Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Redox Biol ; 54: 102392, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35797799

RESUMEN

Granulocyte-monocyte progenitors (GMPs) differentiate into both neutrophils and monocytes. Recently, uni-potential neutrophil progenitors have been identified both in mice and humans using an array of surface markers. However, how human GMPs commit to neutrophil progenitors and the regulatory mechanisms of fate determination remain incompletely understood. In the present study, we established a human neutrophil deficiency model using the small molecule alpha-lipoic acid. Using this neutrophil deficiency model, we determined that the neutrophil progenitor commitment process from CD371+ CD115- GMPs defined by CD34 and CD15 and discovered that critical signals generated by RNA splicing and rRNA biogenesis regulate the process of early commitment for human early neutrophil progenitors derived from CD371+ CD115- GMPs. These processes were elucidated by single-cell RNA sequencing both in vitro and in vivo derived cells. Sequentially, we identified that the transcription factor ELK1 is essential for human neutrophil lineage commitment using the alpha-lipoic acid (ALA)-inducing neutrophil deficiency model. Finally, we also revealed differential roles for long-ELK1 and short-ELK1, balanced by SF3B1, in the commitment process of neutrophil progenitors. Taken together, we discovered a novel function of ALA in regulating neutrophil lineage specification and identified that the SF3B1-ELK axis regulates the commitment of human neutrophil progenitors from CD371+ CD115- GMPs.


Asunto(s)
Neutrófilos , Ácido Tióctico , Diferenciación Celular/genética , Humanos , Monocitos , Células Progenitoras Mieloides/fisiología , Ácido Tióctico/farmacología
2.
Front Immunol ; 12: 621665, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815375

RESUMEN

Toll-like receptor 5 (TLR5) is the receptor of bacterial Flagellin. Reportedly, TLR5 engagement helps to combat infections, especially at mucosal sites, by evoking responses from epithelial cells and immune cells. Here we report that TLR5 is expressed on a previously defined bipotent progenitor of macrophages (MΦs) and osteoclasts (OCs) that resides in the mouse bone marrow (BM) and circulates at low frequency in the blood. In vitro, Flagellin promoted the generation of MΦs, but not OCs from this progenitor. In vivo, MΦ/OC progenitors were recruited from the blood into the lung upon intranasal inoculation of Flagellin, where they rapidly differentiated into MΦs. Recruitment of the MΦ/OC progenitors into the lung was likely promoted by the CCL2/CCR2 axis, since the progenitors expressed CCR2 and type 2 alveolar epithelial cells (AECs) produced CCL2 upon stimulation by Flagellin. Moreover, CCR2 blockade reduced migration of the MΦ/OC progenitors toward lung lavage fluid (LLF) from Flagellin-inoculated mice. Our study points to a novel role of the Flagellin/TLR5 axis in recruiting circulating MΦ/OC progenitors into infected tissue and stimulating these progenitors to locally differentiate into MΦs. The progenitor pathway to produce MΦs may act, next to monocyte recruitment, to fortify host protection against bacterial infection at mucosal sites.


Asunto(s)
Flagelina/metabolismo , Pulmón/inmunología , Macrófagos/fisiología , Células Progenitoras Mieloides/fisiología , Osteoclastos/fisiología , Receptor Toll-Like 5/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Quimiocina CCL2/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/metabolismo , Transducción de Señal
3.
Elife ; 92020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33168134

RESUMEN

Atherosclerosis is the major cause of cardiovascular disease (CVD). Monocyte-derived macrophages are the most abundant immune cells in atherosclerotic plaques. In patients with atherosclerotic CVD, leukocytes have a hyperinflammatory phenotype. We hypothesize that immune cell reprogramming in these patients occurs at the level of myeloid progenitors. We included 13 patients with coronary artery disease due to severe atherosclerosis and 13 subjects without atherosclerosis in an exploratory study. Cytokine production capacity after ex vivo stimulation of peripheral blood mononuclear cells (MNCs) and bone marrow MNCs was higher in patients with atherosclerosis. In BM-MNCs this was associated with increased glycolysis and oxidative phosphorylation. The BM composition was skewed towards myelopoiesis and transcriptome analysis of HSC/GMP cell populations revealed enrichment of neutrophil- and monocyte-related pathways. These results show that in patients with atherosclerosis, activation of innate immune cells occurs at the level of myeloid progenitors, which adds exciting opportunities for novel treatment strategies.


Asunto(s)
Aterosclerosis/metabolismo , Células de la Médula Ósea/fisiología , Técnicas de Reprogramación Celular , Enfermedad de la Arteria Coronaria/metabolismo , Leucocitos Mononucleares/fisiología , Células Progenitoras Mieloides/fisiología , Anciano , Estudios de Casos y Controles , Diferenciación Celular , Proliferación Celular , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Masculino , Persona de Mediana Edad
4.
Mech Ageing Dev ; 192: 111378, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33022333

RESUMEN

Hematopoietic stem cells (HSCs) are characterized by two key features: Self-renewal ability and multilineage differentiation potential (multipotentiality). With aging, these key features gradually change. This is thought to be related to hematological diseases. However, clonal in vivo analysis assessing the potential of HSCs to differentiate along erythroid and platelet lineages ("five-lineage tracing") has not been performed in the aged bone marrow. By contrast, in young HSCs clonal in vivo analysis combined with five-lineage tracing has provided us with novel insights into HSC biology. Understanding HSC aging at the clonal level will help us to elucidate aging mechanisms and disease progression. We review recent progress towards understanding HSC aging at the clonal cell level in the transplantation setting.


Asunto(s)
Diferenciación Celular , Transdiferenciación Celular , Senescencia Celular/fisiología , Hematopoyesis Clonal/fisiología , Células Madre Hematopoyéticas/fisiología , Células Progenitoras Mieloides/fisiología , Animales , Linaje de la Célula , Autorrenovación de las Células/fisiología , Ensayo de Unidades Formadoras de Colonias , Humanos
5.
Front Immunol ; 10: 2133, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552057

RESUMEN

Cardiotrophin-like cytokine factor 1 (CLCF1) is secreted as a complex with the cytokine receptor-like factor 1 (CRLF1). Syndromes caused by mutations in the genes encoding CLCF1 or CRLF1 suggest an important role for CLCF1 in the development and regulation of the immune system. In mice, CLCF1 induces B-cell expansion, enhances humoral responses and triggers autoimmunity. Interestingly, inactivation of CRLF1, which impedes CLCF1 secretion, leads to a marked reduction in the number of bone marrow (BM) progenitor cells, while mice heterozygous for CLCF1 display a significant decrease in their circulating leukocytes. We therefore hypothesized that CLCF1 might be implicated in the regulation of hematopoiesis. To test this hypothesis, murine hematopoietic progenitor cells defined as Lin-Sca1+c-kit+ (LSK) were treated in vitro with ascending doses of CLCF1. The frequency and counts of LSK cells were significantly increased in the presence of CLCF1, which may be mediated by several CLCF1-induced soluble factors including IL-6, G-CSF, IL-1ß, IL-10, and VEGF. CLCF1 administration to non-diseased C57BL/6 mice resulted in a pronounced increase in circulating myeloid cells, which was concomitant with augmented LSK and myeloid cell counts in the BM. Likewise, CLCF1 administration to mice following sub-lethal irradiation or congeneic BM transplantation (BMT) resulted in accelerated LSK recovery along with a sustained increase in BM-derived CD11b+ cells. Altogether, our observations establish an important and unforeseen role for CLCF1 in regulating hematopoiesis with a bias toward myeloid cell differentiation.


Asunto(s)
Hematopoyesis , Células Progenitoras Mieloides/fisiología , Receptores de Citocinas/genética , Animales , Femenino , Hematopoyesis/efectos de los fármacos , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/efectos de los fármacos , Proteínas Recombinantes/farmacología
6.
Cell Metab ; 30(4): 609-613, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31477497

RESUMEN

Hess et al. quantified circulating aldehyde dehydrogenase-expressing (ALDHhi) cell subsets in people with T2DM given either empagliflozin (EMPA) or placebo. EMPA treatment increased circulating pro-angiogenic CD133+ progenitor cells, decreased pro-inflammatory ALDHhi granulocyte precursors, and increased ALDHhi monocytes with M2 polarization. EMPA treatment improved T2DM-associated "regenerative cell depletion" contributing to enhanced vascular health.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Cardiotónicos/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Diabetes Mellitus Tipo 2/complicaciones , Glucósidos/farmacología , Células Progenitoras Mieloides/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Compuestos de Bencidrilo/uso terapéutico , Cardiotónicos/uso terapéutico , Enfermedades Cardiovasculares/complicaciones , Glucósidos/uso terapéutico , Humanos , Persona de Mediana Edad , Células Progenitoras Mieloides/fisiología , Regeneración , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
7.
Transfusion ; 59(9): 2938-2951, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31251408

RESUMEN

BACKGROUND: Dengue virus (DENV) is a significant threat to public health in tropical and subtropical regions, where the frequency of human migration is increasing. Transmission of DENV from donors to recipients after hematopoietic stem cell transplantation has been steadily described. However, the underlying mechanisms remain unclear. STUDY DESIGN AND METHODS: Freshly isolated bone marrow (BM) was subjected to DENV infection, followed by multicolor fluorescence-activated cell sorting (FACS) analysis. Virus in supernatants was collected and analyzed by plaque assay. RESULTS: DENV-1 to DENV-4 could effectively infect freshly obtained BM and produced infectious virus. DENV infection did not change the quantitative population of hematopoietic stem and progenitor cells (HSPCs), megakaryocytic progenitor cells (MkPs) and megakaryocytes. Additionally, DENV antigen, nonstructural protein 1, was enriched in HSPCs and MkPs of DENV infected marrow cells. CD34+, CD133+, or CD61+ cells sorted out from BM were not only the major contributing targets facilitating the DENV infection directly but also facilitated the spread of DENV into other cells when cocultured. CONCLUSION: Results suggest that DENV can efficiently infect HSPCs, which might jeopardize the recipients if DENV-infected cells were subsequently used. We therefore raise the need for DENV screening for both the donors and recipients of hematopoietic stem cell transplantation, especially for donors exposed to endemic areas, to mitigate DENV infection in immunocompromised recipients.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Dengue/patología , Dengue/transmisión , Células Madre Hematopoyéticas/virología , Ensayo de Placa Viral , Antígenos Virales/análisis , Antígenos Virales/aislamiento & purificación , Células de la Médula Ósea/patología , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/virología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Dengue/sangre , Virus del Dengue/patogenicidad , Sangre Fetal/citología , Sangre Fetal/virología , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunofenotipificación , Megacariocitos/patología , Megacariocitos/fisiología , Megacariocitos/virología , Células Progenitoras Mieloides/patología , Células Progenitoras Mieloides/fisiología , Células Progenitoras Mieloides/virología
9.
Immunol Invest ; 48(1): 96-105, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30204524

RESUMEN

Previously, we found that dual therapy by the CXCR4 inhibitor Plerixafor and cytosine arabinoside (Ara-C) effectively eradicated leukemia cells and concurrently activated immune cells in acute myeloid leukemia (AML). To reveal the significance of programmed death-ligand1 (PD-L1) in AML and as a strategic approach, we investigated the anti-leukemic effect of a triple combinational therapy by utilizing Plerixafor and anti-PD-L1 in combination with chemotherapy in an AML mouse model. We examined leukemic myeloid blast cells in multiple organs after the successive treatment with Ara-C, Plerixafor, and anti-PD-L1. The results showed that noticeable benefits of triple combinational therapy for eradication of myeloid blast cells in vivo with prolonged survival rates. The frequencies of regulatory T cells (Tregs), monocytic-myeloid-derived suppressor cells (M-MDSCs), and granulocytic-myeloid-derived suppressor cells (G-MDSCs), in the peripheral blood of leukemic mice were consistently decreased, even when mice were sacrificed alive at D + 26 after completion of the triple combinational therapy, compared to the other subgroups. These findings imply that the modulation by the triple combinational therapy may lead to more efficient leukemic myeloid blast cell ablation through the suppression of Tregs or M-MDSCs and G-MDSCs in AML. Although Plerixafor and PD-L1 antagonist do not have a direct anti-leukemic role, our results provide some clues and guidelines to develop clinically therapeutic strategies for chemotherapy-resistant patients by the modulation of leukemic microenvironments.


Asunto(s)
Antígeno B7-H1/inmunología , Inmunoterapia/métodos , Leucemia Mieloide Aguda/terapia , Células Progenitoras Mieloides/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bencilaminas , Línea Celular Tumoral , Ciclamas , Citarabina/uso terapéutico , Modelos Animales de Enfermedad , Compuestos Heterocíclicos/uso terapéutico , Humanos , Inmunomodulación , Leucemia Mieloide Aguda/inmunología , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/fisiología , Microambiente Tumoral/efectos de los fármacos
10.
J Mol Med (Berl) ; 96(11): 1267-1277, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30324339

RESUMEN

The function and mechanism of action of MLL-TET1 (MT1) fusion protein in hematological cells are unclear and require further investigation. In the present study, we found that the MT1 fusion protein attenuated the expression of Cebpa, Csf1r, and Cd11b and inhibited the differentiation of myeloid progenitor cells. Increased binding of the MT1 fusion protein to the Trib2 promoter upregulated Trib2 mRNA and protein expression and downregulated Cebpa expression. Trib2 knockdown relieved the inhibition of myeloid cell differentiation induced by the MT1 fusion protein. Thus, TRIB2 is important for the survival of leukemia cells during MT1-related leukemogenesis and is important in maintaining differentiation blockade of leukemic cells. KEY MESSAGES: • MLL-TET1 fusion decreases the 5-hmC levels in the myeloid progenitor cells. • MLL-TET1 fusion inhibits myeloid differentiation through decreased expression of Cebpa. • MLL-TET1 fusion blocks the differentiation of the myeloid progenitor cells by overexpressing Trib2. • Knockdown of Trib2 in MLL-TET1 transduced cells induces myeloid differentiation.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Diferenciación Celular/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oxigenasas de Función Mixta/metabolismo , Células Progenitoras Mieloides/fisiología , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7
11.
J Hepatol ; 69(6): 1294-1307, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171870

RESUMEN

BACKGROUND & AIMS: The liver is the main hematopoietic site in embryos, becoming a crucial organ in both immunity and metabolism in adults. However, how the liver adapts both the immune system and enzymatic profile to challenges in the postnatal period remains elusive. We aimed to identify the mechanisms underlying this adaptation. METHODS: We analyzed liver samples from mice on day 0 after birth until adulthood. Human biopsies from newborns and adults were also examined. Liver immune cells were phenotyped using mass cytometry (CyTOF) and expression of several genes belonging to immune and metabolic pathways were measured. Mortality rate, bacteremia and hepatic bacterial retention after E. coli challenge were analyzed using intravital and in vitro approaches. In a set of experiments, mice were prematurely weaned and the impact on gene expression of metabolic pathways was evaluated. RESULTS: Human and mouse newborns have a sharply different hepatic cellular composition and arrangement compared to adults. We also found that myeloid cells and immature B cells primarily compose the neonatal hepatic immune system. Although neonatal mice were more susceptible to infections, a rapid evolution to an efficient immune response was observed. Concomitantly, newborns displayed a reduction of several macronutrient metabolic functions and the normal expression level of enzymes belonging to lipid and carbohydrate metabolism was reached around the weaning period. Interestingly, early weaning profoundly disturbed the expression of several hepatic metabolic pathways, providing novel insights into how dietary schemes affect the metabolic maturation of the liver. CONCLUSION: In newborns, the immune and metabolic profiles of the liver are dramatically different to those of the adult liver, which can be explained by the differences in the liver cell repertoire and phenotype. Also, dietary and antigen cues may be crucial to guide liver development during the postnatal phase. LAY SUMMARY: Newborns face major challenges in the extra-uterine life. In fact, organs need to modify their cellular composition and gene expression profile in order to adapt to changes in both microbiota and diet throughout life. The liver is interposed between the gastrointestinal system and the systemic circulation, being the destination of all macronutrients and microbial products from the gut. Therefore, it is expected that delicately balanced mechanisms govern the transformation of a neonatal liver to a key organ in adults.


Asunto(s)
Recién Nacido , Hígado/inmunología , Hígado/metabolismo , Adulto , Animales , Animales Recién Nacidos , Biopsia , Infecciones por Escherichia coli/inmunología , Femenino , Hepatocitos , Humanos , Metabolismo de los Lípidos , Hígado/citología , Metaboloma , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/fisiología , Valor Nutritivo/fisiología , Fagocitos/inmunología , Células Precursoras de Linfocitos B/inmunología , Destete
12.
Sci Rep ; 8(1): 13005, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158544

RESUMEN

Osteoclasts are the bone resorbing cells that derive from myeloid progenitor cells. Although there have been recent advancements in the ability to identify osteoclast progenitors, very little is known about the molecular mechanisms governing their homeostasis. Here, by analyzing the normalized phylogenetic profiles of the Schlafen (Slfn) gene family, we found that it co-evolved with osteoclast-related genes. Following these findings, we used a Slfn2 loss-of-function mutant mouse, elektra, to study the direct role of Slfn2 in osteoclast development and function. Slfn2eka/eka mice exhibited a profound increase in their cancellous bone mass and a significant reduction in osteoclast numbers. In addition, monocyte cultures from the bone marrow of Slfn2eka/eka mice showed a reduction in osteoclast number and total resorption area. Finally, we show that the bone marrow of Slfn2eka/eka mice have significantly less CD11b-Ly6Chi osteoclast precursors. Overall, our data suggest that Slfn2 is required for normal osteoclast differentiation and that loss of its function in mice results in an osteopetrotic phenotype.


Asunto(s)
Proteínas de Ciclo Celular/deficiencia , Diferenciación Celular , Mutación , Células Progenitoras Mieloides/fisiología , Osteoclastos/fisiología , Osteopetrosis/genética , Osteopetrosis/patología , Animales , Ratones
13.
Thromb Haemost ; 118(9): 1586-1599, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30103245

RESUMEN

Thromboembolic events are the main cause of mortality in BCR-ABL1-negative myeloproliferative neoplasms (MPNs) but their underlying mechanisms are largely unrecognized. The Janus kinase 2 (JAK2)V617F mutation is the most frequent genetic alteration leading to MPN. Usually found in haematopoietic progenitors and stem cells, this mutation has also been described in endothelial cells (ECs) of MPN patients. In this study, we have questioned the impact of the JAK2V617F mutation on EC phenotype and functions. We developed an induced pluripotent stem cells strategy to compare JAK2 mutant and wild-type ECs. Transcriptomic assays showed that several genes and pathways involved in inflammation, cell adhesion and thrombotic events were over-represented in JAK2V617F ECs and expression levels of von Willebrand factor and P-selectin (CD62P) proteins were increased. Finally, we found that leucocytes from MPN patients adhere more tightly to JAK2V617F ECs. Our results show that JAK2V617F ECs have a pro-inflammatory and pro-thrombotic phenotype and were functionally pro-adherent.


Asunto(s)
Plaquetas/fisiología , Células Endoteliales/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Janus Quinasa 2/genética , Células Progenitoras Mieloides/fisiología , Trastornos Mieloproliferativos/genética , Trombosis/genética , Adhesión Celular/genética , Diferenciación Celular , Células Cultivadas , Proteínas de Fusión bcr-abl/metabolismo , Perfilación de la Expresión Génica , Humanos , Mutación/genética , Transgenes/genética
14.
Dev Biol ; 442(1): 138-154, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30016639

RESUMEN

Macrophages are well characterized as immune cells. However, in recent years, a multitude of non-immune functions have emerged many of which play essential roles in a variety of developmental processes (Wynn et al., 2013; DeFalco et al., 2014). In adult animals, macrophages are derived from circulating monocytes originating in the bone marrow, but much of the tissue-resident population arise from erythro-myeloid progenitors (EMPs) in the extra-embryonic yolk sac, appearing around the same time as primitive erythroblasts (Schulz et al., 2012; Kierdorf et al., 2013; McGrath et al., 2015; Gomez Perdiguero et al., 2015; Mass et al., 2016). Of particular interest to our group, macrophages have been shown to act as pro-angiogenic regulators during development (Wynn et al., 2013; DeFalco et al., 2014; Hsu et al., 2015), but there is still much to learn about these early cells. The goal of the present study was to isolate and expand progenitors of yolk-sac-derived Embryonic Macrophages (EMs) in vitro to generate a new platform for mechanistic studies of EM differentiation. To accomplish this goal, we isolated pure (>98%) EGFP+ populations by flow cytometry from embryonic day 9.5 (E9.5) Csf1r-EGFP+/tg mice, then evaluated the angiogenic potential of EMs relative to Bone Marrow-Derived Macrophages (BMDMs). We found that EMs expressed more pro-angiogenic and less pro-inflammatory macrophage markers than BMDMs. EMs also promoted more endothelial cell (EC) cord formation in vitro, as compared to BMDMs in a manner that required direct cell-to-cell contact. Importantly, EMs preferentially matured into microglia when co-cultured with mouse Neural Stem/Progenitor Cells (NSPCs). In conclusion, we have established a protocol to isolate and propagate EMs in vitro, have further defined specialized properties of yolk-sac-derived macrophages, and have identified EM-EC and EM-NSPC interactions as key inducers of EC tube formation and microglial cell maturation, respectively.


Asunto(s)
Células Precursoras Eritroides/fisiología , Macrófagos/fisiología , Células Progenitoras Mieloides/fisiología , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/fisiología , Técnicas de Cocultivo/métodos , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/fisiología , Macrófagos/citología , Ratones/embriología , Fenotipo , Saco Vitelino/citología
15.
J Immunol Res ; 2018: 5370414, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30622977

RESUMEN

Brucellosis is a zoonotic bacterial infection that may persist for long periods causing relapses in antibiotic-treated patients. The ability of Brucella to develop chronic infections is linked to their capacity to invade and replicate within the mononuclear phagocyte system, including the bone marrow (BM). Persistence of Brucella in the BM has been associated with hematological complications such as neutropenia, thrombocytopenia, anemia, and pancytopenia in human patients. In the mouse model, we observed that the number of Brucella abortus in the BM remained constant for up to 168 days of postinfection. This persistence was associated with histopathological changes, accompanied by augmented numbers of BM myeloid GMP progenitors, PMNs, and CD4+ lymphocytes during the acute phase (eight days) of the infection in the BM. Monocytes, PMNs, and GMP cells were identified as the cells harboring Brucella in the BM. We propose that the BM is an essential niche for the bacterium to establish long-lasting infections and that infected PMNs may serve as vehicles for dispersion of Brucella organisms, following the Trojan horse hypothesis. Monocytes are solid candidates for Brucella reservoirs in the BM.


Asunto(s)
Médula Ósea/microbiología , Brucella abortus/fisiología , Brucelosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/fisiología , Neutrófilos/inmunología , Animales , Autofagia , Bovinos , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Ratones
16.
Immunity ; 47(5): 890-902.e4, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166589

RESUMEN

Granulocyte-monocyte progenitors (GMPs) and monocyte-dendritic cell progenitors (MDPs) produce monocytes during homeostasis and in response to increased demand during infection. Both progenitor populations are thought to derive from common myeloid progenitors (CMPs), and a hierarchical relationship (CMP-GMP-MDP-monocyte) is presumed to underlie monocyte differentiation. Here, however, we demonstrate that mouse MDPs arose from CMPs independently of GMPs, and that GMPs and MDPs produced monocytes via similar but distinct monocyte-committed progenitors. GMPs and MDPs yielded classical (Ly6Chi) monocytes with gene expression signatures that were defined by their origins and impacted their function. GMPs produced a subset of "neutrophil-like" monocytes, whereas MDPs gave rise to a subset of monocytes that yielded monocyte-derived dendritic cells. GMPs and MDPs were also independently mobilized to produce specific combinations of myeloid cell types following the injection of microbial components. Thus, the balance of GMP and MDP differentiation shapes the myeloid cell repertoire during homeostasis and following infection.


Asunto(s)
Células Dendríticas/fisiología , Células Precursoras de Granulocitos/fisiología , Monocitos/fisiología , Células Progenitoras Mieloides/fisiología , Animales , Antígenos Ly/análisis , Diferenciación Celular , Leucosialina/análisis , Ratones , Análisis de Secuencia de ARN , Transcriptoma
17.
EMBO J ; 36(24): 3619-3633, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29030486

RESUMEN

Single-cell RNA sequencing is a powerful technology for assessing heterogeneity within defined cell populations. Here, we describe the heterogeneity of a B220+CD117intCD19-NK1.1- uncommitted hematopoietic progenitor having combined lymphoid and myeloid potential. Phenotypic and functional assays revealed four subpopulations within the progenitor with distinct lineage developmental potentials. Among them, the Ly6D+SiglecH-CD11c- fraction was lymphoid-restricted exhibiting strong B-cell potential, whereas the Ly6D-SiglecH-CD11c- fraction showed mixed lympho-myeloid potential. Single-cell RNA sequencing of these subsets revealed that the latter population comprised a mixture of cells with distinct lymphoid and myeloid transcriptional signatures and identified a subgroup as the potential precursor of Ly6D+SiglecH-CD11c- Subsequent functional assays confirmed that B220+CD117intCD19-NK1.1- single cells are, with rare exceptions, not bipotent for lymphoid and myeloid lineages. A B-cell priming gradient was observed within the Ly6D+SiglecH-CD11c- subset and we propose a herein newly identified subgroup as the direct precursor of the first B-cell committed stage. Therefore, the apparent multipotency of B220+CD117intCD19-NK1.1- progenitors results from underlying heterogeneity at the single-cell level and highlights the validity of single-cell transcriptomics for resolving cellular heterogeneity and developmental relationships among hematopoietic progenitors.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Análisis de Secuencia de ARN/métodos , Animales , Linfocitos B/citología , Linfocitos B/fisiología , Diferenciación Celular , Linaje de la Célula , Femenino , Perfilación de la Expresión Génica , Heterogeneidad Genética , Células Madre Hematopoyéticas/citología , Secuenciación de Nucleótidos de Alto Rendimiento , Células Progenitoras Linfoides/citología , Células Progenitoras Linfoides/fisiología , Masculino , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/fisiología , Análisis de la Célula Individual
19.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27695000

RESUMEN

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Asunto(s)
Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Células Progenitoras Linfoides/fisiología , Células Progenitoras Mieloides/fisiología , Receptores Notch/metabolismo , Linfocitos T/fisiología , Timo/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Células Cultivadas , Feto , Regulación del Desarrollo de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
20.
Eur J Immunol ; 46(12): 2737-2748, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27601030

RESUMEN

CSF1R (colony stimulating factor 1 receptor) is the main receptor for CSF1 and has crucial roles in regulating myelopoeisis. CSF1R can be proteolytically released from the cell surface by ADAM17 (A disintegrin and metalloprotease 17). Here, we identified CSF1R as a major substrate of ADAM17 in an unbiased degradomics screen. We explored the impact of CSF1R shedding by ADAM17 and its upstream regulator, inactive rhomboid protein 2 (iRhom2, gene name Rhbdf2), on homeostatic development of mouse myeloid cells. In iRhom2-/- mice, we found constitutive accumulation of membrane-bound CSF1R on myeloid cells at steady state, although cell numbers of these populations were not altered. However, in the context of mixed bone marrow (BM) chimera, under competitive pressure, iRhom2-/- BM progenitor-derived monocytes, tissue macrophages and lung DCs showed a repopulation advantage over those derived from wild-type (WT) BM progenitors, suggesting enhanced CSF1R signaling in the absence of iRhom2. In vitro experiments indicate that iRhom2-/- Lin- SCA-1+ c-Kit+ (LSKs) cells, but not granulocyte-macrophage progenitors (GMPs), had faster growth rates than WT cells in response to CSF1. Our results shed light on an important role of iRhom2/ADAM17 pathway in regulation of CSF1R shedding and repopulation of monocytes, macrophages and DCs.


Asunto(s)
Proteína ADAM17/metabolismo , Células de la Médula Ósea/fisiología , Proteínas Portadoras/metabolismo , Células Progenitoras Mieloides/fisiología , Mielopoyesis , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteína ADAM17/genética , Animales , Proteínas Portadoras/genética , Células Cultivadas , Células Dendríticas/fisiología , Femenino , Regulación de la Expresión Génica , Pulmón/patología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Transducción de Señal , Quimera por Trasplante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...