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.
Genes Dev ; 38(5-6): 253-272, 2024 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-38565249

RESUMEN

Oncogenic activation of MYC in cancers predominantly involves increased transcription rather than coding region mutations. However, MYC-dependent lymphomas frequently acquire point mutations in the MYC phosphodegron, including at threonine 58 (T58), where phosphorylation permits binding via the FBW7 ubiquitin ligase triggering MYC degradation. To understand how T58 phosphorylation functions in normal cell physiology, we introduced an alanine mutation at T58 (T58A) into the endogenous c-Myc locus in the mouse germline. While MYC-T58A mice develop normally, lymphomas and myeloid leukemias emerge in ∼60% of adult homozygous T58A mice. We found that primitive hematopoietic progenitor cells from MYC-T58A mice exhibit aberrant self-renewal normally associated with hematopoietic stem cells (HSCs) and up-regulate a subset of MYC target genes important in maintaining stem/progenitor cell balance. In lymphocytes, genomic occupancy by MYC-T58A was increased at all promoters compared with WT MYC, while genes differentially expressed in a T58A-dependent manner were significantly more proximal to MYC-bound enhancers. MYC-T58A lymphocyte progenitors exhibited metabolic alterations and decreased activation of inflammatory and apoptotic pathways. Our data demonstrate that a single point mutation stabilizing MYC is sufficient to skew target gene expression, producing a profound gain of function in multipotential hematopoietic progenitors associated with self-renewal and initiation of lymphomas and leukemias.


Asunto(s)
Proteína 7 que Contiene Repeticiones F-Box-WD , Neoplasias Hematológicas , Linfoma , Proteínas Proto-Oncogénicas c-myc , Animales , Ratones , Células Germinativas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Mutación Puntual , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo
2.
bioRxiv ; 2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37961183

RESUMEN

Oncogenic activation of MYC in cancers predominantly involves increased transcription rather than coding region mutations. However, MYC-dependent lymphomas frequently contain point mutations in the MYC phospho-degron, including at threonine-58 (T58), where phosphorylation permits binding by the FBW7 ubiquitin ligase triggering MYC degradation. To understand how T58 phosphorylation functions in normal cell physiology, we introduced an alanine mutation at T58 (T58A) into the endogenous c-Myc locus in the mouse germline. While MYC-T58A mice develop normally, lymphomas and myeloid leukemias emerge in ~60% of adult homozygous T58A mice. We find that primitive hematopoietic progenitor cells from MYC-T58A mice exhibit aberrant self-renewal normally associated with hematopoietic stem cells (HSCs) and upregulate a subset of Myc target genes important in maintaining stem/progenitor cell balance. Genomic occupancy by MYC-T58A was increased at all promoters, compared to WT MYC, while genes differentially expressed in a T58A-dependent manner were significantly more proximal to MYC-bound enhancers. MYC-T58A lymphocyte progenitors exhibited metabolic alterations and decreased activation of inflammatory and apoptotic pathways. Our data demonstrate that a single point mutation in Myc is sufficient to produce a profound gain of function in multipotential hematopoietic progenitors associated with self-renewal and initiation of lymphomas and leukemias.

3.
bioRxiv ; 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37333272

RESUMEN

Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.

4.
Nat Commun ; 13(1): 1584, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35332125

RESUMEN

Hematopoietic stem cells (HSCs) develop from hemogenic endothelium within embryonic arterial vessels such as the aorta of the aorta-gonad-mesonephros region (AGM). To identify the signals responsible for HSC formation, here we use single cell RNA-sequencing to simultaneously analyze the transcriptional profiles of AGM-derived cells transitioning from hemogenic endothelium to HSCs, and AGM-derived endothelial cells which provide signals sufficient to support HSC maturation and self-renewal. Pseudotemporal ordering reveals dynamics of gene expression during the hemogenic endothelium to HSC transition, identifying surface receptors specifically expressed on developing HSCs. Transcriptional profiling of niche endothelial cells identifies corresponding ligands, including those signaling to Notch receptors, VLA-4 integrin, and CXCR4, which, when integrated in an engineered platform, are sufficient to support the generation of engrafting HSCs. These studies provide a transcriptional map of the signaling interactions necessary for the development of HSCs and advance the goal of engineering HSCs for therapeutic applications.


Asunto(s)
Hemangioblastos , Transcriptoma , Gónadas , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Mesonefro
5.
Cell Rep ; 36(11): 109675, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525376

RESUMEN

During embryogenesis, waves of hematopoietic progenitors develop from hemogenic endothelium (HE) prior to the emergence of self-renewing hematopoietic stem cells (HSCs). Although previous studies have shown that yolk-sac-derived erythromyeloid progenitors and HSCs emerge from distinct populations of HE, it remains unknown whether the earliest lymphoid-competent progenitors, multipotent progenitors, and HSCs originate from common HE. In this study, we demonstrate by clonal assays and single-cell transcriptomics that rare HE with functional HSC potential in the early murine embryo are distinct from more abundant HE with multilineage hematopoietic potential that fail to generate HSCs. Specifically, HSC-competent HE are characterized by expression of CXCR4 surface marker and by higher expression of genes tied to arterial programs regulating HSC dormancy and self-renewal. Taken together, these findings suggest a revised model of developmental hematopoiesis in which the initial populations of multipotent progenitors and HSCs arise independently from HE with distinct phenotypic and transcriptional properties.


Asunto(s)
Hemangioblastos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Multipotentes/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Autorrenovación de las Células/genética , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Femenino , Hemangioblastos/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transcripción Genética
6.
Stem Cell Reports ; 16(4): 717-726, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33770495

RESUMEN

T cell development is restricted to the thymus and is dependent on high levels of Notch signaling induced within the thymic microenvironment. To understand Notch function in thymic restriction, we investigated the basis for target gene selectivity in response to quantitative differences in Notch signal strength, focusing on the chromatin architecture of genes essential for T cell differentiation. We find that high Notch signal strength is required to activate promoters of known targets essential for T cell commitment, including Il2ra, Cd3ε, and Rag1, which feature low CpG content (LCG) and DNA inaccessibility in hematopoietic stem progenitor cells. Our findings suggest that promoter DNA inaccessibility at LCG T lineage genes provides robust protection against stochastic activation in inappropriate Notch signaling contexts, limiting T cell development to the thymus.


Asunto(s)
Islas de CpG/genética , Regiones Promotoras Genéticas/genética , Receptores Notch/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Animales , ADN/metabolismo , Desoxirribonucleasa I/metabolismo , Ratones Endogámicos C57BL
7.
J Vis Exp ; (135)2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29806841

RESUMEN

The ability to study hematopoietic stem cell (HSC) genesis during embryonic development has been limited by the rarity of HSC precursors in the early embryo and the lack of assays that functionally identify the long-term multilineage engraftment potential of individual putative HSC precursors. Here, we describe methodology that enables the isolation and characterization of functionally validated HSC precursors at the single cell level. First, we utilize index sorting to catalog the precise phenotypic parameter of each individually sorted cell, using a combination of phenotypic markers to enrich for HSC precursors with additional markers for experimental analysis. Second, each index-sorted cell is co-cultured with vascular niche stroma from the aorta-gonad-mesonephros (AGM) region, which supports the maturation of non-engrafting HSC precursors to functional HSC with multilineage, long-term engraftment potential in transplantation assays. This methodology enables correlation of phenotypic properties of clonal hemogenic precursors with their functional engraftment potential or other properties such as transcriptional profile, providing a means for the detailed analysis of HSC precursor development at the single cell level.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Separación Celular , Células Cultivadas , Células Endoteliales/citología , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Embarazo
8.
Stem Cell Reports ; 8(6): 1563-1572, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28479303

RESUMEN

Recent evidence points to the embryonic emergence of some tissue-resident innate immune cells, such as B-1a lymphocytes, prior to and independently of hematopoietic stem cells (HSCs). However, whether the full hematopoietic repertoire of embryonic HSCs initially includes these unique lineages of innate immune cells has been difficult to assess due to lack of clonal assays that identify and assess HSC precursor (pre-HSC) potential. Here, by combining index sorting of single embryonic hemogenic precursors with in vitro HSC maturation and transplantation assays, we analyze emerging pre-HSCs at the single-cell level, revealing their unique stage-specific properties and clonal lineage potential. Remarkably, clonal pre-HSCs detected between E9.5 and E11.5 contribute to the complete B cell repertoire, including B-1a lymphocytes, revealing a previously unappreciated common precursor for all B cell lineages at the pre-HSC stage and a second embryonic origin for B-1a lymphocytes.


Asunto(s)
Linfocitos B/metabolismo , Células Madre Hematopoyéticas/citología , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/metabolismo , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/citología , Cadherinas/metabolismo , Proteínas de Unión al Calcio , Células Cultivadas , Técnicas de Cocultivo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Femenino , Citometría de Flujo , Factores de Intercambio de Guanina Nucleótido/genética , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL
9.
Development ; 142(14): 2452-63, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26062937

RESUMEN

Although Notch1 and Notch2 are closely related paralogs and function through the same canonical signaling pathway, they contribute to different outcomes in some cell and disease contexts. To understand the basis for these differences, we examined in detail mice in which the Notch intracellular domains (N1ICD and N2ICD) were swapped. Our data indicate that strength (defined here as the ultimate number of intracellular domain molecules reaching the nucleus, integrating ligand-mediated release and nuclear translocation) and duration (half-life of NICD-RBPjk-MAML-DNA complexes, integrating cooperativity and stability dependent on shared sequence elements) are the factors that underlie many of the differences between Notch1 and Notch2 in all the contexts we examined, including T-cell development, skin differentiation and carcinogenesis, the inner ear, the lung and the retina. We were able to show that phenotypes in the heart, endothelium, and marginal zone B cells are attributed to haploinsufficiency but not to intracellular domain composition. Tissue-specific differences in NICD stability were most likely caused by alternative scissile bond choices by tissue-specific γ-secretase complexes following the intracellular domain swap. Reinterpretation of clinical findings based on our analyses suggests that differences in outcome segregating with Notch1 or Notch2 are likely to reflect outcomes dependent on the overall strength of Notch signals.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Receptor Notch1/metabolismo , Receptor Notch2/metabolismo , Alelos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Carcinogénesis , Diferenciación Celular , Separación Celular , Oído Interno/embriología , Femenino , Citometría de Flujo , Cardiopatías Congénitas , Homocigoto , Pulmón/embriología , Masculino , Ratones , Fenotipo , Estructura Terciaria de Proteína , Retina/embriología , Transducción de Señal , Piel/embriología , Neoplasias Cutáneas/metabolismo , Linfocitos T/citología , Transcriptoma
10.
J Clin Invest ; 125(5): 2032-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25866967

RESUMEN

Hematopoietic stem cells (HSCs) first emerge during embryonic development within vessels such as the dorsal aorta of the aorta-gonad-mesonephros (AGM) region, suggesting that signals from the vascular microenvironment are critical for HSC development. Here, we demonstrated that AGM-derived endothelial cells (ECs) engineered to constitutively express AKT (AGM AKT-ECs) can provide an in vitro niche that recapitulates embryonic HSC specification and amplification. Specifically, nonengrafting embryonic precursors, including the VE-cadherin-expressing population that lacks hematopoietic surface markers, cocultured with AGM AKT-ECs specified into long-term, adult-engrafting HSCs, establishing that a vascular niche is sufficient to induce the endothelial-to-HSC transition in vitro. Subsequent to hematopoietic induction, coculture with AGM AKT-ECs also substantially increased the numbers of HSCs derived from VE-cadherin⁺CD45⁺ AGM hematopoietic cells, consistent with a role in supporting further HSC maturation and self-renewal. We also identified conditions that included NOTCH activation with an immobilized NOTCH ligand that were sufficient to amplify AGM-derived HSCs following their specification in the absence of AGM AKT-ECs. Together, these studies begin to define the critical niche components and resident signals required for HSC induction and self-renewal ex vivo, and thus provide insight for development of defined in vitro systems targeted toward HSC generation for therapeutic applications.


Asunto(s)
Aorta/embriología , Células Endoteliales/fisiología , Endotelio Vascular/embriología , Gónadas/embriología , Sistema Hematopoyético/embriología , Mesonefro/embriología , Receptor Notch1/fisiología , Receptor Notch2/fisiología , Nicho de Células Madre/fisiología , Animales , Antígenos CD/análisis , Cadherinas/análisis , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Endotelio Vascular/citología , Femenino , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Péptidos y Proteínas de Señalización Intracelular/fisiología , Antígenos Comunes de Leucocito/análisis , Masculino , Proteínas de la Membrana/fisiología , Ratones Congénicos , Ratones Endogámicos C57BL , Quimera por Radiación , Transducción de Señal , Células del Estroma/fisiología
11.
Blood ; 119(8): 1856-60, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22219225

RESUMEN

SIRT1 is an NAD(+)-dependent histone deacetylase implicated in the establishment of the primitive hematopoietic system during mouse embryonic development. However, investigation of the role of SIRT1 in adult hematopoiesis has been complicated by the high perinatal mortality of SIRT1-deficient mice (SIRT1(-/-)). We performed a comprehensive in vivo study of the hematopoietic stem cell (HSC) compartment in adult SIRT1(-/-) mice and show that, apart from anemia and leukocytosis in older mice, the production of mature blood cells, lineage distribution within hematopoietic organs, and frequencies of the most primitive HSC populations are comparable to those of wild-type littermate controls. Furthermore, we show that SIRT1-deficient BM cells confer stable long-term reconstitution in competitive repopulation and serial transplantation experiments. The results of the present study rule out an essential physiologic role for cell-autonomous SIRT1 signaling in the maintenance of the adult HSC compartment in mice.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Sirtuina 1/fisiología , Factores de Edad , Animales , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Recuento de Células Sanguíneas , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Citometría de Flujo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Superficie Celular/metabolismo , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Sirtuina 1/deficiencia , Sirtuina 1/genética , Factores de Tiempo
12.
J Clin Invest ; 121(3): 1207-16, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21285514

RESUMEN

HSCs either self-renew or differentiate to give rise to multipotent cells whose progeny provide blood cell precursors. However, surprisingly little is known about the factors that regulate this choice of self-renewal versus differentiation. One candidate is the Notch signaling pathway, with ex vivo studies suggesting that Notch regulates HSC differentiation, although a functional role for Notch in HSC self-renewal in vivo remains controversial. Here, we have shown that Notch2, and not Notch1, inhibits myeloid differentiation and enhances generation of primitive Sca-1(+)c-kit(+) progenitors following in vitro culture of enriched HSCs with purified Notch ligands. In mice, Notch2 enhanced the rate of formation of short-term repopulating multipotential progenitor cells (MPPs) as well as long-term repopulating HSCs, while delaying myeloid differentiation in BM following injury. However, consistent with previous reports, once homeostasis was achieved, neither Notch1 nor Notch2 affected repopulating cell self-renewal. These data indicate a Notch2-dependent role in assuring orderly repopulation by HSCs, MPPs, myeloid cells, and lymphoid cells during BM regeneration.


Asunto(s)
Receptor Notch2/metabolismo , Células Madre/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Células Cultivadas , Ligandos , Ratones , Ratones Transgénicos , Microscopía Fluorescente/métodos , Receptor Notch1/metabolismo , Regeneración , Transducción de Señal
13.
Cell Stem Cell ; 6(3): 251-64, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20207228

RESUMEN

Bone marrow endothelial cells (ECs) are essential for reconstitution of hematopoiesis, but their role in self-renewal of long-term hematopoietic stem cells (LT-HSCs) is unknown. We have developed angiogenic models to demonstrate that EC-derived angiocrine growth factors support in vitro self-renewal and in vivo repopulation of authentic LT-HSCs. In serum/cytokine-free cocultures, ECs, through direct cellular contact, stimulated incremental expansion of repopulating CD34(-)Flt3(-)cKit(+)Lineage(-)Sca1(+) LT-HSCs, which retained their self-renewal ability, as determined by single-cell and serial transplantation assays. Angiocrine expression of Notch ligands by ECs promoted proliferation and prevented exhaustion of LT-HSCs derived from wild-type, but not Notch1/Notch2-deficient, mice. In transgenic notch-reporter (TNR.Gfp) mice, regenerating TNR.Gfp(+) LT-HSCs were detected in cellular contact with sinusoidal ECs. Interference with angiocrine, but not perfusion, function of SECs impaired repopulation of TNR.Gfp(+) LT-HSCs. ECs establish an instructive vascular niche for clinical-scale expansion of LT-HSCs and a cellular platform to identify stem cell-active trophogens.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Transducción de Señal , Animales , Comunicación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Ligandos , Ratones , Ratones Noqueados , Receptor Notch1/deficiencia , Receptor Notch1/metabolismo , Receptor Notch2/deficiencia , Receptor Notch2/metabolismo
14.
Cell Stem Cell ; 3(6): 611-24, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-19041778

RESUMEN

Myc activity is emerging as a key element in acquisition and maintenance of stem cell properties. We have previously shown that c-Myc deficiency results in accumulation of defective hematopoietic stem cells (HSCs) due to niche-dependent differentiation defects. Here we report that immature HSCs coexpress c-myc and N-myc mRNA at similar levels. Although conditional deletion of N-myc in the bone marrow does not affect hematopoiesis, combined deficiency of c-Myc and N-Myc (dKO) results in pancytopenia and rapid lethality. Interestingly, proliferation of HSCs depends on both myc genes during homeostasis, but is c-Myc/N-Myc independent during bone marrow repair after injury. Strikingly, while most dKO hematopoietic cells undergo apoptosis, only self-renewing HSCs accumulate the cytotoxic molecule Granzyme B, normally employed by the innate immune system, thereby revealing an unexpected mechanism of stem cell apoptosis. Collectively, Myc activity (c-Myc and N-Myc) controls crucial aspects of HSC function including proliferation, differentiation, and survival.


Asunto(s)
Diferenciación Celular/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Linaje de la Célula/genética , Proliferación Celular , Supervivencia Celular/genética , Células Cultivadas , Supervivencia de Injerto/genética , Granzimas/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Pancitopenia/genética , Pancitopenia/fisiopatología , Transducción de Señal/genética , Estrés Fisiológico/genética
15.
J Immunol ; 181(9): 5885-94, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941177

RESUMEN

Little is known about the transcriptional regulators that control the proliferation of multipotent bone marrow progenitors. Understanding the mechanisms that restrict proliferation is of significant interest since the loss of cell cycle integrity can be associated with hematopoietic exhaustion, bone marrow failure, or even oncogenic transformation. Herein, we show that multipotent LSKs (lineage(-)Sca(high)c-kit(+)) from E47-deficient mice exhibit a striking hyperproliferation associated with a loss of cell cycle quiescence and increased susceptibility to in vivo challenge with a mitotoxic drug. Total LSKs contain long-term self-renewing hematopoietic stem cells and downstream multipotential progenitors (MPPs) that possess very limited or no self-renewal ability. Within total LSKs, we found specific developmental and functional deficits in the MPP subset. E47 knockout mice have grossly normal numbers of self-renewing hematopoietic stem cells but a 50-70% reduction in nonrenewing MPPs and downstream lineage-restricted populations. The residual MPPs in E47 knockout mice fail to fully up-regulate flk2 or initiate V(D)J recombination, hallmarks of normal lymphoid lineage progression. Consistent with the loss of normal cell cycle restraints, we show that E47-deficient LSKs have a 50% decrease in p21, a cell cycle inhibitor and known regulator of LSK proliferation. Moreover, enforced expression studies identify p21 as an E47 target gene in primary bone marrow LSKs. Thus, E47 appears to regulate the developmental and functional integrity of early hematopoietic subsets in part through effects on p21-mediated cell cycle quiescence.


Asunto(s)
Ciclo Celular/inmunología , Diferenciación Celular/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Multipotentes/metabolismo , Fase de Descanso del Ciclo Celular/inmunología , Factores de Transcripción TCF/fisiología , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Marcación de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Fase de Descanso del Ciclo Celular/genética , Factores de Transcripción TCF/biosíntesis , Factores de Transcripción TCF/deficiencia , Factores de Transcripción TCF/genética , Proteína 1 Similar al Factor de Transcripción 7 , Transducción Genética
16.
Genes Dev ; 22(12): 1677-89, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559482

RESUMEN

Phosphorylations within N- and C-terminal degrons independently control the binding of cyclin E to the SCF(Fbw7) and thus its ubiquitination and proteasomal degradation. We have now determined the physiologic significance of cyclin E degradation by this pathway. We describe the construction of a knockin mouse in which both degrons were mutated by threonine to alanine substitutions (cyclin E(T74A T393A)) and report that ablation of both degrons abolished regulation of cyclin E by Fbw7. The cyclin E(T74A T393A) mutation disrupted cyclin E periodicity and caused cyclin E to continuously accumulate as cells reentered the cell cycle from quiescence. In vivo, the cyclin E(T74A T393A) mutation greatly increased cyclin E activity and caused proliferative anomalies. Cyclin E(T74A T393A) mice exhibited abnormal erythropoiesis characterized by a large expansion of abnormally proliferating progenitors, impaired differentiation, dysplasia, and anemia. This syndrome recapitulates many features of early stage human refractory anemia/myelodysplastic syndrome, including ineffective erythropoiesis. Epithelial cells also proliferated abnormally in cyclin E knockin mice, and the cyclin E(T74A T393A) mutation delayed mammary gland involution, implicating cyclin E degradation in this anti-mitogenic response. Hyperproliferative mammary epithelia contained increased apoptotic cells, suggesting that apoptosis contributes to tissue homeostasis in the setting of cyclin E deregulation. Overall these data show the critical role of both degrons in regulating cyclin E activity and reveal that complete loss of Fbw7-mediated cyclin E degradation causes spontaneous and cell type-specific proliferative anomalies.


Asunto(s)
Proliferación Celular , Ciclina E/metabolismo , Ciclina E/fisiología , Células Epiteliales/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Linaje de la Célula/fisiología , Células Cultivadas , Ciclina E/genética , Células Epiteliales/metabolismo , Células Eritroides/patología , Eritropoyesis/genética , Femenino , Marcación de Gen , Enfermedades Hematológicas/genética , Células Madre Hematopoyéticas/metabolismo , Masculino , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Embarazo , Proteínas Quinasas/metabolismo , Proteínas Quinasas/fisiología , Procesamiento Proteico-Postraduccional/fisiología
17.
Blood ; 111(5): 2615-20, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18048645

RESUMEN

Notch signaling establishes boundaries in the thymus by inducing T-cell commitment and inhibiting a B-cell choice. Here, we show a significant 1.6-fold increased generation of B-cell precursors in thymuses from mice deficient for Notch target Hes5 compared with wild-type littermates. We further show that culture of bone marrow-derived progenitors with increasing densities of purified immobilized Notch ligand (Delta1(ext-IgG)) induced increased expression of Notch targets Hes1 and Hes5, and that although Hes5-deficient progenitors responded appropriately to high densities of ligand, they misread intermediate and low densities. Together, our results suggest that to ensure an appropriate outcome in the thymus in response to a lower threshold of induced Notch signaling, induction of the additional target Hes5 is required.


Asunto(s)
Linfocitos B/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Receptores Notch/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T/citología , Timo/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Recuento de Células , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Proteínas de la Membrana/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Células Madre/citología , Subgrupos de Linfocitos T/citología
18.
Stem Cells ; 25(10): 2488-97, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17641244

RESUMEN

The Wnt and Notch signaling pathways have been independently shown to play a critical role in regulating hematopoietic cell fate decisions. We previously reported that induction of Notch signaling in human CD34(+)CD38(-) cord blood cells by culture with the Notch ligand Delta 1 resulted in more cells with T or natural killer (NK) lymphoid precursor phenotype. Here, we show that addition of Wnt3a to Delta 1 further increased the percentage of CD34(-)CD7(+) and CD34(-)CD7(+)cyCD3(+) cells with increased expression of CD3 epsilon and preT alpha. In contrast, culture with Wnt3a alone did not increase generation of CD34(-)CD7(+) precursors or expression of CD3 epsilon or preT alpha gene. Furthermore, Wnt3a increased the amount of activated Notch1, suggesting that Wnt modulates Notch signaling by affecting Notch protein levels. In contrast, addition of a Wnt signaling inhibitor to Delta 1 increased the percentage of CD56(+) NK cells. Overall, these results demonstrate that regulation of Notch signaling by the Wnt pathway plays a critical role in differentiation of precursors along the early T or NK differentiation pathways. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Células Asesinas Naturales/citología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Linfocitos T/citología , Proteínas Wnt/fisiología , Antígenos CD/análisis , Diferenciación Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Citotoxicidad Inmunológica , Sangre Fetal/citología , Genes Reporteros , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/farmacología , Receptor Notch1/fisiología , Transducción de Señal/efectos de los fármacos , Transducción Genética , Proteínas Wnt/farmacología , Proteína Wnt3 , Proteína Wnt3A
19.
Blood ; 109(8): 3579-87, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17213287

RESUMEN

A physiologic role for Notch signaling in hematopoiesis has been clearly defined in lymphoid differentiation, with evidence suggesting a critical role in T-cell versus B-cell fate decisions. Previously, we demonstrated that activation of endogenous Notch receptors by culture of murine lin(-)Sca-1(+)c-kit(+) (LSK) hematopoietic progenitors with exogenously presented Notch ligand, Delta1(ext-IgG), consisting of the extracellular domain of Delta1 fused to the Fc domain of human IgG(1), promoted early T-cell differentiation and increased the number of progenitors capable of short-term lymphoid and myeloid reconstitution. Here we show that culture of LSK precursors with Delta1(ext-IgG) increases the number of progenitors that are able to rapidly repopulate the thymus and accelerate early T-cell reconstitution with a diversified T-cell receptor repertoire. Most of the early T-cell reconstitution originated from cells that expressed lymphoid-associated antigens: B220, Thy1, CD25, and/or IL7Ralpha, whereas the most efficient thymic repopulation on a per cell basis originated from the smaller number of cultured cells that did not express lymphoid-associated antigens. These findings demonstrate the potential of Delta1(ext-IgG)-cultured cells for accelerating early immune reconstitution after hematopoietic cell transplantation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Proteínas de la Membrana/farmacología , Receptores Notch/metabolismo , Recuperación de la Función/efectos de los fármacos , Linfocitos T/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Trasplante de Células Madre Hematopoyéticas , Inmunoglobulina G/genética , Inmunoglobulina G/farmacología , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Ratones , Estructura Terciaria de Proteína/genética , Receptores Notch/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T/citología , Quimera por Trasplante/metabolismo , Trasplante Homólogo
20.
Blood ; 106(8): 2693-9, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15976178

RESUMEN

Although significant advances have been made over the last decade with respect to our understanding of stem cell biology, progress has been limited in the development of successful techniques for clinically significant ex vivo expansion of hematopoietic stem and progenitor cells. We here describe the effect of Notch ligand density on induction of Notch signaling and subsequent cell fate of human CD34+CD38- cord blood progenitors. Lower densities of Delta1(ext-IgG) enhanced the generation of CD34+ cells as well as CD14+ and CD7+ cells, consistent with early myeloid and lymphoid differentiation, respectively. However, culture with increased amounts of Delta1(ext-IgG) induced apoptosis of CD34+ precursors resulting in decreased cell numbers, without affecting generation of CD7+ cells. RNA interference studies revealed that the promotion of lymphoid differentiation was primarily mediated by Delta1 activation of Notch1. Furthermore, enhanced generation of NOD/SCID repopulating cells was seen following culture with lower but not higher densities of ligand. These studies indicate critical, quantitative aspects of Notch signaling in affecting hematopoietic precursor cell-fate outcomes and suggest that density of Notch ligands in different organ systems may be an important determinant in regulating cell-fate outcomes. Moreover, these findings contribute to the development of methodology for manipulation of hematopoietic precursors for therapeutic purposes.


Asunto(s)
Diferenciación Celular , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Receptores de Superficie Celular/metabolismo , Receptores de Citocinas/metabolismo , Factores de Transcripción/metabolismo , Proliferación Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Ligandos , Receptor Notch1 , Receptor Notch2 , Receptores de Superficie Celular/genética , Transducción de Señal , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA