Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Genes Dev ; 29(18): 1915-29, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26385962

RESUMEN

The balance between self-renewal and differentiation is crucial for the maintenance of hematopoietic stem cells (HSCs). Whereas numerous gene regulatory factors have been shown to control HSC self-renewal or drive their differentiation, we have relatively few insights into transcription factors that serve to restrict HSC differentiation. In the present work, we identify ETS (E-twenty-six)-related gene (ERG) as a critical factor protecting HSCs from differentiation. Specifically, loss of Erg accelerates HSC differentiation by >20-fold, thus leading to rapid depletion of immunophenotypic and functional HSCs. Molecularly, we could demonstrate that ERG, in addition to promoting the expression of HSC self-renewal genes, also represses a group of MYC targets, thereby explaining why Erg loss closely mimics Myc overexpression. Consistently, the BET domain inhibitor CPI-203, known to repress Myc expression, confers a partial phenotypic rescue. In summary, ERG plays a critical role in coordinating the balance between self-renewal and differentiation of HSCs.


Asunto(s)
Diferenciación Celular/genética , Células Madre Hematopoyéticas/citología , Proteínas Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Animales , Células de la Médula Ósea/fisiología , Adhesión Celular/genética , Movimiento Celular/genética , Transformación Celular Neoplásica/genética , Células Cultivadas , Eliminación de Gen , Ratones , Proteínas Oncogénicas/genética , Factores de Transcripción/genética , Regulador Transcripcional ERG
2.
PLoS Genet ; 10(1): e1004079, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24415956

RESUMEN

Transcription factors are key regulators of hematopoietic stem cells (HSCs) and act through their ability to bind DNA and impact on gene transcription. Their functions are interpreted in the complex landscape of chromatin, but current knowledge on how this is achieved is very limited. C/EBPα is an important transcriptional regulator of hematopoiesis, but its potential functions in HSCs have remained elusive. Here we report that C/EBPα serves to protect adult HSCs from apoptosis and to maintain their quiescent state. Consequently, deletion of Cebpa is associated with loss of self-renewal and HSC exhaustion. By combining gene expression analysis with genome-wide assessment of C/EBPα binding and epigenetic configurations, we show that C/EBPα acts to modulate the epigenetic states of genes belonging to molecular pathways important for HSC function. Moreover, our data suggest that C/EBPα acts as a priming factor at the HSC level where it actively promotes myeloid differentiation and counteracts lymphoid lineage choice. Taken together, our results show that C/EBPα is a key regulator of HSC biology, which influences the epigenetic landscape of HSCs in order to balance different cell fate options.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Animales , Apoptosis , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Linaje de la Célula , Proliferación Celular , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Ratones
3.
Blood ; 123(15): 2389-400, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24516045

RESUMEN

The loss of regulation of cell proliferation is a key event in leukemic transformation, and the oncogene tribbles (Trib)2 is emerging as a pivotal target of transcription factors in acute leukemias. Deregulation of the transcription factor E2F1, normally repressed by CCAAT enhancer-binding protein α (C/EBPα)-p42, occurs in acute myeloid leukemia (AML), resulting in the perturbation of cell cycle and apoptosis, emphasizing its importance in the molecular pathogenesis of AML. Here we show that E2F family members directly regulate Trib2 in leukemic cells and identify a feedback regulatory loop for E2F1, C/EBPα, and Trib2 in AML cell proliferation and survival. Further analyses revealed that E2F1-mediated Trib2 expression was repressed by C/EBPα-p42, and in normal granulocyte/macrophage progenitor cells, we detect C/EBPα bound to the Trib2 promoter. Pharmacological inhibition of the cell cycle or Trib2 knockdown resulted in a block in AML cell proliferation. Our work proposes a novel paradigm whereby E2F1 plays a key role in the regulation of Trib2 expression important for AML cell proliferation control. Importantly, we identify the contribution of dysregulated C/EBPα and E2F1 to elevated Trib2 expression and leukemic cell survival, which likely contributes to the initiation and maintenance of AML and may have significant implications for normal and malignant hematopoiesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Transformación Celular Neoplásica/genética , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia Mieloide Aguda/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Células 3T3 , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proliferación Celular , Inmunoprecipitación de Cromatina , Factor de Transcripción E2F1/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Retroalimentación Fisiológica/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
4.
BMC Genomics ; 16: 46, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25652644

RESUMEN

BACKGROUND: Chromatin-Immunoprecipitation coupled with deep sequencing (ChIP-seq) is used to map transcription factor occupancy and generate epigenetic profiles genome-wide. The requirement of nano-scale ChIP DNA for generation of sequencing libraries has impeded ChIP-seq on in vivo tissues of low cell numbers. RESULTS: We describe a robust, simple and scalable methodology for ChIP-seq of low-abundant cell populations, verified down to 10,000 cells. By employing non-mammalian genome mapping bacterial carrier DNA during amplification, we reliably amplify down to 50 pg of ChIP DNA from transcription factor (CEBPA) and histone mark (H3K4me3) ChIP. We further demonstrate that genomic profiles are highly resilient to changes in carrier DNA to ChIP DNA ratios. CONCLUSIONS: This represents a significant advance compared to existing technologies, which involve either complex steps of pre-selection for nucleosome-containing chromatin or pre-amplification of precipitated DNA, making them prone to introduce experimental biases.


Asunto(s)
Inmunoprecipitación de Cromatina , Técnicas de Amplificación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción/genética , Animales , Cromatina/genética , ADN Bacteriano/genética , Genoma Bacteriano/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ADN
5.
Bioinformatics ; 30(8): 1154-1162, 2014 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-24403540

RESUMEN

MOTIVATION: Histone modifications are a key epigenetic mechanism to activate or repress the transcription of genes. Datasets of matched transcription data and histone modification data obtained by ChIP-seq exist, but methods for integrative analysis of both data types are still rare. Here, we present a novel bioinformatics approach to detect genes that show different transcript abundances between two conditions putatively caused by alterations in histone modification. RESULTS: We introduce a correlation measure for integrative analysis of ChIP-seq and gene transcription data measured by RNA sequencing or microarrays and demonstrate that a proper normalization of ChIP-seq data is crucial. We suggest applying Bayesian mixture models of different types of distributions to further study the distribution of the correlation measure. The implicit classification of the mixture models is used to detect genes with differences between two conditions in both gene transcription and histone modification. The method is applied to different datasets, and its superiority to a naive separate analysis of both data types is demonstrated. AVAILABILITY AND IMPLEMENTATION: R/Bioconductor package epigenomix. CONTACT: h.klein@uni-muenster.de Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Inmunoprecipitación de Cromatina , Biología Computacional/métodos , Histonas/química , Teorema de Bayes , Epigénesis Genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ARN/métodos , Transcripción Genética
6.
Nat Commun ; 13(1): 3595, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35739121

RESUMEN

Differentiation of multipotent stem cells into mature cells is fundamental for development and homeostasis of mammalian tissues, and requires the coordinated induction of lineage-specific transcriptional programs and cell cycle withdrawal. To understand the underlying regulatory mechanisms of this fundamental process, we investigated how the tissue-specific transcription factors, CEBPA and CEBPE, coordinate cell cycle exit and lineage-specification in vivo during granulocytic differentiation. We demonstrate that CEBPA promotes lineage-specification by launching an enhancer-primed differentiation program and direct activation of CEBPE expression. Subsequently, CEBPE confers promoter-driven cell cycle exit by sequential repression of MYC target gene expression at the G1/S transition and E2F-meditated G2/M gene expression, as well as by the up-regulation of Cdk1/2/4 inhibitors. Following cell cycle exit, CEBPE unleashes the CEBPA-primed differentiation program to generate mature granulocytes. These findings highlight how tissue-specific transcription factors coordinate cell cycle exit with differentiation through the use of distinct gene regulatory elements.


Asunto(s)
Regulación de la Expresión Génica , Factores de Transcripción , Animales , Ciclo Celular , Diferenciación Celular/genética , Granulocitos/metabolismo , Mamíferos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
J Exp Med ; 202(1): 85-96, 2005 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-15983063

RESUMEN

CCAAT/enhancer binding protein (C/EBP)alpha is a myeloid-specific transcription factor that couples lineage commitment to terminal differentiation and cell cycle arrest, and is found mutated in 9% of patients who have acute myeloid leukemia (AML). We previously showed that mutations which dissociate the ability of C/EBP alpha to block cell cycle progression through E2F inhibition from its function as a transcriptional activator impair the in vivo development of the neutrophil granulocyte and adipose lineages. We now show that such mutations increase the capacity of bone marrow (BM) myeloid progenitors to proliferate, and predispose mice to a granulocytic myeloproliferative disorder and transformation of the myeloid compartment of the BM. Both of these phenotypes were transplantable into lethally irradiated recipients. BM transformation was characterized by a block in granulocyte differentiation, accumulation of myeloblasts and promyelocytes, and expansion of myeloid progenitor populations--all characteristics of AML. Circulating myeloblasts and hepatic leukocyte infiltration were observed, but thrombocytopenia, anemia, and elevated leukocyte count--normally associated with AML-were absent. These results show that disrupting the cell cycle regulatory function of C/EBP alpha is sufficient to initiate AML-like transformation of the granulocytic lineage, but only partially the peripheral pathology of AML.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Ciclo Celular/fisiología , Granulocitos/citología , Células Progenitoras Mieloides/citología , Neutrófilos/citología , Animales , Trasplante de Médula Ósea , Proteína alfa Potenciadora de Unión a CCAAT/genética , Ciclo Celular/genética , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fenotipo
8.
Mol Cell Biol ; 26(3): 1028-37, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16428455

RESUMEN

The C/EBPalpha transcription factor regulates growth and differentiation of several tissues during embryonic development. Several hypotheses as to how C/EBPalpha inhibits cellular growth in vivo have been derived, mainly from studies of tissue culture cells. In fetal liver it has been proposed that a short, centrally located, 15-amino-acid proline-histidine-rich region (PHR) of C/EBPalpha is responsible for the growth-inhibitory function of the protein through its ability to interact with CDK2 and CDK4, thereby inhibiting their activities. Homozygous Cebpa(DeltaPHR/DeltaPHR) (DeltaPHR) mice, carrying a modified cebpa allele lacking amino acids 180 to 194, were born at the Mendelian ratio, reached adulthood, and displayed no apparent adverse phenotypes. When fetal livers from the DeltaPHR mice were analyzed for their expression of cell cycle markers, bromodeoxyuridine incorporation, cyclin-dependent kinase 2 kinase activity, and global gene expression, we failed to detect any cell cycle or developmental differences between the DeltaPHR mice and their control littermates. These in vivo data demonstrate that any C/EBPalpha-mediated growth repression via the PHR as well as the basic region is dispensable for proper embryonic development of, and cell cycle control in, the liver. Surprisingly, control experiments performed in C/EBPalpha null fetal livers yielded similar results.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Histidina/química , Hígado/embriología , Prolina/química , Adipocitos/citología , Secuencia de Aminoácidos , Animales , Proteína alfa Potenciadora de Unión a CCAAT/química , Proteína alfa Potenciadora de Unión a CCAAT/genética , Diferenciación Celular , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Histidina/genética , Humanos , Hígado/metabolismo , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Prolina/genética , Estructura Terciaria de Proteína , Ratas , Eliminación de Secuencia
9.
Cell Rep ; 29(9): 2756-2769.e6, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31775043

RESUMEN

B cell development depends on the coordinated expression and cooperation of several transcription factors. Here we show that the transcription factor ETS-related gene (ERG) is crucial for normal B cell development and that its deletion results in a substantial loss of bone marrow B cell progenitors and peripheral B cells, as well as a skewing of splenic B cell populations. We find that ERG-deficient B lineage cells exhibit an early developmental block at the pre-B cell stage and proliferate less. The cells fail to express the immunoglobulin heavy chain due to inefficient V-to-DJ recombination, and cells that undergo recombination display a strong bias against incorporation of distal V gene segments. Furthermore, antisense transcription at PAX5-activated intergenic repeat (PAIR) elements, located in the distal region of the Igh locus, depends on ERG. These findings show that ERG serves as a critical regulator of B cell development by ensuring efficient and balanced V-to-DJ recombination.


Asunto(s)
Linfocitos B/metabolismo , Cadenas Pesadas de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Humanos , Regulador Transcripcional ERG/metabolismo
10.
Cell Rep ; 23(9): 2744-2757, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29847803

RESUMEN

Transcription factors PU.1 and CEBPA are required for the proper coordination of enhancer activity during granulocytic-monocytic (GM) lineage differentiation to form myeloid cells. However, precisely how these factors control the chronology of enhancer establishment during differentiation is not known. Through integrated analyses of enhancer dynamics, transcription factor binding, and proximal gene expression during successive stages of murine GM-lineage differentiation, we unravel the distinct kinetics by which PU.1 and CEBPA coordinate GM enhancer activity. We find no evidence of a pioneering function of PU.1 during late GM-lineage differentiation. Instead, we delineate a set of enhancers that gain accessibility in a CEBPA-dependent manner, suggesting a pioneering function of CEBPA. Analyses of Cebpa null bone marrow demonstrate that CEBPA controls PU.1 levels and, unexpectedly, that the loss of CEBPA results in an early differentiation block. Taken together, our data provide insights into how PU.1 and CEBPA functionally interact to drive GM-lineage differentiation.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/deficiencia , Diferenciación Celular/genética , Elementos de Facilitación Genéticos/genética , Células Mieloides/citología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Línea Celular , Linaje de la Célula , Cromatina/metabolismo , Femenino , Regulación de la Expresión Génica , Granulocitos/citología , Granulocitos/metabolismo , Ratones , Monocitos/citología , Monocitos/metabolismo , Células Mieloides/metabolismo , Unión Proteica
11.
Cell Rep ; 24(3): 766-780, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30021172

RESUMEN

Hematopoietic stem cells (HSCs) are considered a heterogeneous cell population. To further resolve the HSC compartment, we characterized a retinoic acid (RA) reporter mouse line. Sub-fractionation of the HSC compartment in RA-CFP reporter mice demonstrated that RA-CFP-dim HSCs were largely non-proliferative and displayed superior engraftment potential in comparison with RA-CFP-bright HSCs. Gene expression analysis demonstrated higher expression of RA-target genes in RA-CFP-dim HSCs, in contrast to the RA-CFP reporter expression, but both RA-CFP-dim and RA-CFP-bright HSCs responded efficiently to RA in vitro. Single-cell RNA sequencing (RNA-seq) of >1,200 HSCs showed that differences in cell cycle activity constituted the main driver of transcriptional heterogeneity in HSCs. Moreover, further analysis of the single-cell RNA-seq data revealed that stochastic low-level expression of distinct lineage-affiliated transcriptional programs is a common feature of HSCs. Collectively, this work demonstrates the utility of the RA-CFP reporter line as a tool for the isolation of superior HSCs.


Asunto(s)
Compartimento Celular , Ciclo Celular/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Transcripción Genética , Animales , Ciclo Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Genoma , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Proteínas Luminiscentes/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcriptoma/genética , Tretinoina/farmacología
12.
PLoS One ; 9(1): e84728, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24404186

RESUMEN

Ageing and cancer is often associated with altered T cell distributions and this phenomenon has been suggested to be the main driver in the development of immunosenescence. Memory phenotype PD-1+ CD4+ T cells accumulate with age and during leukemic development, and they might account for the attenuated T cell response in elderly or diseased individuals. The transcription factor C/EBPα has been suggested to be responsible for the accumulation as well as for the senescent features of these cells including impaired TCR signaling and decreased proliferation. Thus modulating the activity of C/EBPα could potentially target PD-1+ CD4+ T cells and consequently, impede the development of immunosenescence. To exploit this possibility we tested the importance of C/EBPα for the development of age-dependent PD-1+ CD4+ T cells as well as its role in the accumulation of PD-1+ CD4+ T cells during leukemic progression. In contrast to earlier suggestions, we find that loss of C/EBPα expression in the lymphoid compartment led to an increase of PD-1+ CD4+ T cells specifically in old mice, suggesting that C/EBPα repress the accumulation of these cells in elderly by inhibiting their proliferation. Furthermore, C/EBPα-deficiency in the lymphoid compartment had no effect on leukemic development and did not affect the accumulation of PD-1+ CD4+ T cells. Thus, in addition to contradict earlier suggestions of a role for C/EBPα in immunosenescence, these findings efficiently discard the potential of using C/EBPα as a target for the alleviation of ageing/cancer-associated immunosenescence.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Memoria Inmunológica , Receptor de Muerte Celular Programada 1/metabolismo , Factores de Edad , Envejecimiento , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Senescencia Celular , Progresión de la Enfermedad , Expresión Génica , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo , Leucemia/mortalidad , Activación de Linfocitos , Ratones , Bazo/inmunología , Bazo/metabolismo
13.
J Exp Med ; 211(1): 5-13, 2014 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-24367003

RESUMEN

MLL-fusion proteins are potent inducers of oncogenic transformation, and their expression is considered to be the main oncogenic driving force in ∼10% of human acute myeloid leukemia (AML) patients. These oncogenic fusion proteins are responsible for the initiation of a downstream transcriptional program leading to the expression of factors such as MEIS1 and HOXA9, which in turn can replace MLL-fusion proteins in overexpression experiments. To what extent MLL fusion proteins act on their own during tumor initiation, or if they collaborate with other transcriptional regulators, is unclear. Here, we have compared gene expression profiles from human MLL-rearranged AML to normal progenitors and identified the myeloid tumor suppressor C/EBPα as a putative collaborator in MLL-rearranged AML. Interestingly, we find that deletion of Cebpa rendered murine hematopoietic progenitors completely resistant to MLL-ENL-induced leukemic transformation, whereas C/EBPα was dispensable in already established AMLs. Furthermore, we show that Cebpa-deficient granulocytic-monocytic progenitors were equally resistant to transformation and that C/EBPα collaborates with MLL-ENL in the induction of a transcriptional program, which is also apparent in human AML. Thus, our studies demonstrate a key role of C/EBPα in MLL fusion-driven transformation and find that it sharply demarcates tumor initiation and maintenance.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Leucemia Mieloide Aguda/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/genética , Biología Computacional , Cartilla de ADN/genética , Citometría de Flujo , Eliminación de Gen , Perfilación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide , Proteínas de Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa
14.
PLoS One ; 7(6): e38841, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22715416

RESUMEN

BACKGROUND: Transcription factors play a key role in lineage commitment and differentiation of stem cells into distinct mature cells. In hematopoiesis, they regulate lineage-specific gene expression in a stage-specific manner through various physical and functional interactions with regulatory proteins that are simultanously recruited and activated to ensure timely gene expression. The transcription factor CCAAT/enhancer binding protein α (C/EBPα) is such a factor and is essential for the development of granulocytic/monocytic cells. The activity of C/EBPα is regulated on several levels including gene expression, alternative translation, protein interactions and posttranslational modifications, such as phosphorylation. In particular, the phosphorylation of serine 248 of the transactivation domain has been shown to be of crucial importance for granulocytic differentiation of 32Dcl3 cells in vitro. METHODOLOGY/PRINCIPAL FINDINGS: Here, we use mouse genetics to investigate the significance of C/EBPα serine 248 in vivo through the construction and analysis of Cebpa(S248A/S248A) knock-in mice. Surprisingly, 8-week old Cebpa(S248A/S248A) mice display normal steady-state hematopoiesis including unaltered development of mature myeloid cells. However, over time some of the animals develop a hematopoietic disorder with accumulation of multipotent, megakaryocytic and erythroid progenitor cells and a mild impairment of differentiation along the granulocytic-monocytic lineage. Furthermore, BM cells from Cebpa(S248A/S248A) animals display a competitive advantage compared to wild type cells in a transplantation assay. CONCLUSIONS/SIGNIFICANCE: Taken together, our data shows that the substitution of C/EBPα serine 248 to alanine favors the selection of the megakaryocytic/erythroid lineage over the monocytic/granulocytic compartment in old mice and suggests that S248 phosphorylation may be required to maintain proper hematopoietic homeostasis in response to changes in the wiring of cellular signalling networks. More broadly, the marked differences between the phenotype of the S248A variant in vivo and in vitro highlight the need to exert caution when extending in vitro phenotypes to the more appropriate in vivo context.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Precursoras Eritroides/metabolismo , Células Progenitoras de Megacariocitos/metabolismo , Mutación Missense , Mielopoyesis , Trastornos Mieloproliferativos/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular , Células Precursoras Eritroides/patología , Células Progenitoras de Megacariocitos/patología , Ratones , Ratones Mutantes , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Fosforilación , Serina/genética , Serina/metabolismo
15.
Blood ; 111(8): 4309-21, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18212247

RESUMEN

The CCAAT enhancer binding protein alpha (C/EBPalpha) is an important myeloid tumor suppressor that is frequently mutated in human acute myeloid leukemia (AML). We have previously shown that mice homozygous for the E2F repression-deficient Cebpa(BRM2) allele develop nonfatal AML with long latency and incomplete penetrance, suggesting that accumulation of secondary mutations is necessary for disease progression. Here, we use SRS19-6-driven retroviral insertional mutagenesis to compare the phenotypes of leukemias arising in Cebpa(+/+), Cebpa(+/BRM2), and Cebpa(BRM2/BRM2) mice, with respect to disease type, latency of tumor development, and identity of the retroviral insertion sites (RISs). Both Cebpa(+/BRM2) and Cebpa(BRM2/BRM2) mice preferentially develop myeloid leukemias, but with differing latencies, thereby demonstrating the importance of gene dosage. Determination of RISs led to the identification of several novel candidate oncogenes, some of which may collaborate specifically with the E2F repression-deficient allele of Cebpa. Finally, we used an in silico pathway analysis approach to extract additional information from single RISs, leading to the identification of signaling pathways which were preferentially deregulated in a disease- and/or genotype-specific manner.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Predisposición Genética a la Enfermedad , Leucemia Mieloide/genética , Leucemia Mieloide/patología , Mutagénesis Insercional , Mutación/genética , Retroviridae/genética , Alelos , Animales , Células Clonales , Biología Computacional , Regulación Leucémica de la Expresión Génica , Reordenamiento Génico , Genes Relacionados con las Neoplasias , Inestabilidad Genómica , Inmunoglobulinas/genética , Inyecciones , Ratones , Ratones Endogámicos C57BL , Fenotipo , Lesiones Precancerosas/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Retroviridae/fisiología , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA