Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Hum Mutat ; 31(12): 1294-303, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20848650

RESUMEN

Hereditary fructose intolerance (HFI) is an autosomal recessive metabolic disease caused by impaired functioning of human liver aldolase (ALDOB). At least 54 subtle/point mutations and only two large intragenic deletions have been found in the ALDOB gene. Here we report two novel ALDOB variants (p.R46W and p.Y343H) and an intragenic deletion that we found in patients with suspected HFI. The residual catalytic activity of the recombinant p.R46W and p.Y343H variants toward F1P was particularly altered. We also characterized a large intragenic deletion that we found in six unrelated patients. This is the first report of six unrelated patients sharing the same ALDOB deletion, thus indicating a founder effect for this allele in our geographic area. Because this deletion involves ALDOB exon 5, it can mimic worldwide common pathogenic genotypes, that is, homozygous p.A150P and p.A175D. Finally, the identification of only one ALDOB mutation in symptomatic patients suggests that HFI symptoms can, albeit rarely, appear also in heterozygotes. Therefore, an excessive and continuous fructose dietary intake may have deleterious effects even in apparently asymptomatic HFI carriers.


Asunto(s)
Intolerancia a la Fructosa/enzimología , Intolerancia a la Fructosa/genética , Fructosa-Bifosfato Aldolasa/genética , Eliminación de Gen , Alelos , Secuencia de Bases , Niño , Dicroismo Circular , Resultado Fatal , Femenino , Intolerancia a la Fructosa/diagnóstico , Fructosa-Bifosfato Aldolasa/química , Frecuencia de los Genes/genética , Humanos , Lactante , Italia , Cinética , Masculino , Datos de Secuencia Molecular , Linaje , Estructura Secundaria de Proteína , Proteínas Recombinantes/metabolismo
2.
Exp Hematol ; 92: 62-74, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33152396

RESUMEN

Acute myeloid leukemia development occurs in a stepwise fashion whereby an original driver mutation is followed by additional mutations. The first type of mutations tends to be in genes encoding members of the epigenetic/transcription regulatory machinery (i.e., RUNX1, DNMT3A, TET2), while the secondary mutations often involve genes encoding members of signaling pathways that cause uncontrolled growth of such cells such as the growth factor receptors c-KIT of FLT3. Patients usually present with both types of mutations, but it is currently unclear how both mutational events shape the epigenome in developing AML cells. To this end we generated an in vitro model of t(8;21) AML by expressing its driver oncoprotein RUNX1-ETO with or without a mutated (N822K) KIT protein. Expression of N822K-c-KIT strongly increases the self-renewal capacity of RUNX1-ETO-expressing cells. Global analysis of gene expression changes and alterations in the epigenome revealed that N822K-c-KIT expression profoundly influences the open chromatin landscape and transcription factor binding. However, our experiments also revealed that double mutant cells still differ from their patient-derived counterparts, highlighting the importance of studying patient cells to obtain a true picture of how gene regulatory networks have been reprogrammed during tumorigenesis.


Asunto(s)
Cromatina/metabolismo , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda , Mutación Missense , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Transcripción Genética , Translocación Genética , Sustitución de Aminoácidos , Cromatina/patología , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 21/metabolismo , Cromosomas Humanos Par 8/genética , Cromosomas Humanos Par 8/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Femenino , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Proteínas Proto-Oncogénicas c-kit/genética , Proteína 1 Compañera de Translocación de RUNX1/genética
4.
Nat Genet ; 51(1): 151-162, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30420649

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease caused by a variety of alterations in transcription factors, epigenetic regulators and signaling molecules. To determine how different mutant regulators establish AML subtype-specific transcriptional networks, we performed a comprehensive global analysis of cis-regulatory element activity and interaction, transcription factor occupancy and gene expression patterns in purified leukemic blast cells. Here, we focused on specific subgroups of subjects carrying mutations in genes encoding transcription factors (RUNX1, CEBPα), signaling molecules (FTL3-ITD, RAS) and the nuclear protein NPM1). Integrated analysis of these data demonstrates that each mutant regulator establishes a specific transcriptional and signaling network unrelated to that seen in normal cells, sustaining the expression of unique sets of genes required for AML growth and maintenance.


Asunto(s)
Regulación Leucémica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Leucemia Mieloide Aguda/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nucleofosmina , Transducción de Señal/genética , Factores de Transcripción/genética , Adulto Joven
5.
Blood Adv ; 2(3): 271-284, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29431622

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease caused by recurrent mutations in the transcription regulatory machinery, resulting in abnormal growth and a block in differentiation. One type of recurrent mutations affects RUNX1, which is subject to mutations and translocations, the latter giving rise to fusion proteins with aberrant transcriptional activities. We recently compared the mechanism by which the products of the t(8;21) and the t(3;21) translocation RUNX1-ETO and RUNX1-EVI1 reprogram the epigenome. We demonstrated that a main component of the block in differentiation in both types of AML is direct repression of the gene encoding the myeloid regulator C/EBPα by both fusion proteins. Here, we examined at the global level whether C/EBPα is able to reverse aberrant chromatin programming in t(8;21) and t(3;21) AML. C/EBPα overexpression does not change oncoprotein expression or globally displace these proteins from their binding sites. Instead, it upregulates a core set of common target genes important for myeloid differentiation and represses genes regulating leukemia maintenance. This study, therefore, identifies common CEBPA-regulated pathways as targets for therapeutic intervention.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Reprogramación Celular , Epigénesis Genética , Leucemia Mieloide Aguda/genética , Factores de Transcripción , Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular/genética , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Leucemia Mieloide Aguda/patología , Mutación
6.
Cancer Cell ; 34(4): 674-689.e8, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30245083

RESUMEN

Intra-tumor heterogeneity caused by clonal evolution is a major problem in cancer treatment. To address this problem, we performed label-free quantitative proteomics on primary acute myeloid leukemia (AML) samples. We identified 50 leukemia-enriched plasma membrane proteins enabling the prospective isolation of genetically distinct subclones from individual AML patients. Subclones differed in their regulatory phenotype, drug sensitivity, growth, and engraftment behavior, as determined by RNA sequencing, DNase I hypersensitive site mapping, transcription factor occupancy analysis, in vitro culture, and xenograft transplantation. Finally, we show that these markers can be used to identify and longitudinally track distinct leukemic clones in patients in routine diagnostics. Our study describes a strategy for a major improvement in stratifying cancer diagnosis and treatment.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mutación/genética , Fenotipo , Factores de Transcripción/genética , Adulto , Anciano , Secuencia de Bases/genética , Evolución Clonal/genética , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Tirosina Quinasa 3 Similar a fms/genética
7.
Cell Rep ; 19(8): 1654-1668, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28538183

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous disease caused by mutations in transcriptional regulator genes, but how different mutant regulators shape the chromatin landscape is unclear. Here, we compared the transcriptional networks of two types of AML with chromosomal translocations of the RUNX1 locus that fuse the RUNX1 DNA-binding domain to different regulators, the t(8;21) expressing RUNX1-ETO and the t(3;21) expressing RUNX1-EVI1. Despite containing the same DNA-binding domain, the two fusion proteins display distinct binding patterns, show differences in gene expression and chromatin landscape, and are dependent on different transcription factors. RUNX1-EVI1 directs a stem cell-like transcriptional network reliant on GATA2, whereas that of RUNX1-ETO-expressing cells is more mature and depends on RUNX1. However, both types of AML are dependent on the continuous expression of the fusion proteins. Our data provide a molecular explanation for the differences in clinical prognosis for these types of AML.


Asunto(s)
Cromatina/metabolismo , Cromosomas Humanos/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/genética , Translocación Genética/genética , Secuencia de Bases , Sitios de Unión , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Supervivencia Celular/genética , Factor de Transcripción GATA2/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Fenotipo
8.
Int J Hematol ; 101(4): 319-29, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25749719

RESUMEN

The differentiation from multipotent hematopoietic stem cells (HSC) to mature and functional blood cells requires the finely tuned regulation of gene expression at each stage of development. Specific transcription factors play a key role in this process as they modulate the expression of their target genes in an exquisitely lineage-specific manner. A large number of important transcriptional regulators have been identified which establish and maintain specific gene expression patterns during hematopoietic development. Hematopoiesis is therefore a paradigm for investigating how transcription factors function in mammalian cells, thanks also to the evolution of genome-wide and the next-generation sequencing technologies. In this review, we focus on the current knowledge of the biological and functional properties of the hematopoietic master regulator RUNX1 (also known as AML1, CBFA2, PEBP2aB) transcription factor and its main downstream target PU.1. We will outline their relationship in determining the fate of the myeloid lineage during normal stem cell development and under conditions when hematopoietic development is subverted by leukemic transformation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hematopoyesis , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/análisis , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Transactivadores/análisis , Transactivadores/genética
9.
Cell Rep ; 12(5): 821-36, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212328

RESUMEN

Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect the epigenetic regulatory machinery and signaling molecules, leading to a block in hematopoietic differentiation. Constitutive signaling from mutated growth factor receptors is a major driver of leukemic growth, but how aberrant signaling affects the epigenome in AML is less understood. Furthermore, AML cells undergo extensive clonal evolution, and the mutations in signaling genes are often secondary events. To elucidate how chronic growth factor signaling alters the transcriptional network in AML, we performed a system-wide multi-omics study of primary cells from patients suffering from AML with internal tandem duplications in the FLT3 transmembrane domain (FLT3-ITD). This strategy revealed cooperation between the MAP kinase (MAPK) inducible transcription factor AP-1 and RUNX1 as a major driver of a common, FLT3-ITD-specific gene expression and chromatin signature, demonstrating a major impact of MAPK signaling pathways in shaping the epigenome of FLT3-ITD AML.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/enzimología , Sistema de Señalización de MAP Quinasas , Mutación , Tirosina Quinasa 3 Similar a fms/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Estructura Terciaria de Proteína , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Tirosina Quinasa 3 Similar a fms/genética
10.
Cell Rep ; 8(6): 1974-1988, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25242324

RESUMEN

Oncogenic transcription factors such as RUNX1/ETO, which is generated by the chromosomal translocation t(8;21), subvert normal blood cell development by impairing differentiation and driving malignant self-renewal. Here, we use digital footprinting and chromatin immunoprecipitation sequencing (ChIP-seq) to identify the core RUNX1/ETO-responsive transcriptional network of t(8;21) cells. We show that the transcriptional program underlying leukemic propagation is regulated by a dynamic equilibrium between RUNX1/ETO and RUNX1 complexes, which bind to identical DNA sites in a mutually exclusive fashion. Perturbation of this equilibrium in t(8;21) cells by RUNX1/ETO depletion leads to a global redistribution of transcription factor complexes within preexisting open chromatin, resulting in the formation of a transcriptional network that drives myeloid differentiation. Our work demonstrates on a genome-wide level that the extent of impaired myeloid differentiation in t(8;21) is controlled by the dynamic balance between RUNX1/ETO and RUNX1 activities through the repression of transcription factors that drive differentiation.


Asunto(s)
Leucemia Mieloide Aguda/patología , Translocación Genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Cromosomas Humanos Par 21 , Cromosomas Humanos Par 8 , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Redes Reguladoras de Genes , Humanos , Proteínas con Dominio LIM/metabolismo , Leucemia Mieloide Aguda/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Análisis de Secuencia de ARN , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA