Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 201(11): 3307-3319, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30366956

RESUMO

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.


Assuntos
Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Células Matadoras Naturais/fisiologia , Células Progenitoras Linfoides/fisiologia , Linfopoese , Receptores Notch/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Quimera , Citotoxicidade Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
2.
Dev Cell ; 44(3): 362-377.e7, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29290585

RESUMO

ETV6-RUNX1 is associated with childhood acute B-lymphoblastic leukemia (cALL) functioning as a first-hit mutation that initiates a clinically silent pre-leukemia in utero. Because lineage commitment hierarchies differ between embryo and adult, and the impact of oncogenes is cell-context dependent, we hypothesized that the childhood affiliation of ETV6-RUNX1 cALL reflects its origins in a progenitor unique to embryonic life. We characterize the first emerging B cells in first-trimester human embryos, identifying a developmentally restricted CD19-IL-7R+ progenitor compartment, which transitions from a myeloid to lymphoid program during ontogeny. This developmental series is recapitulated in differentiating human pluripotent stem cells (hPSCs), thereby providing a model for the initiation of cALL. Genome-engineered hPSCs expressing ETV6-RUNX1 from the endogenous ETV6 locus show expansion of the CD19-IL-7R+ compartment, show a partial block in B lineage commitment, and produce proB cells with aberrant myeloid gene expression signatures and potential: features (collectively) consistent with a pre-leukemic state.


Assuntos
Linfócitos B/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Desenvolvimento Embrionário , Regulação Leucêmica da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/patologia , Células Mieloides/patologia , Proteínas de Fusão Oncogênica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Doença Aguda , Linfócitos B/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Células Mieloides/metabolismo , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Receptores de Interleucina-7 , Transcriptoma
3.
Br J Haematol ; 183(4): 588-600, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30596405

RESUMO

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.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular , Células Progenitoras Linfoides/metabolismo , Linfopoese , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Animais , Células da Medula Óssea/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Células Progenitoras Linfoides/patologia , Camundongos , Camundongos Knockout , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Timo/metabolismo , Timo/patologia , Tirosina Quinase 3 Semelhante a fms/genética
4.
Immunity ; 45(2): 346-57, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533015

RESUMO

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.


Assuntos
Linfócitos B/fisiologia , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Fígado/fisiologia , Subpopulações de Linfócitos/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Plasticidade Celular , Autorrenovação Celular , Células Clonais , Proteínas de Ligação a DNA/genética , Feminino , Hematopoese/genética , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Ligação a RNA , Análise de Célula Única
5.
Blood ; 128(2): 217-26, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27207794

RESUMO

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.


Assuntos
Linhagem da Célula/fisiologia , Feto/metabolismo , Linfopoese/fisiologia , Células Precursoras de Linfócitos B/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Animais , Feto/citologia , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/citologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética
6.
Nat Immunol ; 17(6): 666-676, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27043410

RESUMO

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.


Assuntos
Diferenciação Celular , Linhagem da Célula , Linfócitos/fisiologia , Células Mieloides/fisiologia , Células Progenitoras Mieloides/fisiologia , Animais , Antígenos CD/metabolismo , Células Cultivadas , Biologia Computacional , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Hematopoese , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Análise de Sequência de RNA , Análise de Célula Única , Análise Serial de Tecidos , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
7.
Immunity ; 43(2): 394-407, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26287684

RESUMO

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.


Assuntos
Sangue Fetal/citologia , Feto/citologia , Hematopoese , Células Matadoras Naturais/fisiologia , Células Progenitoras Linfoides/fisiologia , Adulto , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem da Célula , Citotoxicidade Imunológica , Desenvolvimento Fetal , Hematopoese/imunologia , Humanos , Imunidade Inata , Imunofenotipagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...