Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 143(10): 882-894, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38207291

RESUMEN

ABSTRACT: Ex vivo expansion of hematopoietic stem cells (HSCs) is gaining importance for cell and gene therapy, and requires a shift from dormancy state to activation and cycling. However, abnormal or excessive HSC activation results in reduced self-renewal ability and increased propensity for myeloid-biased differentiation. We now report that activation of the E3 ligase complex CRL3KBTBD4 by UM171 not only induces epigenetic changes through CoREST1 degradation but also controls chromatin-bound master regulator of cell cycle entry and proliferative metabolism (MYC) levels to prevent excessive activation and maintain lympho-myeloid potential of expanded populations. Furthermore, reconstitution activity and multipotency of UM171-treated HSCs are specifically compromised when MYC levels are experimentally increased despite degradation of CoREST1.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas/métodos , Ciclo Celular , Diferenciación Celular
2.
Cell ; 137(2): 369-79, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19379700

RESUMEN

Despite tremendous progress made toward the identification of the molecular circuitry that governs cell fate in embryonic stem cells, genes controlling this process in the adult hematopoietic stem cell have proven to be more difficult to unmask. We now report the results of a novel gain-of-function screening approach, which identified a series of 18 nuclear factors that affect hematopoietic stem cell activity. Overexpression of ten of these factors resulted in an increased repopulating activity compared to unmanipulated cells. Interestingly, at least four of the 18 factors, Fos, Tcfec, Hmgb1, and Sfpi1, show non-cell-autonomous functions. The utilization of this screening method together with the creation of a database enriched for potential determinants of hematopoietic stem cell self-renewal will serve as a resource to uncover regulatory networks in these cells.


Asunto(s)
Células Madre Adultas/citología , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/citología , Proteínas Nucleares/análisis , Células Madre Adultas/metabolismo , Animales , Células de la Médula Ósea , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Organismos Libres de Patógenos Específicos
3.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33692124

RESUMEN

Zinc finger (ZnF) proteins represent one of the largest families of human proteins, although most remain uncharacterized. Given that numerous ZnF proteins are able to interact with DNA and poly(ADP ribose), there is growing interest in understanding their mechanism of action in the maintenance of genome integrity. We now report that the ZnF protein E4F transcription factor 1 (E4F1) is an actor in DNA repair. Indeed, E4F1 is rapidly recruited, in a poly(ADP ribose) polymerase (PARP)-dependent manner, to DNA breaks and promotes ATR/CHK1 signaling, DNA-end resection, and subsequent homologous recombination. Moreover, we identify E4F1 as a regulator of the ATP-dependent chromatin remodeling SWI/SNF complex in DNA repair. E4F1 binds to the catalytic subunit BRG1/SMARCA4 and together with PARP-1 mediates its recruitment to DNA lesions. We also report that a proportion of human breast cancers show amplification and overexpression of E4F1 or BRG1 that are mutually exclusive with BRCA1/2 alterations. Together, these results reveal a function of E4F1 in the DNA damage response that orchestrates proper signaling and repair of double-strand breaks and document a molecular mechanism for its essential role in maintaining genome integrity and cell survival.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias de la Mama/genética , Proliferación Celular , Supervivencia Celular , Ensamble y Desensamble de Cromatina , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Recombinación Homóloga , Humanos , Unión Proteica , Proteínas Represoras/deficiencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia
4.
Blood ; 138(25): 2642-2654, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34499717

RESUMEN

Hematopoietic stem cells (HSCs) sustain blood cell homeostasis throughout life and can regenerate all blood lineages after transplantation. Despite this clear functional definition, highly enriched isolation of human HSCs can currently only be achieved through combinatorial assessment of multiple surface antigens. Although several transgenic HSC reporter mouse strains have been described, no analogous approach to prospectively isolate human HSCs has been reported. To identify genes with the most selective expression in human HSCs, we profiled population and single-cell transcriptomes of unexpanded and ex vivo cultured cord blood-derived hematopoietic stem and progenitor cells as well as peripheral blood, adult bone marrow, and fetal liver. On the basis of these analyses, we propose the master transcription factor HLF (hepatic leukemia factor) as one of the most specific HSC marker genes. To directly track its expression in human hematopoietic cells, we developed a genomic HLF reporter strategy, capable of selectively labeling the most immature blood cells on the basis of a single engineered parameter. Most importantly, HLF-expressing cells comprise all stem cell activity in culture and in vivo during serial transplantation. Taken together, these results experimentally establish HLF as a defining gene of the human HSC state and outline a new approach to continuously mark these cells with high fidelity.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Transcriptoma , Hematopoyesis , Células Madre Hematopoyéticas/citología , Humanos , Análisis de la Célula Individual
5.
Nucleic Acids Res ; 47(14): 7532-7547, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31219578

RESUMEN

Fanconi Anemia (FA) clinical phenotypes are heterogenous and rely on a mutation in one of the 22 FANC genes (FANCA-W) involved in a common interstrand DNA crosslink-repair pathway. A critical step in the activation of FA pathway is the monoubiquitination of FANCD2 and its binding partner FANCI. To better address the clinical phenotype associated with FANCI and the epistatic relationship with FANCD2, we created the first conditional inactivation model for FANCI in mouse. Fanci -/- mice displayed typical FA features such as delayed development in utero, microphtalmia, cellular sensitivity to mitomycin C, occasional limb abnormalities and hematological deficiencies. Interestingly, the deletion of Fanci leads to a strong meiotic phenotype and severe hypogonadism. FANCI was localized in spermatocytes and spermatids and in the nucleus of oocytes. Both FANCI and FANCD2 proteins co-localized with RPA along meiotic chromosomes, albeit at different levels. Consistent with a role in meiotic recombination, FANCI interacted with RAD51 and stimulated D-loop formation, unlike FANCD2. The double knockout Fanci-/- Fancd2-/- also showed epistatic relationship for hematological defects while being not epistatic with respect to generating viable mice in crosses of double heterozygotes. Collectively, this study highlights common and distinct functions of FANCI and FANCD2 during mouse development, meiotic recombination and hematopoiesis.


Asunto(s)
Reparación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oocitos/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Espermatocitos/metabolismo
6.
Genes Dev ; 26(7): 651-6, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22431509

RESUMEN

In this study, we show the high frequency of spontaneous γδ T-cell leukemia (T-ALL) occurrence in mice with biallelic deletion of enhancer of zeste homolog 2 (Ezh2). Tumor cells show little residual H3K27 trimethylation marks compared with controls. EZH2 is a component of the PRC2 Polycomb group protein complex, which is associated with DNA methyltransferases. Using next-generation sequencing, we identify alteration in gene expression levels of EZH2 and acquired mutations in PRC2-associated genes (DNMT3A and JARID2) in human adult T-ALL. Together, these studies document that deregulation of EZH2 and associated genes leads to the development of mouse, and likely human, T-ALL.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia-Linfoma de Células T del Adulto/metabolismo , Factores de Transcripción/metabolismo , Enfermedad Aguda , Animales , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética
7.
Blood ; 130(20): 2204-2214, 2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-28855157

RESUMEN

Neomorphic missense mutations affecting crucial lysine residues in histone H3 genes significantly contribute to a variety of solid cancers. Despite the high prevalence of H3K27M mutations in pediatric glioblastoma and their well-established impact on global histone H3 lysine 27 di- and trimethylation (H3K27me2/3), the relevance of these mutations has not been studied in acute myeloid leukemia (AML). Here, we report the first identification of H3K27M and H3K27I mutations in patients with AML. We find that these lesions are major determinants of reduced H3K27me2/3 in these patients and that they are associated with common aberrations in the RUNX1 gene. We demonstrate that H3K27I/M mutations are strong disease accelerators in a RUNX1-RUNX1T1 AML mouse model, suggesting that H3K27me2/3 has an important and selective leukemia-suppressive activity in this genetic context.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Histonas/genética , Leucemia Mieloide Aguda/genética , Mutación Missense , Transformación Genética , Adolescente , Anciano de 80 o más Años , Animales , Metilación de ADN , Femenino , Humanos , Lisina/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Proteína 1 Compañera de Translocación de RUNX1/genética , Análisis de Secuencia de ADN
8.
Blood ; 129(25): 3344-3351, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28408459

RESUMEN

A small subset of human cord blood CD34+ cells express endothelial protein C receptor (EPCR/CD201/PROCR) when exposed to the hematopoietic stem cell (HSC) self-renewal agonist UM171. In this article, we show that EPCR-positive UM171-treated cells, as opposed to EPCR-negative cells, exhibit robust multilineage repopulation and serial reconstitution ability in immunocompromised mice. In contrast to other stem cell markers, such as CD38, EPCR expression is maintained when cells are introduced in culture, irrespective of UM171 treatment. Although engineered overexpression of EPCR fails to reproduce the effects of UM171 on HSC activity, its expression is required for the repopulating activity of human HSCs. Altogether, our results indicate that EPCR is a reliable and cell culture-compatible marker of UM171-expanded human cord blood HSCs.


Asunto(s)
Antígenos CD34/análisis , Antígenos CD/análisis , Sangre Fetal/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Indoles/farmacología , Pirimidinas/farmacología , Receptores de Superficie Celular/análisis , Animales , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Receptor de Proteína C Endotelial , Femenino , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Humanos , Ratones Endogámicos NOD , Ratones SCID
9.
FASEB J ; 31(11): 5012-5018, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28754713

RESUMEN

The ubiquitin-associated protein 2-like (UBAP2L) gene remains poorly studied in human and mouse development. UBAP2L interacts with the Polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog (BMI1) and determines the activity of mouse hematopoietic stem cells in vivo Here we show that loss of Ubap2l leads to disorganized respiratory epithelium of mutant neonates, which die of respiratory failure. We also show that UBAP2L overexpression leads to epithelial-mesenchymal transition-like phenotype in a non-small cell lung carcinoma (NSCLC) cell line. UBAP2L is amplified in 15% of human primary lung adenocarcinoma specimens. Such patients express higher levels of UBAP2L and show a reduction in survival when compared with those who do not have this gene amplification. Supporting a possible role for UBAP2L in lung tumor progression, NSCLC cells engineered to express low levels of this gene produce much smaller tumors in vivo than wild-type control cells. Together, these results suggest that UBAP2L contributes to epithelial lung cell identity in mice and that it plays an important role in human lung adenocarcinoma.-Aucagne, R., Girard, S., Mayotte, N., Lehnertz, B., Lopes-Paciencia, S., Gendron, P., Boucher, G., Chagraoui, J., Sauvageau, G. UBAP2L is amplified in a large subset of human lung adenocarcinoma and is critical for epithelial lung cell identity and tumor metastasis.


Asunto(s)
Adenocarcinoma/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Portadoras/biosíntesis , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/biosíntesis , Mucosa Respiratoria/metabolismo , Células A549 , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Portadoras/genética , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Mucosa Respiratoria/patología
10.
Immunity ; 29(1): 57-67, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18617424

RESUMEN

Despite studies based on deletion or activation of intracellular components of the canonical Wingless related (Wnt) pathway, the role of Wnts in hematolymphopoiesis remains controversial. Using gain-of-function and loss-of-function models, we found that Wnt4 differentially affected diverse subsets of hematopoietic stem and progenitor cells. Bone-marrow and thymic Lin(-)Sca1(+)Kit(hi) cells (LSKs) were the key targets of Wnt4. In adult mice, Wnt4-induced expansion of Flt3(+) bone-marrow LSKs (lymphoid-primed multipotent progenitors) led to a sizeable accumulation of the most immature thymocyte subsets (upstream of beta-selection) and a major increase in thymopoiesis. Conversely, Wnt4(-/-) neonates showed low frequencies of bone-marrow LSKs and thymic hypocellularity. We provide compelling evidence that Wnt4 activates noncanonical (beta-catenin-independent) signaling and that its effects on hematopoietic cells are mainly non-cell-autonomous. Our work shows that Wnt4 overexpression has a unique ability to expand Flt3(+) LSKs in adults and demonstrates that noncanonical Wnt signaling regulates thymopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Multipotentes/citología , Transducción de Señal/inmunología , Timo/crecimiento & desarrollo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Diferenciación Celular/inmunología , Citometría de Flujo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Immunoblotting , Ratones , Células Madre Multipotentes/inmunología , Células Madre Multipotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/citología , Timo/inmunología , Proteínas Wnt/inmunología , Proteína Wnt4 , beta Catenina/inmunología
11.
Blood ; 124(15): 2362-9, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25185265

RESUMEN

Multipotent long-term repopulating hematopoietic stem cells (LT-HSCs) can self-renew or differentiate into the less primitive short-term repopulating stem cells (ST-HSCs), which themselves produce progenitors that ensure the daily supply of all essential blood components. The Polycomb group (PcG) protein BMI1 is essential for the activity of both HSCs and progenitor cells. Although BMI1 operates by suppressing the Ink4a/Arf locus in progenitors and ST-HSCs, the mechanisms through which this gene regulates the activity of LT-HSCs remain poorly understood. Toward this goal, we isolated BMI1-containing protein complexes and identified UBAP2L as a novel BMI1-interacting protein. We also showed that UBAP2L is preferentially expressed in mouse and human HSC-enriched populations when compared with more mature cell types, and that this gene is essential for the activity of LT-HSCs. In contrast to what is observed for Bmi1 knockdown, we found that UBAP2L depletion does not affect the Ink4a/Arf locus. Given that we demonstrated that BMI1 overexpression is able to rescue the deleterious effects of Ubap2l downregulation on LT-HSC activity and that UBAP2L is part of a PcG subcomplex comprising BMI1, we propose a model in which at least 2 different BMI1-containing PcG complexes regulate HSC activity, which are distinguishable by the presence of UBAP2L.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Madre Hematopoyéticas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Regulación hacia Abajo , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones Endogámicos C57BL , Proteínas del Grupo Polycomb/metabolismo , Unión Proteica , ARN Interferente Pequeño/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
12.
Blood ; 122(9): 1545-55, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23777767

RESUMEN

Histone methylation is a dynamic and reversible process proposed to directly impact on stem cell fate. The Jumonji (JmjC) domain-containing family of demethylases comprises 27 members that target mono-, di-, and trimethylated lysine residues of histone (or nonhistone) proteins. To evaluate their role in regulation of hematopoietic stem cell (HSC) behavior, we performed an in vivo RNAi-based functional screen and demonstrated that Jarid1b and Jhdm1f play opposing roles in regulation of HSC activity. Decrease in Jarid1b levels correlated with an in vitro expansion of HSCs with preserved long-term in vivo lymphomyeloid differentiation potential. Through RNA sequencing analysis, Jarid1b knockdown was associated with increased expression levels of several HSC regulators (Hoxa7, Hoxa9, Hoxa10, Hes1, Gata2) and reduced levels of differentiation-associated genes. shRNA against Jhdmlf, in contrast, impaired hematopoietic reconstitution of bone marrow cells. Together, our studies identified Jarid1b as a negative regulator of HSC activity and Jhdmlf as a positive regulator of HSC activity.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Histona Demetilasas con Dominio de Jumonji/fisiología , Interferencia de ARN/fisiología , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/fisiología , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Estudios de Validación como Asunto
13.
Blood ; 120(8): e17-27, 2012 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-22802335

RESUMEN

We recently generated 2 phenotypically similar Hoxa9+Meis1 overexpressing acute myeloid leukemias that differ by their in vivo biologic behavior. The first leukemia, named FLA2, shows a high frequency of leukemia stem cells (LSCs; 1 in 1.4 cells), whereas the second, FLB1, is more typical with a frequency of LSCs in the range of 1 per several hundred cells. To gain insights into possible mechanisms that determine LSC self-renewal, we profiled and compared the abundance of nuclear and cytoplasmic proteins and phosphoproteins from these leukemias using quantitative proteomics. These analyses revealed differences in proteins associated with stem cell fate, including a hyperactive p38 MAP kinase in FLB1 and a differentially localized Polycomb group protein Ezh2, which is mostly nuclear in FLA2 and predominantly cytoplasmic in FLB1. Together, these newly documented proteomes and phosphoproteomes represent a unique resource with more than 440 differentially expressed proteins and 11 543 unique phosphopeptides, of which 80% are novel and 7% preferentially phosphorylated in the stem cell-enriched leukemia.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Células Madre Neoplásicas/metabolismo , Proteoma/análisis , Proteoma/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Activación Enzimática , N-Metiltransferasa de Histona-Lisina/análisis , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Proteínas Represoras/análisis , Proteínas Represoras/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Blood ; 119(11): 2510-22, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22174158

RESUMEN

The stem cell-intrinsic model of self-renewal via asymmetric cell division (ACD) posits that fate determinants be partitioned unequally between daughter cells to either activate or suppress the stemness state. ACD is a purported mechanism by which hematopoietic stem cells (HSCs) self-renew, but definitive evidence for this cellular process remains open to conjecture. To address this issue, we chose 73 candidate genes that function within the cell polarity network to identify potential determinants that may concomitantly alter HSC fate while also exhibiting asymmetric segregation at cell division. Initial gene-expression profiles of polarity candidates showed high and differential expression in both HSCs and leukemia stem cells. Altered HSC fate was assessed by our established in vitro to in vivo screen on a subcohort of candidate polarity genes, which revealed 6 novel positive regulators of HSC function: Ap2a2, Gpsm2, Tmod1, Kif3a, Racgap1, and Ccnb1. Interestingly, live-cell videomicroscopy of the endocytic protein AP2A2 shows instances of asymmetric segregation during HSC/progenitor cell cytokinesis. These results contribute further evidence that ACD is functional in HSC self-renewal, suggest a role for Ap2a2 in HSC activity, and provide a unique opportunity to prospectively analyze progeny from HSC asymmetric divisions.


Asunto(s)
Complejo 2 de Proteína Adaptadora/metabolismo , Subunidades alfa de Complejo de Proteína Adaptadora/metabolismo , División Celular Asimétrica/fisiología , Polaridad Celular/genética , Endocitosis/genética , Células Madre Hematopoyéticas/citología , Células Madre Neoplásicas/patología , Células Madre/citología , Complejo 2 de Proteína Adaptadora/antagonistas & inhibidores , Complejo 2 de Proteína Adaptadora/genética , Subunidades alfa de Complejo de Proteína Adaptadora/antagonistas & inhibidores , Subunidades alfa de Complejo de Proteína Adaptadora/genética , Animales , Biomarcadores/metabolismo , Western Blotting , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Leucemia/metabolismo , Leucemia/patología , Ratones , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/fisiología
15.
Proc Natl Acad Sci U S A ; 108(13): 5284-9, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21402923

RESUMEN

BMI1 is a key component of multiprotein Polycomb repression complex 1 (PRC1), and its disruption in mice induces severe aplastic anemia by early adulthood. The contributing mechanisms responsible for this phenotype remain elusive. Here we show that transformed human cell lines as well as primitive hematopoietic cells exhibit a high frequency of spontaneous chromosome breaks upon BMI1 depletion and are hypersensitive to genotoxic agents. Consistent with these observations, we found that BMI1 is recruited rapidly to DNA damage foci where it blocks transcriptional elongation. We also show that BMI1 contributes to homologous recombination DNA repair and is required for checkpoint recovery. Taken together, our results suggest that BMI1 is critical for the maintenance of chromosome integrity in both normal and transformed cells.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Cromosomas/metabolismo , Daño del ADN , Reparación del ADN , Fibroblastos/citología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Proteínas Proto-Oncogénicas/genética , Recombinación Genética , Proteínas Represoras/genética
16.
Leukemia ; 38(3): 530-537, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38102204

RESUMEN

Monosomy 5 and deletions of the chromosome 5q (-5/del(5q)) are recurrent events in de novo adult acute myeloid leukemia (AML), reaching up to 40% of cases in secondary AML. These chromosome anomalies are associated with TP53 mutations and with very poor prognosis. Using the large Leucegene genomic and transcriptomic dataset composed of 48 -5/del(5q) patient specimens and 367 control AML, we identified DELE1 - located in the common deleted region - as the most consistently downregulated gene in these leukemias. DELE1 encodes a mitochondrial protein recently characterized as the relay of mitochondrial stress to the cytosol through a newly defined OMA1-DELE1-HRI pathway which ultimately leads to the activation of ATF4, the master transcription factor of the integrated stress response. Here, we showed that the partial loss of DELE1 expression observed in -5/del(5q) patients was sufficient to significantly reduce the sensitivity to mitochondrial stress in AML cells. Overall, our results suggest that DELE1 haploinsufficiency could represent a new driver mechanism in -5/del(5q) AML.


Asunto(s)
Haploinsuficiencia , Leucemia Mieloide Aguda , Proteínas Mitocondriales , Monosomía , Adulto , Humanos , Apoptosis/genética , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Leucemia Mieloide Aguda/genética , Proteínas Mitocondriales/genética
17.
Cell Rep ; 43(6): 114260, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38838225

RESUMEN

Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities.


Asunto(s)
Inmunoterapia , Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/patología , Inmunoterapia/métodos , Proteínas de la Membrana/metabolismo
18.
Blood Adv ; 8(1): 112-129, 2024 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-37729615

RESUMEN

ABSTRACT: Acute megakaryoblastic leukemia (AMKL) is a rare, developmentally restricted, and highly lethal cancer of early childhood. The paucity and hypocellularity (due to myelofibrosis) of primary patient samples hamper the discovery of cell- and genotype-specific treatments. AMKL is driven by mutually exclusive chimeric fusion oncogenes in two-thirds of the cases, with CBFA2T3::GLIS2 (CG2) and NUP98 fusions (NUP98r) representing the highest-fatality subgroups. We established CD34+ cord blood-derived CG2 models (n = 6) that sustain serial transplantation and recapitulate human leukemia regarding immunophenotype, leukemia-initiating cell frequencies, comutational landscape, and gene expression signature, with distinct upregulation of the prosurvival factor B-cell lymphoma 2 (BCL2). Cell membrane proteomic analyses highlighted CG2 surface markers preferentially expressed on leukemic cells compared with CD34+ cells (eg, NCAM1 and CD151). AMKL differentiation block in the mega-erythroid progenitor space was confirmed by single-cell profiling. Although CG2 cells were rather resistant to BCL2 genetic knockdown or selective pharmacological inhibition with venetoclax, they were vulnerable to strategies that target the megakaryocytic prosurvival factor BCL-XL (BCL2L1), including in vitro and in vivo treatment with BCL2/BCL-XL/BCL-W inhibitor navitoclax and DT2216, a selective BCL-XL proteolysis-targeting chimera degrader developed to limit thrombocytopenia in patients. NUP98r AMKL were also sensitive to BCL-XL inhibition but not the NUP98r monocytic leukemia, pointing to a lineage-specific dependency. Navitoclax or DT2216 treatment in combination with low-dose cytarabine further reduced leukemic burden in mice. This work extends the cellular and molecular diversity set of human AMKL models and uncovers BCL-XL as a therapeutic vulnerability in CG2 and NUP98r AMKL.


Asunto(s)
Antineoplásicos , Leucemia Megacarioblástica Aguda , Humanos , Niño , Preescolar , Animales , Ratones , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Leucemia Megacarioblástica Aguda/genética , Leucemia Megacarioblástica Aguda/patología , Proteómica , Factores de Transcripción , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Represoras
19.
PLoS Genet ; 6(12): e1001241, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21170304

RESUMEN

Understanding the function of important DNA elements in mammalian stem cell genomes would be enhanced by the availability of deletion collections in which segmental haploidies are precisely characterized. Using a modified Cre-loxP-based system, we now report the creation and characterization of a collection of ∼1,300 independent embryonic stem cell (ESC) clones enriched for nested chromosomal deletions. Mapping experiments indicate that this collection spans over 25% of the mouse genome with good representative coverage of protein-coding genes, regulatory RNAs, and other non-coding sequences. This collection of clones was screened for in vitro defects in differentiation of ESC into embryoid bodies (EB). Several putative novel haploinsufficient regions, critical for EB development, were identified. Functional characterization of one of these regions, through BAC complementation, identified the ribosomal gene Rps14 as a novel haploinsufficient determinant of embryoid body formation. This new library of chromosomal deletions in ESC (DelES: http://bioinfo.iric.ca/deles) will serve as a unique resource for elucidation of novel protein-coding and non-coding regulators of ESC activity.


Asunto(s)
Diferenciación Celular , Deleción Cromosómica , Células Madre Embrionarias/citología , Genoma , Mamíferos/genética , Animales , Línea Celular , Mapeo Cromosómico , Femenino , Humanos , Masculino , Ratones
20.
Blood ; 116(10): 1678-84, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20522713

RESUMEN

It is believed that hemopoietic stem cells (HSC), which colonize the fetal liver (FL) rapidly, expand to establish a supply of HSCs adequate for maintenance of hemopoiesis throughout life. Accordingly, FL HSCs are actively cycling as opposed to their predominantly quiescent bone marrow counterparts, suggesting that the FL microenvironment provides unique signals that support HSC proliferation and self-renewal. We now report the generation and characterization of mice with a mutant allele of Baf250a lacking exons 2 and 3. Baf250a(E2E3/E2E3) mice are viable until E19.5, but do not survive beyond birth. Most interestingly, FL HSC numbers are markedly higher in these mice than in control littermates, thus raising the possibility that Baf250a determines the HSC pool size in vivo. Limit dilution experiments indicate that the activity of Baf250a(E2E3/E2E3) HSC is equivalent to that of the wild-type counterparts. The Baf250a(E2E3/E2E3) FL-derived stroma, in contrast, exhibits a hemopoiesis-supporting potential superior to the developmentally matched controls. To our knowledge, this demonstration is the first that a mechanism operating in a cell nonautonomous manner canexpand the pool size of the fetal HSC populations.


Asunto(s)
Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/metabolismo , Hígado/metabolismo , Mutación , Proteínas Nucleares/genética , Alelos , Animales , Western Blotting , Recuento de Células , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Proteínas de Unión al ADN/metabolismo , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Hígado/embriología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Tiempo , Factores de Transcripción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA