Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 49(4): 640-653.e5, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30332630

RESUMO

Tissue-resident mast cells are associated with many inflammatory and physiological processes. Although mast cells arise from the yolk sac, the exact ontogeny of adult mast cells remains unclear. Here we have investigated the hematopoietic origin of mast cells using fate-mapping systems. We have shown that early erythro-myeloid progenitors (EMPs), late EMPs, and definitive hematopoietic stem cells (HSCs) each gave rise to mast cells in succession via an intermediate integrin ß7+ progenitor. From late embryogenesis to adult, early EMP-derived mast cells were largely replaced by late EMP-derived cells in most connective tissues except adipose and pleural cavity. Thus, mast cells with distinct origin displayed tissue-location preferences: early EMP-derived cells were limited to adipose and pleural cavity and late EMP-derived cells dominated most connective tissues, while HSC-derived cells were a main group in mucosa. Therefore, embryonic origin shapes the heterogeneity of adult mast cells, with diverse functions in immunity and development.


Assuntos
Células Eritroides/imunologia , Mastócitos/imunologia , Células Progenitoras Mieloides/imunologia , Animais , Linhagem da Célula/imunologia , Células Cultivadas , Tecido Conjuntivo/imunologia , Tecido Conjuntivo/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/imunologia , Células Eritroides/citologia , Células Eritroides/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Cadeias beta de Integrinas/imunologia , Cadeias beta de Integrinas/metabolismo , Mastócitos/citologia , Mastócitos/metabolismo , Camundongos Transgênicos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/metabolismo
2.
Immunity ; 49(1): 93-106.e7, 2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-29958804

RESUMO

There is a growing body of research on the neural control of immunity and inflammation. However, it is not known whether the nervous system can regulate the production of inflammatory myeloid cells from hematopoietic progenitor cells in disease conditions. Myeloid cell numbers in diabetic patients were strongly correlated with plasma concentrations of norepinephrine, suggesting the role of sympathetic neuronal activation in myeloid cell production. The spleens of diabetic patients and mice contained higher numbers of tyrosine hydroxylase (TH)-expressing leukocytes that produced catecholamines. Granulocyte macrophage progenitors (GMPs) expressed the ß2 adrenergic receptor, a target of catecholamines. Ablation of splenic sympathetic neuronal signaling using surgical, chemical, and genetic approaches diminished GMP proliferation and myeloid cell development. Finally, mice lacking TH-producing leukocytes had reduced GMP proliferation, resulting in diminished myelopoiesis. Taken together, our study demonstrates that catecholamines produced by leukocytes and sympathetic nerve termini promote GMP proliferation and myeloid cell development.


Assuntos
Diabetes Mellitus/fisiopatologia , Células Progenitoras de Granulócitos e Macrófagos/citologia , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Mielopoese , Neuroimunomodulação , Sistema Nervoso Simpático/metabolismo , Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus/sangue , Modelos Animais de Doenças , Feminino , Humanos , Leucócitos/enzimologia , Leucócitos/metabolismo , Masculino , Camundongos , Células Mieloides/citologia , Mielopoese/efeitos dos fármacos , Neuroimunomodulação/efeitos dos fármacos , Norepinefrina/sangue , Transdução de Sinais/efeitos dos fármacos , Baço/citologia , Baço/inervação , Baço/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos
3.
Immunol Rev ; 315(1): 31-53, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36752151

RESUMO

Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.


Assuntos
Células-Tronco Hematopoéticas , Mastócitos , Animais , Humanos , Macrófagos , Medula Óssea , Hematopoese/genética , Desenvolvimento Fetal , Diferenciação Celular
4.
Development ; 149(8)2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35438172

RESUMO

Hofbauer cells (HBCs) are tissue macrophages of the placenta thought to be important for fetoplacental vascular development and innate immune protection. The developmental origins of HBCs remain unresolved and could implicate functional diversity of HBCs in placenta development and disease. In this study, we used flow cytometry and paternally inherited reporters to phenotype placenta macrophages and to identify fetal-derived HBCs and placenta-associated maternal macrophages in the mouse. In vivo pulse-labeling traced the ontogeny of HBCs from yolk sac-derived erythro-myeloid progenitors, with a minor contribution from fetal hematopoietic stem cells later on. Single-cell RNA-sequencing revealed transcriptional similarities between placenta macrophages and erythro-myeloid progenitor-derived fetal liver macrophages and microglia. As with other fetal tissue macrophages, HBCs were dependent on the transcription factor Pu.1, the loss-of-function of which in embryos disrupted fetoplacental labyrinth morphology, supporting a role for HBC in labyrinth angiogenesis and/or remodeling. HBC were also sensitive to Pu.1 (Spi1) haploinsufficiency, which caused an initial deficiency in the numbers of macrophages in the early mouse placenta. These results provide groundwork for future investigation into the relationship between HBC ontogeny and function in placenta pathophysiology.


Assuntos
Macrófagos , Placenta , Animais , Feminino , Células-Tronco Hematopoéticas , Camundongos , Células Progenitoras Mieloides , Gravidez , Saco Vitelino
5.
Development ; 149(17)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35929537

RESUMO

Mutations that occur in RNA-splicing machinery may contribute to hematopoiesis-related diseases. How splicing factor mutations perturb hematopoiesis, especially in the differentiation of erythro-myeloid progenitors (EMPs), remains elusive. Dhx38 is a pre-mRNA splicing-related DEAH box RNA helicase, for which the physiological functions and splicing mechanisms during hematopoiesis currently remain unclear. Here, we report that Dhx38 exerts a broad effect on definitive EMPs as well as the differentiation and maintenance of hematopoietic stem and progenitor cells (HSPCs). In dhx38 knockout zebrafish, EMPs and HSPCs were found to be arrested in mitotic prometaphase, accompanied by a 'grape' karyotype, owing to the defects in chromosome alignment. Abnormal alternatively spliced genes related to chromosome segregation, the microtubule cytoskeleton, cell cycle kinases and DNA damage were present in the dhx38 mutants. Subsequently, EMPs and HSPCs in dhx38 mutants underwent P53-dependent apoptosis. This study provides novel insights into alternative splicing regulated by Dhx38, a process that plays a crucial role in the proliferation and differentiation of fetal EMPs and HSPCs.


Assuntos
Processamento Alternativo , Peixe-Zebra , Processamento Alternativo/genética , Animais , Hematopoese/genética , Células-Tronco Hematopoéticas , Células Progenitoras Mieloides , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
6.
EMBO Rep ; 24(2): e55503, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36440617

RESUMO

Myeloid cell development in bone marrow is essential for the maintenance of peripheral immune homeostasis. However, the role of intracellular protein trafficking pathways during myeloid cell differentiation is currently unknown. By mining bioinformatics data, we identify trafficking protein particle complex subunit 1 (TRAPPC1) as continuously upregulated during myeloid cell development. Using inducible ER-TRAPPC1 knockout mice and bone marrow chimeric mouse models, we demonstrate that TRAPPC1 deficiency causes severe monocyte and neutrophil defects, accompanied by a selective decrease in common myeloid progenitors (CMPs) and subsequent cell subsets in bone marrow. TRAPPC1-deleted CMPs differentiate poorly into monocytes and neutrophils in vivo and in vitro, in addition to exhibiting enhanced endoplasmic reticulum stress and apoptosis via a Ca2+ -mitochondria-dependent pathway. Cell cycle arrest and senescence of TRAPPC1-deleted CMPs are mediated by the activation of pancreatic endoplasmic reticulum kinase and the upregulation of cyclin-dependent kinase inhibitor p21. This study reveals the essential role of TRAPPC1 in the maintenance and differentiation of CMPs and highlights the significance of protein processing and trafficking processes in myeloid cell development.


Assuntos
Medula Óssea , Células Progenitoras Mieloides , Proteínas de Transporte Vesicular , Animais , Camundongos , Medula Óssea/metabolismo , Diferenciação Celular , Camundongos Knockout , Monócitos , Células Progenitoras Mieloides/metabolismo , Neutrófilos , Proteínas de Transporte Vesicular/metabolismo
7.
Br J Haematol ; 205(1): 20-21, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763166

RESUMO

Histiocytic diseases arise from MAPK mutations in myeloid progenitors. Depending on whether the progenitor follows a dendritic cell or macrophage/monocyte lineage the final histology results in Langerhans cell histiocytosis, Rosai-Dorfman disease or Erdheim-Chester disease. Commentary on: Friedman et al. Mixed histiocytic neoplasms: A multicentre series revealing diverse somatic mutations and responses to targeted therapy. Br J Haematol 2024;205:127-137.


Assuntos
Mutação , Humanos , Doença de Erdheim-Chester/genética , Doença de Erdheim-Chester/patologia , Doença de Erdheim-Chester/diagnóstico , Histiocitose de Células de Langerhans/genética , Histiocitose de Células de Langerhans/patologia , Histiocitose de Células de Langerhans/diagnóstico , Histiocitose Sinusal/patologia , Histiocitose Sinusal/diagnóstico , Histiocitose Sinusal/genética
8.
Cytometry A ; 105(8): 580-594, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38995093

RESUMO

Senescence is an irreversible arrest of the cell cycle that can be characterized by markers of senescence such as p16, p21, and KI-67. The characterization of different senescence-associated phenotypes requires selection of the most relevant senescence markers to define reliable cytometric methodologies. Mass cytometry (a.k.a. Cytometry by time of flight, CyTOF) can monitor up to 40 different cell markers at the single-cell level and has the potential to integrate multiple senescence and other phenotypic markers to identify senescent cells within a complex tissue such as skeletal muscle, with greater accuracy and scalability than traditional bulk measurements and flow cytometry-based measurements. This article introduces an analysis framework for detecting putative senescent cells based on clustering, outlier detection, and Boolean logic for outliers. Results show that the pipeline can identify putative senescent cells in skeletal muscle with well-established markers such as p21 and potential markers such as GAPDH. It was also found that heterogeneity of putative senescent cells in skeletal muscle can partly be explained by their cell type. Additionally, autophagy-related proteins ATG4A, LRRK2, and GLB1 were identified as important proteins in predicting the putative senescent population, providing insights into the association between autophagy and senescence. It was observed that sex did not affect the proportion of putative senescent cells among total cells. However, age did have an effect, with a higher proportion observed in fibro/adipogenic progenitors (FAPs), satellite cells, M1 and M2 macrophages from old mice. Moreover, putative senescent cells from muscle of old and young mice show different expression levels of senescence-related proteins, with putative senescent cells of old mice having higher levels of p21 and GAPDH, whereas putative senescent cells of young mice had higher levels of IL-6. Overall, the analysis framework prioritizes multiple senescence-associated proteins to characterize putative senescent cells sourced from tissue made of different cell types.


Assuntos
Biomarcadores , Senescência Celular , Citometria de Fluxo , Músculo Esquelético , Animais , Senescência Celular/fisiologia , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Citometria de Fluxo/métodos , Biomarcadores/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Análise de Célula Única/métodos
9.
Fetal Pediatr Pathol ; : 1-15, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39108072

RESUMO

OBJECTIVE: Wide inter-individual variations in ionizing radiation (IR) responses of neonatal hematopoietic system calls for identifying reliable biomarkers to effectively estimate radiation exposure damages in neonates. METHODS: Association between telomere length (TL) at birth and radiation sensitivity of cord blood hematopoietic stem cells (HSC) from 166 healthy newborns were investigated by assessing their clonogenic differentiation. TL was determined as terminal restriction fragment (TRF) by Southern blot method. RESULTS: TL correlated with surviving fractions of total progenitor colony forming cell (CFC) content at 0.75 Gy (p < 0.05), granulo-macrophagic lineage colony forming units (CFU-GM) at 0.75 Gy (p < 0.05) and erythroid burst forming unit (BFU-E) at 0.75 Gy (p < 0.05) & at 3 Gy (p < 0.05) of newborns. CONCLUSION: Our results indicate risks for HSC clonogenic survival in neonates with shorter telomeres after IR exposure. These observations might aid in considering TL at birth as an assessment factor for radiation related hematopoietic challenges in children.

10.
Blood Cells Mol Dis ; 102: 102760, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37267696

RESUMO

Hematopoiesis and lineage commitment are regulated by several conserved cell-intrinsic signaling pathways, including MAPKs and ß-catenin/TCF/LEF. The Inhibitor of MyoD Family A (I-MFA), a transcriptional repressor and tumor suppressor gene, interacts with these pathways and is dysregulated in chronic and acute myeloid leukemias, suggesting it may play a role in development and differentiation during hematopoiesis. To study this, immune cell populations in the bone marrow (BM) and periphery were analyzed in mice lacking Mdfi, encoding I-MFA (I-MFA-/-), and wild type (WT) controls. I-MFA-/- mice had reduced spleen and BM cellularity, with significant hyposplenism, compared to WT mice. In blood, total red blood cells and platelet counts were significantly reduced in I-MFA-/- mice, accompanied by a reduction in megakaryocyte (MK)/erythrocyte progenitor cells and an increase in myeloid progenitors in BM compared to WT mice. The K562 cell line exhibits PMA-induced MK differentiation, and shRNA knockdown of I-MFA resulted in reduced differentiation compared to control, with an increase and prolongation in phospho-JNK and phospho-ERK signaling. Overexpression of I-MFA promoted MK differentiation. These results suggest I-MFA plays a cell-intrinsic role in the response to differentiation signals, an effect that can be explored in the context of hematological cancers or other blood proliferative disorders.


Assuntos
Medula Óssea , Megacariócitos , Camundongos , Animais , Medula Óssea/metabolismo , Diferenciação Celular , Hematopoese , Células da Medula Óssea/patologia , Linhagem da Célula
11.
Rinsho Ketsueki ; 64(9): 853-860, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37793858

RESUMO

Myelopoiesis is a process that produces myeloid cells including granulocytes and mononuclear phagocytes. The differentiation and proliferation of hematopoietic stem and progenitor cells are tightly regulated to meet demands for such myeloid cells both at steady state and under stressed conditions. CCAAT/enhancer-binding protein family transcription factors are involved not only in the appropriate regulation of myelopoiesis but also in dysregulated myelopoiesis. A recent study has revealed that inflammation, in addition to the established concepts or mechanisms of dysregulated myelopoiesis, triggers long-term epigenetic memory in hematopoietic stem/progenitor cells. Further, clonal hematopoiesis develops and impairs host health conditions via inflammatory conditions. Intensive studies covering both the basic and clinical aspects of myelopoiesis are required to establish therapeutic and even prophylactic approaches to different types of human diseases including hematopoietic and nonhematopoietic origins.


Assuntos
Células-Tronco Hematopoéticas , Mielopoese , Humanos , Mielopoese/fisiologia , Diferenciação Celular , Fatores de Transcrição , Células Mieloides
12.
J Transl Med ; 17(1): 247, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31366356

RESUMO

BACKGROUND: The molecular pathways that drive bone marrow myeloid progenitors (BMMP) development are very well understood and include a tight controlled multi-stage gene hierarch. Monocytes are versatile cells that display remarkable plasticity and may give rise to specific subsets of macrophages to proper promote tissue homesostasis upon an injury. However, the epigenetic mechanisms that underlie monocyte differentiation into the pro-inflammatory Ly6Chigh or the repairing Ly6Clow subsets are yet to be elucidated. We have previously shown that Epigenetic mechanisms Histone Deacetylase (HDAC) dependent are crucial for monocyte behavior and plasticity and in this work, we propose that this same mechanism underlies BMMP plasticity upon an inflammatory challenge in vivo. METHODS: BMMP were culture in the presence of GM-CSF alone or in combination with HDAC inhibitor (iHDAC) and phenotyped by flow cytometry, immune staining or western blot. iHDAC was topically added to skin wounds for 7 consecutive days and wound healing was monitored by flow cytometry and histopathological analysis. RESULTS: When BMMP were cultured in the presence of iHDAC, we showed that the CD11blow/Ly6Clow subset was the specific target of iHDAC that underwent chromatin hyperacetylation in vitro. Upon 13 days in the presence of iHDAC, BMMP gave rise to very elongated macrophages, that in turn, displayed a remarkable plasticity in a HDAC activity dependent fashion. HDAC-dependent cell shape was tight related to macrophage behavior and phenotype through the control of iNOS protein levels, showing that chromatin remodeling is a key component of macrophage plasticity and function. We then hypothesized that iHDAC would modulate the inflammatory response and favor tissue repair in vivo. To test this hypothesis, we topically added iHDAC to skin wounds during 7 consecutive days and followed tissue repair dynamics. In fact, iHDAC treated skin wounds presented an increase in wound closure at day 5 that was correlated to an enrichment in the CD11blow/Ly6Clow subset and in very elongated F4/80 positives macrophages in vivo, fully recapitulating the behavior previously observed in vitro. CONCLUSION: Our work provides the biological basis that connects chromatin remodeling to phenotypic plasticity, which in turn, may become a tractable therapeutic strategy in further translational studies.


Assuntos
Epigenoma , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Células Progenitoras Mieloides/citologia , Pele/efeitos dos fármacos , Pele/patologia , Cicatrização , Animais , Cromatina/química , Epigênese Genética , Histona Desacetilases/genética , Humanos , Inflamação , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Fenótipo
13.
Cell Immunol ; 330: 5-15, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29475558

RESUMO

Tissue-resident macrophages have pivotal functions for tissue defense and homeostasis. Two main discoveries have changed our current understanding of macrophage development: Their embryonic origin and their ability to self-renew throughout the lifespan. It is now well accepted that most tissue-resident macrophages are long-lived cells derived from a transient hematopoietic wave of erythro-myeloid progenitors (EMPs) emerging in the yolk sac. At least two distinct pathways derived from EMPs have been implicated in macrophage development. The first one, c-Myb-independent is giving rise to yolk sac macrophages also called primitive macrophages, and bypassing the classical monocytic intermediates. The second requires c-Myb expression and start once EMPs seed the fetal liver where they generate fetal monocytes. Sequentially, primitive macrophages seed every tissue and will ultimately give rise to microglia in the brain, rapidly isolated by the blood brain barrier, while EMP-derived fetal monocytes infiltrate every other tissues and gradually generate the major pool of adult tissue-resident macrophages by diluting the initial primitive macrophage contribution. A third wave of hematopoietic stem cells (HSC)-derived monocytes is also emerging from the fetal liver to contribute to the long-lived macrophage pool established at birth while the adult hematopoiesis is only starting in the bone marrow. We propose here to review recent insights about the different embryonic hematopoietic programs responsible for the generation of long-lived tissue-resident macrophages and their maintenance after birth.


Assuntos
Células-Tronco Hematopoéticas/citologia , Macrófagos/citologia , Monócitos/citologia , Saco Vitelino/citologia , Animais , Linhagem da Célula , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/citologia , Fígado/embriologia , Microglia/citologia , Proteínas Proto-Oncogênicas c-myb/metabolismo , Saco Vitelino/embriologia
14.
Stem Cells ; 33(3): 976-87, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25446279

RESUMO

Previous studies have predicted that reciprocal activation of GATA-1 and PU.1 regulates myelo-erythroid versus myelo-lymphoid lineage commitment in early hematopoiesis. Such PU.1-activating myelo-lymphoid progenitors exist within the lymphoid-primed multipotent progenitor (LMPP) population at the primitive Lineage(-) Sca-1(+) c-Kit(+) (LSK) stage. We here show that the counterpart of GATA-1-activating myelo-erythroid progenitor resides also at the LSK stage, expressing CD41 at a high level. Purified CD41(hi) LSK cells showed exceedingly strong and prolonged myelo-erythroid-restricted reconstitution, and primed myelo-erythroid gene expression with a more primitive molecular signature as compared to the original common myeloid progenitor (CMP). The CD41(hi) LSK cells more strongly contributed to emergent and malignant myelopoiesis than LMPPs, and produced the original CMP by downregulating Sca-1 and CD41, suggesting that they are the earliest CMPs. Thus, the hematopoietic developmental map should be revised by integrating the primary branchpoint comprised of the new, isolatable CD41(hi) CMP and the LMPP populations.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Leucemia Mieloide/patologia , Células Progenitoras Mieloides/citologia , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Células Cultivadas , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Progenitoras Mieloides/metabolismo
15.
Dev Growth Differ ; 56(6): 469-79, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25093513

RESUMO

Sry related high mobility group box 17 (Sox17), which is a marker of endodermal cells and a transcriptional regulator, has a critical role in the maintenance of fetal and neonatal hematopoietic stem cells (HSC). Sox17 has been identified as a key regulator of the development and differentiation of fetal hematopoietic progenitors from the aorta-gonad-mesonephros (AGM) region. The co-culture of Sox17-transduced hematopoietic progenitor cells (CD45(low) c-Kit(high) cells) from AGM regions on OP9 stromal cells gives rise to multipotential hematopoietic stem/progenitor cells. Here, we show that in a primary transplantation experiment, Sox17-transduction in CD45(low) c-Kit(high) cells of embryonic day (E) 10.5 AGM increased the absolute number of common myeloid progenitors (CMPs) in the bone marrow (BM) of recipient mice in comparison to that of granulocyte/macrophage progenitors (GMPs) and the megakaryocyte/erythroid progenitors (MEPs). When Sox17-transduced cells were cultured with OP9 stromal cells, Sox17-transduced GMPs (Sox17-GMPs), Sox17-transduced CMPs (Sox17-CMPs), and Sox17-transduced MEPs (Sox17-MEPs) were generated. Sox17-GMPs and Sox17-CMPs maintained their self-renewal capacity and the hematopoietic ability upon co-culture with the OP9 stromal cells for some passages. Moreover, Sox17-GMPs exhibited the increase in expression of c-Mpl and GATA-2 in comparison to GMPs of BM and Sox17-CMPs showed the increase in expression of c-Mpl, NF-E2, and ß-globin genes in comparison to CMPs of BM. Furthermore, when Sox17-transduced cells were cultured in methylcellulose to examine the colony-forming ability, Sox17-GMPs and Sox17-CMPs maintained the formation of mixed colonies for some passages. Taken together, Sox17 is suggested to regulate the maintenance and differentiation of hematopoietic progenitors derived from AGM regions at midgestation, in particular myeloid progenitors.


Assuntos
Proteínas HMGB/metabolismo , Células Mieloides/citologia , Fatores de Transcrição SOXF/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Técnicas de Cocultura , Proteínas HMGB/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células Progenitoras de Megacariócitos e Eritrócitos/citologia , Células Progenitoras de Megacariócitos e Eritrócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXF/genética
16.
Exp Cell Res ; 319(17): 2739-46, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23850973

RESUMO

The balance between quiescence and proliferation of HSCs is an important regulator of hematopoiesis. Loss of quiescence frequently results in HSCs exhaustion, which underscores the importance of tight regulation of proliferation in these cells. Studies have indicated that cyclin-dependent kinases are involved in the regulation of quiescence in HSCs. BRCA1 plays an important role in the repair of DNA double-stranded breaks, cell cycle, apoptosis and transcription. BRCA1 is expressed in the bone marrow. However, the function of BRCA1 in HSCs is unknown. In our study, we generated BRCA1 transgenic mice to investigate the effects of BRCA1 on the mechanisms of quiescence and differentiation in HSCs. The results demonstrate that over-expression of BRCA1 in the bone marrow impairs the development of B lymphocytes. Furthermore, BRCA1 induced an increase in the number of LSKs, LT-HSCs, ST-HSCs and MPPs. A competitive transplantation assay found that BRCA1 transgenic mice failed to reconstitute hematopoiesis. Moreover, BRCA1 regulates the expression of p21(waf1)/cip1 and p57(kip2), which results in a loss of quiescence in LSKs. Together, over-expression of BRCA1 in bone marrow disrupted the quiescent of LSKs, induced excessive accumulation of LSKs, and disrupted differentiation of the HSCs, which acts through the down-regulated of p21(waf1)/cip1 and p57(kip2).


Assuntos
Proteína BRCA1/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Animais , Linfócitos B/citologia , Proteína BRCA1/genética , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/genética , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Regulação para Baixo , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos
17.
Methods Mol Biol ; 2713: 11-43, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37639113

RESUMO

Macrophages are vital to the physiological function of most tissues, but also contribute to disease through a multitude of pathological roles. They are thus highly plastic and heterogeneous. It is now well recognized that macrophages develop from several distinct progenitors from embryogenesis onwards and extending throughout life. Tissue-resident macrophages largely originate from embryonic sources and in many cases self-maintain independently without monocyte input. However, in certain tissues, monocyte-derived macrophages replace these over time or as a result of tissue injury and inflammation. This additional layer of heterogeneity has introduced many questions regarding the influence of origin on fate and function of macrophages in health and disease. To comprehensively address these questions, appropriate methods of tracing macrophage ontogeny are required. This chapter explores why ontogeny is of vital importance in macrophage biology and how to delineate macrophage populations by origin through genetic fate mapping. First, we summarize the current view of macrophage ontogeny and briefly discuss how origin may influence macrophage function in homeostasis and pathology. We go on to make the case for genetic fate mapping as the gold standard and briefly review different fate-mapping models. We then put forward our recommendations for fate-mapping strategies best suited to answer specific research questions and finally discuss the strengths and limitations of currently available models.


Assuntos
Linhagem da Célula , Marcadores Genéticos , Macrófagos , Macrófagos/citologia , Monócitos/citologia , Células-Tronco Hematopoéticas/citologia , Animais , Camundongos , Diferenciação Celular
18.
Cell Rep ; 43(5): 114251, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38761374

RESUMO

Phagocytic macrophages are crucial for innate immunity and tissue homeostasis. Most tissue-resident macrophages develop from embryonic precursors that populate every organ before birth to lifelong self-renew. However, the mechanisms for versatile macrophage differentiation remain unknown. Here, we use in vivo genetic and cell biological analysis of the Drosophila larval hematopoietic organ, the lymph gland that produces macrophages. We show that the developmentally regulated transient activation of caspase-activated DNase (CAD)-mediated DNA strand breaks in intermediate progenitors is essential for macrophage differentiation. Insulin receptor-mediated PI3K/Akt signaling regulates the apoptosis signal-regulating kinase 1 (Ask1)/c-Jun kinase (JNK) axis to control sublethal levels of caspase activation, causing DNA strand breaks during macrophage development. Furthermore, caspase activity is also required for embryonic-origin macrophage development and efficient phagocytosis. Our study provides insights into developmental signaling and CAD-mediated DNA strand breaks associated with multifunctional and heterogeneous macrophage differentiation.


Assuntos
Diferenciação Celular , Dano ao DNA , Proteínas de Drosophila , Macrófagos , Fagocitose , Animais , Macrófagos/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Transdução de Sinais , Caspases/metabolismo , Ativação Enzimática , Desoxirribonucleases/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo
19.
Biochem Biophys Res Commun ; 440(4): 545-50, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24103757

RESUMO

Interleukin (IL)-3, a multilineage hematopoietic growth factor, is implicated in the regulation of osteoclastogenesis. However, the role of IL-3 in osteoclastogenesis remains controversial; whereas early studies showed that IL-3 stimulates osteoclastogenesis, recent investigations demonstrated that IL-3 inhibits osteoclast formation. The objective of this work is to further address the role of IL-3 in osteoclastogenesis. We found that IL-3 treatment of bone marrow cells generated a population of cells capable of differentiating into osteoclasts in tissue culture dishes in response to the stimulation of the monocyte/macrophage-colony stimulating factor (M-CSF) and the receptor activator of nuclear factor kappa B ligand (RANKL). The IL-3-dependent hematopoietic cells were able to further proliferate and differentiate in response to M-CSF stimulation and the resulting cells were also capable of forming osteoclasts with M-CSF and RANKL treatment. Interestingly, IL-3 inhibits M-CSF-/RANKL-induced differentiation of the IL-3-dependent hematopoietic cells into osteoclasts. The flow cytometry analysis indicates that while IL-3 treatment of bone marrow cells slightly affected the percentage of osteoclast precursors in the surviving populations, it considerably increased the percentage of osteoclast precursors in the populations after subsequent M-CSF treatment. Moreover, osteoclasts derived from IL-3-dependent hematopoietic cells were fully functional. Thus, we conclude that IL-3 plays dual roles in osteoclastogenesis by promoting the development of osteoclast progenitors but inhibiting the osteoclastogenic process. These findings provide a better understanding of the role of IL-3 in osteoclastogenesis.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Interleucina-3/fisiologia , Osteoclastos/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interleucina-3/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL
20.
Blood Cells Mol Dis ; 51(4): 220-5, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24095199

RESUMO

Erythro-myeloid progenitors (EMP) serve as a major source of hematopoiesis in the developing conceptus prior to the formation of a permanent blood system. In this review, we summarize the current knowledge regarding the emergence, fate, and potential of this hematopoietic stem cell (HSC)-independent wave of hematopoietic progenitors, focusing on the murine embryo as a model system. A better understanding of the temporal and spatial control of hematopoietic emergence in the embryo will ultimately improve our ability to derive hematopoietic stem and progenitor cells from embryonic stem cells and induced pluripotent stem cells to serve therapeutic purposes.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/fisiologia , Hematopoese/fisiologia , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/fisiologia , Animais , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA