megacarioblástica Aguda/patología , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Fusión Oncogénica/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Proteínas Represoras/genética
16.
Nat Commun ; 3: 717, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22395608

RESUMEN

Megakaryocytes are unique mammalian cells that undergo polyploidization (endomitosis) during differentiation, leading to an increase in cell size and protein production that precedes platelet production. Recent evidence demonstrates that endomitosis is a consequence of a late failure in cytokinesis associated with a contractile ring defect. Here we show that the non-muscle myosin IIB heavy chain (MYH10) is expressed in immature megakaryocytes and specifically localizes in the contractile ring. MYH10 downmodulation by short hairpin RNA increases polyploidization by inhibiting the return of 4N cells to 2N, but other regulators, such as of the G1/S transition, might regulate further polyploidization of the 4N cells. Conversely, re-expression of MYH10 in the megakaryocytes prevents polyploidization and the transition of 2N to 4N cells. During polyploidization, MYH10 expression is repressed by the major megakaryocyte transcription factor RUNX1. Thus, RUNX1-mediated silencing of MYH10 is required for the switch from mitosis to endomitosis, linking polyploidization with megakaryocyte differentiation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Megacariocitos/citología , Cadenas Pesadas de Miosina/genética , Miosina Tipo IIB no Muscular/genética , Poliploidía , Animales , Antígenos CD34/biosíntesis , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Citocinesis , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Megacariocitos/metabolismo , Ratones , Ratones Noqueados , Mitosis , Cadenas Pesadas de Miosina/biosíntesis , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo IIB no Muscular/biosíntesis , Miosina Tipo IIB no Muscular/metabolismo , Interferencia de ARN , ARN Interferente Pequeño
17.
Blood ; 117(1): 246-9, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20944070

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is the most important cytokine involved in the promotion of myelofibrosis. Mechanisms leading to its local activation in the bone marrow environment remain unclear. As a recent study has highlighted the role of thrombospondin-1 (TSP-1) in platelet-derived TGF-ß1 activation, we investigated the role of TSP-1 in the TPO(high) murine model of myelofibrosis. Two groups of engrafted mice, WT TPO(high) and Tsp-1-null TPO(high), were constituted. All mice developed a similar myeloproliferative syndrome and an increase in total TGF-ß1 levels in the plasma and in extracellular fluids of marrow and spleen. Surprisingly, we were able to detect the active form of TGF-ß1 in Tsp-1-null TPO(high) mice. Accordingly, these mice developed marrow and spleen fibrosis, with intriguingly a higher grade than in WT TPO(high) mice. Our results show that TSP-1 is not the major activator of TGF-ß1 in TPO-induced myelofibrosis, suggesting the contribution of another mechanism in the megakaryocyte/platelet compartment.


Asunto(s)
Mielofibrosis Primaria/inducido químicamente , Mielofibrosis Primaria/patología , Trombopoyetina/efectos adversos , Trombospondina 1/fisiología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Plaquetas/metabolismo , Plaquetas/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Femenino , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mielofibrosis Primaria/metabolismo , Bazo/metabolismo , Bazo/patología
18.
Development ; 137(9): 1573-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20388655

RESUMEN

The intestinal epithelium is a complex system characterized by massive and continuous cell renewal and differentiation. In this context, cell-type-specific transcription factors are thought to play a crucial role by modulating specific transcription networks and signalling pathways. Hnf1alpha and beta are closely related atypical homeoprotein transcription factors expressed in several epithelia, including the gut. With the use of a conditional inactivation system, we generated mice in which Hnf1b is specifically inactivated in the intestinal epithelium on a wild-type or Hnf1a(-/-) genetic background. Whereas the inactivation of Hnf1a or Hnf1b alone did not lead to any major intestinal dysfunction, the concomitant inactivation of both genes resulted in a lethal phenotype. Double-mutant animals had defective differentiation and cell fate commitment. The expression levels of markers of all the differentiated cell types, both enterocytes and secretory cells, were affected. In addition, the number of goblet cells was increased, whereas mature Paneth cells were missing. At the molecular level, we show that Hnf1alpha and beta act upstream of the Notch pathway controlling directly the expression of two crucial components: Jag1 and Atoh1. We demonstrate that the double-mutant mice present with a defect in intestinal water absorption and that Hnf1alpha and beta directly control the expression of Slc26a3, a gene whose mutations are associated with chloride diarrhoea in human patients. Our study identifies new direct target genes of the Hnf1 transcription factors and shows that they play crucial roles in both defining cell fate and controlling terminal functions in the gut epithelium.


Asunto(s)
Diferenciación Celular/fisiología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-beta del Hepatocito/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Animales , Antiportadores/genética , Antiportadores/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/genética , Inmunoprecipitación de Cromatina , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/genética , Inmunohistoquímica , Hibridación in Situ , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Proteínas Serrate-Jagged , Transportadores de Sulfato
20.
J Hepatol ; 41(2): 292-8, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15288479

RESUMEN

BACKGROUND/AIMS: Combined hepatocellular-cholangiocarcinoma (HCC-CC) show dual hepatocellular and biliary epithelial differentiation. To better understand the relations between cholangiocarcinoma (CC), HCC-CC and hepatocellular carcinoma (HCC), we screened for genetic alterations. METHODS: A series of nine CC, 15 HCC-CC and three separated HCC and CC lesions ('collision tumors') were screened for loss of heterozygosity (LOH) using 400 microsatellite markers and for p53 and beta-catenin mutations. A comparison with a previously characterized series of 137 HCC was performed. RESULTS: In six cases of CC and HCC-CC, we identified TP53 gene mutations. A CTNNB1/beta-catenin was identified in two patients presenting collision tumors, but no mutations were found in CC or in HCC-CC. A high level of chromosome instability in both CC and HCC-CC was found. Recurrent specific LOH were identified at 3p and 14q in more than 50% of the CC and the HCC-CC cases, whereas these chromosomal regions were deleted in less than 10% of the HCC cases (P<10(-5)). Minimal common regions of deletion (MCRD) were defined at 3p24-p14 and 14q24-q32, respectively. CONCLUSIONS: These results suggest that combined HCC-CC are genetically closer to CC than HCC and common carcinogenesis pathways may be altered in HCC-CC and CC.


Asunto(s)
Carcinoma Hepatocelular/genética , Colangiocarcinoma/genética , Neoplasias Hepáticas/genética , Anciano , Alelos , Carcinoma Hepatocelular/patología , Colangiocarcinoma/patología , Inestabilidad Cromosómica , Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 3/genética , Proteínas del Citoesqueleto/genética , Femenino , Eliminación de Gen , Genes p53 , Genoma Humano , Humanos , Neoplasias Hepáticas/patología , Pérdida de Heterocigocidad , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Mutación , Transactivadores/genética , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA