Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nat Immunol ; 17(6): 666-676, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27043410

RESUMEN

According to current models of hematopoiesis, lymphoid-primed multi-potent progenitors (LMPPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)Flt3(hi)) and common myeloid progenitors (CMPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)CD41(hi)) establish an early branch point for separate lineage-commitment pathways from hematopoietic stem cells, with the notable exception that both pathways are proposed to generate all myeloid innate immune cell types through the same myeloid-restricted pre-granulocyte-macrophage progenitor (pre-GM) (Lin(-)Sca-1(-)c-Kit(+)CD41(-)FcγRII/III(-)CD150(-)CD105(-)). By single-cell transcriptome profiling of pre-GMs, we identified distinct myeloid differentiation pathways: a pathway expressing the gene encoding the transcription factor GATA-1 generated mast cells, eosinophils, megakaryocytes and erythroid cells, and a pathway lacking expression of that gene generated monocytes, neutrophils and lymphocytes. These results identify an early hematopoietic-lineage bifurcation that separates the myeloid lineages before their segregation from other hematopoietic-lineage potential.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Linfocitos/fisiología , Células Mieloides/fisiología , Células Progenitoras Mieloides/fisiología , Animales , Antígenos CD/metabolismo , Células Cultivadas , Biología Computacional , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Hematopoyesis , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Análisis de Matrices Tisulares , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
2.
Immunity ; 45(2): 346-57, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27533015

RESUMEN

Hematopoietic stem cells (HSCs) undergo a functional switch in neonatal mice hallmarked by a decrease in self-renewing divisions and entry into quiescence. Here, we investigated whether the developmental attenuation of B-1a cell output is a consequence of a shift in stem cell state during ontogeny. Using cellular barcoding for in vivo single-cell fate analyses, we found that fetal liver definitive HSCs gave rise to both B-1a and B-2 cells. Whereas B-1a potential diminished in all HSCs with time, B-2 output was maintained. B-1a and B-2 plasticity could be reinitiated in a subset of adult HSCs by ectopic expression of the RNA binding protein LIN28B, a key regulator of fetal hematopoiesis, and this coincided with the clonal reversal to fetal-like elevated self-renewal and repopulation potential. These results anchor the attenuation of B-1a cell output to fetal HSC behavior and demonstrate that the developmental decline in regenerative potential represents a reversible HSC state.


Asunto(s)
Linfocitos B/fisiología , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/fisiología , Hígado/fisiología , Subgrupos Linfocitarios/fisiología , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Plasticidad de la Célula , Autorrenovación de las Células , Células Clonales , Proteínas de Unión al ADN/genética , Femenino , Hematopoyesis/genética , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Unión al ARN , Análisis de la Célula Individual
3.
Immunity ; 43(2): 394-407, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26287684

RESUMEN

Natural killer (NK) cells are cytotoxic lymphocytes and play a vital role in controlling viral infections and cancer. In contrast to B and T lymphopoiesis where cellular and regulatory pathways have been extensively characterized, the cellular stages of early human NK cell commitment remain poorly understood. Here we demonstrate that a Lin(-)CD34(+)CD38(+)CD123(-)CD45RA(+)CD7(+)CD10(+)CD127(-) population represents a NK lineage-restricted progenitor (NKP) in fetal development, umbilical cord blood, and adult tissues. The newly identified NKP has robust NK cell potential both in vitro and in vivo, generates functionally cytotoxic NK cells, and lacks the ability to produce T cells, B cells, myeloid cells, and innate lymphoid-like cells (ILCs). Our findings identify an early step to human NK cell commitment and provide new insights into the human hematopoietic hierarchy.


Asunto(s)
Sangre Fetal/citología , Feto/citología , Hematopoyesis , Células Asesinas Naturales/fisiología , Células Progenitoras Linfoides/fisiología , Adulto , Antígenos CD/metabolismo , Diferenciación Celular , Linaje de la Célula , Citotoxicidad Inmunológica , Desarrollo Fetal , Hematopoyesis/inmunología , Humanos , Inmunidad Innata , Inmunofenotipificación
4.
J Immunol ; 205(7): 1830-1841, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32839237

RESUMEN

The ß-catenin/Wnt signaling pathway plays an important role in all stages of T cell development. Nemo-like kinase (NLK) is an evolutionary conserved serine/threonine kinase and a negative regulator of the Wnt signaling pathway. NLK can directly phosphorylate histone deacetylase 1 (HDAC1), as well as T cell factor/lymphoid enhancer-binding factor (TCF/LEF), causing subsequent repression of target gene transcription. By engineering mice lacking NLK in early stages of T cell development, we set out to characterize the role NLK plays in T cell development and found that deletion of NLK does not affect mouse health or lymphoid tissue development. Instead, these mice harbored a reduced number of single-positive (SP) CD8+ thymocytes without any defects in the SP CD4+ thymocyte population. The decrease in SP CD8+ thymocytes was not caused by a block in differentiation from double-positive CD4+CD8+ cells. Neither TCR signaling nor activation was altered in the absence of NLK. Instead, we observed a significant increase in cell death and reduced phosphorylation of LEF1 as well as HDAC1 among NLK-deleted SP CD8+ cells. Thus, NLK seems to play an important role in the survival of CD8+ thymocytes. Our data provide evidence for a new function for NLK with regard to its involvement in T cell development and supporting survival of SP CD8+ thymocytes.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Subgrupos de Linfocitos T/fisiología , Timocitos/fisiología , Animales , Diferenciación Celular , Supervivencia Celular , Histona Desacetilasa 1/metabolismo , Activación de Linfocitos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Proteínas Wnt/metabolismo
5.
J Immunol ; 201(11): 3307-3319, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366956

RESUMEN

Within the hematopoietic system, the Notch pathway is critical for promoting thymic T cell development and suppressing the B and myeloid lineage fates; however, its impact on NK lymphopoiesis is less understood. To study the role of Notch during NK cell development in vivo, we investigated different NK cell compartments and function in Rbp-Jkfl/flVav-Cretg/+ mice, in which Rbp-Jk, the major transcriptional effector of canonical Notch signaling, was specifically deleted in all hematopoietic cells. Peripheral conventional cytotoxic NK cells in Rbp-Jk-deleted mice were significantly reduced and had an activated phenotype. Furthermore, the pool of early NK cell progenitors in the bone marrow was decreased, whereas immature NK cells were increased, leading to a block in NK cell maturation. These changes were cell intrinsic as the hematopoietic chimeras generated after transplantation of Rbp-Jk-deficient bone marrow cells had the same NK cell phenotype as the Rbp-Jk-deleted donor mice, whereas the wild-type competitors did not. The expression of several crucial NK cell regulatory pathways was significantly altered after Rbp-Jk deletion. Together, these results demonstrate the involvement of canonical Notch signaling in regulation of multiple stages of NK cell development.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Células Asesinas Naturales/fisiología , Células Progenitoras Linfoides/fisiología , Linfopoyesis , Receptores Notch/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Quimera , Citotoxicidad Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
6.
Br J Haematol ; 183(4): 588-600, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30596405

RESUMEN

Given that FLT3 expression is highly restricted on lymphoid progenitors, it is possible that the established role of FLT3 in the regulation of B and T lymphopoiesis reflects its high expression and role in regulation of lymphoid-primed multipotent progenitors (LMPPs) or common lymphoid progenitors (CLPs). We generated a Flt3 conditional knock-out (Flt3fl/fl) mouse model to address the direct role of FLT3 in regulation of lymphoid-restricted progenitors, subsequent to turning on Rag1 expression, as well as potentially ontogeny-specific roles in B and T lymphopoiesis. Our studies establish a prominent and direct role of FLT3, independently of the established role of FLT3 in regulation of LMPPs and CLPs, in regulation of fetal as well as adult early B cell progenitors, and the early thymic progenitors (ETPs) in adult mice but not in the fetus. Our findings highlight the potential benefit of targeting poor prognosis acute B-cell progenitor leukaemia and ETP leukaemia with recurrent FLT3 mutations using clinical FLT3 inhibitors.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Progenitoras Linfoides/metabolismo , Linfopoyesis , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Células de la Médula Ósea/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Progenitoras Linfoides/patología , Ratones , Ratones Noqueados , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Timo/metabolismo , Timo/patología , Tirosina Quinasa 3 Similar a fms/genética
7.
Blood ; 128(2): 217-26, 2016 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-27207794

RESUMEN

Although it is well established that unique B-cell lineages develop through distinct regulatory mechanisms during embryonic development, much less is understood about the differences between embryonic and adult B-cell progenitor cells, likely to underpin the genetics and biology of infant and childhood PreB acute lymphoblastic leukemia (PreB-ALL), initiated by distinct leukemia-initiating translocations during embryonic development. Herein, we establish that a distinct subset of the earliest CD19(+) B-cell progenitors emerging in the E13.5 mouse fetal liver express the colony-stimulating factor-1 receptor (CSF1R), previously thought to be expressed, and play a lineage-restricted role in development of myeloid lineages, and macrophages in particular. These early embryonic CSF1R(+)CD19(+) ProB cells also express multiple other myeloid genes and, in line with this, possess residual myeloid as well as B-cell, but not T-cell lineage potential. Notably, these CSF1R(+) myeloid-primed ProB cells are uniquely present in a narrow window of embryonic fetal liver hematopoiesis and do not persist in adult bone marrow. Moreover, analysis of CSF1R-deficient mice establishes a distinct role of CSF1R in fetal B-lymphopoiesis. CSF1R(+) myeloid-primed embryonic ProB cells are relevant for infant and childhood PreB-ALLs, which frequently have a bi-phenotypic B-myeloid phenotype, and in which CSF1R-rearrangements have recently been reported.


Asunto(s)
Linaje de la Célula/fisiología , Feto/metabolismo , Linfopoyesis/fisiología , Células Precursoras de Linfocitos B/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Animales , Feto/citología , Ratones , Ratones Noqueados , Células Precursoras de Linfocitos B/citología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética
8.
Semin Immunol ; 26(2): 114-26, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24594002

RESUMEN

Bone marrow-derived natural killer (NK) cells constitute the major subset of cytotoxic lymphocytes in peripheral blood. They provide innate defense against intracellular infection or malignancy and contribute to immune homeostasis. Large numbers of NK cells are also present in tissues, including the liver and uterus, where they can mediate immunosurveillance but also play important roles in tissue remodeling and vascularization. Here, we review the pathways involved in NK cell lineage commitment and differentiation, discussing relationships to other lymphocyte populations and highlighting genetic determinants. Characterizing NK cells from distinct tissues and during infections have revealed subset specializations, reflecting inherent cellular plasticity. In this context, we discuss how different environmental and inflammatory stimuli may shape NK cells. Particular emphasis is placed on genes identified as being critical for NK cell development, differentiation, and function from studies of model organisms or associations with disease. Such studies are also revealing important cellular redundancies. Here, we provide a view of the genetic framework constraining NK cell development and function, pinpointing molecules required for these processes but also underscoring plasticity and redundancy that may underlie robust immunological function. With this view, built in redundancy may highlight the importance of NK cells to immunity.


Asunto(s)
Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Animales , Diferenciación Celular/inmunología , Humanos , Inmunidad/fisiología , Células Asesinas Naturales/citología
9.
Blood ; 120(1): 63-75, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22072559

RESUMEN

The earliest stages of natural killer (NK)-cell development are not well characterized. In this study, we investigated in different fetal hematopoietic tissues how NK-cell progenitors and their mature NK-cell progeny emerge and expand during fetal development. Here we demonstrate, for the first time, that the counterpart of adult BM Lin(-)CD122(+)NK1.1(-)DX5(-) NK-cell progenitor (NKP) emerges in the fetal liver at E13.5. After NKP expansion, immature NK cells emerge at E14.5 in the liver and E15.5 in the spleen. Thymic NK cells arise at E15.5, whereas functionally competent cytotoxic NK cells were present in the liver and spleen at E16.5 and E17.5, respectively. Fetal NKPs failed to produce B and myeloid cells but sustained combined NK- and T-lineage potential at the single-cell level. NKPs were also found in the fetal blood, spleen, and thymus. These findings show the emergence and expansion of bipotent NK/T-cell progenitor during fetal and adult lymphopoiesis, further supporting that NK/T-lineage restriction is taking place prethymically. Uncovering the earliest NK-cell developmental stages will provide important clues, helping to understand the origin of diverse NK-cell subsets, their progenitors, and key regulators.


Asunto(s)
Linaje de la Célula/inmunología , Sistema Inmunológico/citología , Sistema Inmunológico/embriología , Células Asesinas Naturales/citología , Células Madre/citología , Animales , Antígenos Ly/metabolismo , Linfocitos B/citología , Diferenciación Celular/inmunología , Células Cultivadas , Femenino , Subunidad beta del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Hígado/citología , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Embarazo , Bazo/citología , Bazo/embriología , Células del Estroma/citología , Linfocitos T/citología , Timo/citología , Timo/embriología
10.
Blood Adv ; 8(11): 2933-2951, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38484189

RESUMEN

ABSTRACT: Natural killer (NK) cells represent the cytotoxic member within the innate lymphoid cell (ILC) family that are important against viral infections and cancer. Although the NK cell emergence from hematopoietic stem and progenitor cells through multiple intermediate stages and the underlying regulatory gene network has been extensively studied in mice, this process is not well characterized in humans. Here, using a temporal in vitro model to reconstruct the developmental trajectory of NK lineage, we identified an ILC-restricted oligopotent stage 3a CD34-CD117+CD161+CD45RA+CD56- progenitor population, that exclusively gave rise to CD56-expressing ILCs in vitro. We also further investigated a previously nonappreciated heterogeneity within the CD56+CD94-NKp44+ subset, phenotypically equivalent to stage 3b population containing both group-1 ILC and RORγt+ ILC3 cells, that could be further separated based on their differential expression of DNAM-1 and CD161 receptors. We confirmed that DNAM-1hi S3b and CD161hiCD117hi ILC3 populations distinctively differed in their expression of effector molecules, cytokine secretion, and cytotoxic activity. Furthermore, analysis of lineage output using DNA-barcode tracing across these stages supported a close developmental relationship between S3b-NK and S4-NK (CD56+CD94+) cells, whereas distant to the ILC3 subset. Cross-referencing gene signatures of culture-derived NK cells and other noncytotoxic ILCs with publicly available data sets validated that these in vitro stages highly resemble transcriptional profiles of respective in vivo ILC counterparts. Finally, by integrating RNA velocity and gene network analysis through single-cell regulatory network inference and clustering we unravel a network of coordinated and highly dynamic regulons driving the cytotoxic NK cell program, as a guide map for future studies on NK cell regulation.


Asunto(s)
Células Asesinas Naturales , Análisis de la Célula Individual , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Análisis de la Célula Individual/métodos , Linaje de la Célula , Inmunidad Innata , Diferenciación Celular
11.
Blood ; 118(13): 3613-21, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21813452

RESUMEN

Acquisition of homozygous activating growth factor receptor mutations might accelerate cancer progression through a simple gene-dosage effect. Internal tandem duplications (ITDs) of FLT3 occur in approximately 25% cases of acute myeloid leukemia and induce ligand-independent constitutive signaling. Homozygous FLT3-ITDs confer an adverse prognosis and are frequently detected at relapse. Using a mouse knockin model of Flt3-internal tandem duplication (Flt3-ITD)-induced myeloproliferation, we herein demonstrate that the enhanced myeloid phenotype and expansion of granulocyte-monocyte and primitive Lin(-)Sca1(+)c-Kit(+) progenitors in Flt3-ITD homozygous mice can in part be mediated through the loss of the second wild-type allele. Further, whereas autocrine FLT3 ligand production has been implicated in FLT3-ITD myeloid malignancies and resistance to FLT3 inhibitors, we demonstrate here that the mouse Flt3(ITD/ITD) myeloid phenotype is FLT3 ligand-independent.


Asunto(s)
Dosificación de Gen/fisiología , Duplicación de Gen/fisiología , Pérdida de Heterocigocidad/fisiología , Proteínas de la Membrana/genética , Trastornos Mieloproliferativos/genética , Tirosina Quinasa 3 Similar a fms/fisiología , Alelos , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Células de la Médula Ósea/fisiología , Proliferación Celular , Células Cultivadas , Técnicas de Sustitución del Gen , Pérdida de Heterocigocidad/genética , Masculino , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Secuencias Repetidas en Tándem/genética , Secuencias Repetidas en Tándem/fisiología , Tirosina Quinasa 3 Similar a fms/metabolismo
12.
Blood ; 116(2): 183-92, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20421450

RESUMEN

Although bone marrow (BM) is the main site of natural killer (NK)-cell development in adult mice, recent studies have identified a distinct thymic-dependent NK pathway, implicating a possible close link between NK- and T-cell development in adult hematopoiesis. To investigate whether a potential NK-/T-lineage restriction of multipotent progenitors might take place already in the BM, we tested the full lineage potentials of NK-cell progenitors in adult BM. Notably, although Lin(-)CD122(+)NK1.1(-)DX5(-) NK-cell progenitors failed to commit to the B and myeloid lineages, they sustained a combined NK- and T-cell potential in vivo and in vitro at the single-cell level. Whereas T-cell development from NK/T progenitors is Notch-dependent, their contribution to thymic and BM NK cells remains Notch-independent. These findings demonstrate the existence of bipotent NK-/T-cell progenitors in adult BM.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/inmunología , Células Madre Hematopoyéticas/citología , Células Asesinas Naturales/citología , Linfocitos T/citología , Animales , Células de la Médula Ósea/inmunología , Linaje de la Célula , Separación Celular , Citometría de Flujo , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Células Madre Multipotentes/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Timo/citología , Timo/inmunología
13.
Blood ; 115(24): 5061-8, 2010 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-20393130

RESUMEN

Mice deficient in c-fms-like tyrosine kinase 3 (FLT3) signaling have reductions in early multipotent and lymphoid progenitors, whereas no evident myeloid phenotype has been reported. However, activating mutations of Flt3 are among the most common genetic events in acute myeloid leukemia and mice harboring internal tandem duplications within Flt3 (Flt3-ITD) develop myeloproliferative disease, with characteristic expansion of granulocyte-monocyte (GM) progenitors (GMP), possibly compatible with FLT3-ITD promoting a myeloid fate of multipotent progenitors. Alternatively, FLT3 might be expressed at the earliest stages of GM development. Herein, we investigated the expression, function, and role of FLT3 in recently identified early GMPs. Flt3-cre fate-mapping established that most progenitors and mature progeny of the GM lineage are derived from Flt3-expressing progenitors. A higher expression of FLT3 was found in preGMP compared with GMP, and preGMPs were more responsive to stimulation with FLT3 ligand (FL). Whereas preGMPs and GMPs were reduced in Fl(-/-) mice, megakaryocyte-erythroid progenitors were unaffected and lacked FLT3 expression. Notably, mice deficient in both thrombopoietin (THPO) and FL had a more pronounced GMP phenotype than Thpo(-/-) mice, establishing a role of FL in THPO-dependent and -independent regulation of GMPs, of likely significance for myeloid malignancies with Flt3-ITD mutations.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide Aguda/fisiopatología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Linaje de la Célula/fisiología , Expresión Génica/fisiología , Granulocitos/citología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Células Mieloides/citología , Transducción de Señal/fisiología , Trombopoyetina/genética
14.
Cell Rep ; 39(6): 110798, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545037

RESUMEN

The emerging notion of hematopoietic stem and progenitor cells (HSPCs) as a low-primed cloud without sharply demarcated gene expression programs raises the question on how cellular-fate options emerge and at which stem-like stage lineage priming is initiated. Here, we investigate single-cell chromatin accessibility of Lineage-, cKit+, and Sca1+ (LSK) HSPCs spanning the early differentiation landscape. Application of a signal-processing algorithm to detect transition points corresponding to massive alterations in accessibility of 571 transcription factor motifs reveals a population of LSK FMS-like tyrosine kinase 3 (Flt3)intCD9high cells that concurrently display stem-like and lineage-affiliated chromatin signatures, pointing to a simultaneous gain of both lympho-myeloid and megakaryocyte-erythroid programs. Molecularly and functionally, these cells position between stem cells and committed progenitors and display multi-lineage capacity in vitro and in vivo but lack self-renewal activity. This integrative molecular analysis resolves the heterogeneity of cells along hematopoietic differentiation and permits investigation of chromatin-mediated transition between multipotency and lineage restriction.


Asunto(s)
Cromatina , Células Madre Hematopoyéticas , Diferenciación Celular , Linaje de la Célula , Cromatina/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Megacariocitos
15.
Blood ; 113(15): 3453-60, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19188666

RESUMEN

Originally cloned from hematopoietic stem cell (HSC) populations and its ligand being extensively used to promote ex vivo HSC expansion, the FMS-like tyrosine kinase 3 (FLT3; also called FLK2) receptor and its ligand (FL) were expected to emerge as an important physiologic regulator of HSC maintenance and expansion. However, the role of FLT3 receptor and ligand in HSC regulation remains unclear and disputed. Herein, using Fl-deficient mice, we establish for the first time that HSC expansion in fetal liver and after transplantation is FL independent. Because previous findings in Flk2(-/-) mice were compatible with an important role of FLT3 receptor in HSC regulation and because alternative ligands might potentially interact directly or indirectly with FLT3 receptor, we here also characterized HSCs in Flk2(-/-) mice. Advanced phenotypic as well as functional evaluation of Flk2(-/-) HSCs showed that the FLT3 receptor is dispensable for HSC steady-state maintenance and expansion after transplantation. Taken together, these studies show that the FLT3 receptor and ligand are not critical regulators of mouse HSCs, neither in steady state nor during fetal or posttransplantation expansion.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Proteínas de la Membrana/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , División Celular/fisiología , Citometría de Flujo , Supervivencia de Injerto/fisiología , Hematopoyesis/fisiología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fenotipo , Tirosina Quinasa 3 Similar a fms/genética
16.
J Immunol ; 182(3): 1460-8, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19155493

RESUMEN

Although bone marrow (BM) represents the main site for postnatal NK cell development, recently a distinct thymic-dependent NK cell pathway was identified. These studies were designed to investigate the role of cytokines in regulation of thymic NK cells and to compare with established regulatory pathways of BM-dependent NK cell compartment. The common cytokine receptor gamma-chain (Il2rg) essential for IL-15-induced signaling, and FMS-like tyrosine kinase 3 (FLT3) receptor ligand (Flt3l) were previously identified as important regulatory pathways of the BM NK cell compartment based on lack of function studies in mice, however their complementary action remains unknown. By investigating mice double-deficient in Il2rg and Flt3l (Flt3l(-/-) Il2rg(-/-)), we demonstrate that FLT3L is important for IL2Rg-independent maintenance of both immature BM as well as peripheral NK cells. In contrast to IL-7, which is dispensable for BM but important for thymic NK cells, IL-15 has a direct and important role in both thymic and BM NK cell compartments. Although thymic NK cells were not affected in Flt3l(-/-) mice, Flt3l(-/-)Il2rg(-/-) mice lacked detectable thymic NK cells, suggesting that FLT3L is also important for IL-2Rg-independent maintenance of thymic NK cells. Thus, IL-2Rg cytokines and FLT3L play complementary roles and are indispensable for homeostasis of both BM and thymic dependent NK cell development, suggesting that the cytokine pathways crucial for these two distinct NK cell pathways are largely overlapping.


Asunto(s)
Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Subunidad gamma Común de Receptores de Interleucina/fisiología , Células Asesinas Naturales/inmunología , Proteínas de la Membrana/fisiología , Transducción de Señal/inmunología , Timo/citología , Timo/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/genética , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-15/deficiencia , Interleucina-15/genética , Interleucina-15/fisiología , Interleucina-7/deficiencia , Interleucina-7/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Timo/metabolismo , Tirosina Quinasa 3 Similar a fms/deficiencia , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/fisiología
17.
Cell Rep ; 34(12): 108894, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33761361

RESUMEN

The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.


Asunto(s)
Linaje de la Célula , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteómica , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Diferenciación Celular , Feto/citología , Granulocitos/citología , Células HEK293 , Humanos , Inmunofenotipificación , Factores Reguladores del Interferón/metabolismo , Cinética , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/metabolismo , Ratones Endogámicos C57BL , Monocitos/citología , Monocitos/metabolismo , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Proteoma/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
18.
J Exp Med ; 198(10): 1495-506, 2003 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-14610045

RESUMEN

Extensive studies of mice deficient in one or several cytokine receptors have failed to support an indispensable role of cytokines in development of multiple blood cell lineages. Whereas B1 B cells and Igs are sustained at normal levels throughout life of mice deficient in IL-7, IL-7Ralpha, common cytokine receptor gamma chain, or flt3 ligand (FL), we report here that adult mice double deficient in IL-7Ralpha and FL completely lack visible LNs, conventional IgM+ B cells, IgA+ plasma cells, and B1 cells, and consequently produce no Igs. All stages of committed B cell progenitors are undetectable in FL-/- x IL-7Ralpha-/- BM that also lacks expression of the B cell commitment factor Pax5 and its direct target genes. Furthermore, in contrast to IL-7Ralpha-/- mice, FL-/- x IL-7Ralpha-/- mice also lack mature B cells and detectable committed B cell progenitors during fetal development. Thus, signaling through the cytokine tyrosine kinase receptor flt3 and IL-7Ralpha are indispensable for fetal and adult B cell development.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo , Receptores de Interleucina-7/metabolismo , Animales , Linfocitos B/citología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Ganglios Linfáticos Agregados/metabolismo , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
19.
Blood ; 112(6): 2297-304, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18566323

RESUMEN

Phenotypically and functionally distinct progenitors and developmental pathways have been proposed to exist for fetally derived B-1 and conventional B-2 cells. Although IL-7 appears to be the primary cytokine regulator of fetal and adult B lymphopoiesis in mice, considerable fetal B lymphopoiesis and postnatal B cells are sustained in the absence of IL-7; in humans, B-cell generation is suggested to be largely IL-7-independent, as severe combined immune-deficient patients with IL-7 deficiency appear to have normal B-cell numbers. However, the role of other cytokines in IL-7-independent B lymphopoiesis remains to be established. Although thymic stromal lymphopoietin (TSLP) has been proposed to be the main factor driving IL-7-independent B lymphopoiesis and to distinguish fetal from adult B-cell progenitor development in mice, recent studies failed to support a primary role of TSLP in IL-7-independent fetal B-cell development. However, the role of TSLP in IL-7-independent adult B lymphopoiesis and in particular in regulation of B-1 cells remains to be established. Here we demonstrate that, rather than TSLP, IL-7 and FLT3 ligand are combined responsible for all B-cell generation in mice, including recently identified B-1-specified cell progenitors. Thus, the same IL-7- and FLT3 ligand-mediated signal-ing regulates alternative pathways of fetal and adult B-1 and B-2 lymphopoiesis.


Asunto(s)
Linfocitos B/citología , Citocinas/fisiología , Interleucina-7/fisiología , Linfopoyesis , Proteínas de la Membrana/fisiología , Animales , Linaje de la Célula , Feto/citología , Ratones , Linfopoyetina del Estroma Tímico
20.
Crit Rev Immunol ; 29(6): 487-530, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20121697

RESUMEN

The thymus produces new T cells throughout life but has no self-renewing ability and requires replenishment and recruitment of progenitors derived from the bone marrow. Despite the progress in delineation of mature blood cell development several questions remain regarding T lymphopoiesis. Understanding the developmental stages from multipotent hematopoietic stem cells (HSCs) to the T-cell lineage-restricted progenitors has many potential clinical implications as it is important for understanding malignant transformation in T-cell cancer, accelerating T-cell regeneration after bone marrow transplantation and chemotherapy, and establishing new therapies to treat T-cell immune deficiencies. This review focuses on the steps leading from the HSCs in the bone marrow to the lineage committed T cells inside the thymus.


Asunto(s)
Células de la Médula Ósea , Linfocitos T/citología , Linfocitos T/inmunología , Timo/citología , Animales , Médula Ósea/inmunología , Diferenciación Celular , Humanos , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA