Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
1.
Rinsho Ketsueki ; 64(9): 853-860, 2023.
Artículo en Japonés | MEDLINE | ID: mdl-37793858

RESUMEN

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.


Asunto(s)
Células Madre Hematopoyéticas , Mielopoyesis , Humanos , Mielopoyesis/fisiología , Diferenciación Celular , Factores de Transcripción , Células Mieloides
2.
Nat Immunol ; 23(1): 109-121, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937919

RESUMEN

Anemia is a major comorbidity in aging, chronic kidney and inflammatory diseases, and hematologic malignancies. However, the transcriptomic networks governing hematopoietic differentiation in blood cell development remain incompletely defined. Here we report that the atypical kinase RIOK2 (right open reading frame kinase 2) is a master transcription factor (TF) that not only drives erythroid differentiation, but also simultaneously suppresses megakaryopoiesis and myelopoiesis in primary human stem and progenitor cells. Our study reveals the previously uncharacterized winged helix-turn-helix DNA-binding domain and two transactivation domains of RIOK2 that are critical to regulate key hematopoietic TFs GATA1, GATA2, SPI1, RUNX3 and KLF1. This establishes RIOK2 as an integral component of the transcriptional regulatory network governing human hematopoietic differentiation. Importantly, RIOK2 mRNA expression significantly correlates with these TFs and other hematopoietic genes in myelodysplastic syndromes, acute myeloid leukemia and chronic kidney disease. Further investigation of RIOK2-mediated transcriptional pathways should yield therapeutic approaches to correct defective hematopoiesis in hematologic disorders.


Asunto(s)
Células Sanguíneas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Eritropoyesis/fisiología , Regulación de la Expresión Génica/fisiología , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Humanos , Células K562 , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicos/metabolismo , Mielopoyesis/fisiología , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología
3.
Nat Cell Biol ; 24(1): 99-111, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34961794

RESUMEN

Histone variants and the associated post-translational modifications that govern the stemness of haematopoietic stem cells (HSCs) and differentiation thereof into progenitors (HSPCs) have not been well defined. H3.3 is a replication-independent H3 histone variant in mammalian systems that is enriched at both H3K4me3- and H3K27me3-marked bivalent genes as well as H3K9me3-marked endogenous retroviral repeats. Here we show that H3.3, but not its chaperone Hira, prevents premature HSC exhaustion and differentiation into granulocyte-macrophage progenitors. H3.3-null HSPCs display reduced expression of stemness and lineage-specific genes with a predominant gain of H3K27me3 marks at their promoter regions. Concomitantly, loss of H3.3 leads to a reduction of H3K9me3 marks at endogenous retroviral repeats, opening up binding sites for the interferon regulatory factor family of transcription factors, allowing the survival of rare, persisting H3.3-null HSCs. We propose a model whereby H3.3 maintains adult HSC stemness by safeguarding the delicate interplay between H3K27me3 and H3K9me3 marks, enforcing chromatin adaptability.


Asunto(s)
Cromatina/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Mielopoyesis/fisiología , Animales , Linfocitos T CD8-positivos/citología , Proteínas de Ciclo Celular , Línea Celular , Granulocitos/citología , Hematopoyesis/fisiología , Chaperonas de Histonas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Macrófagos/citología , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/fisiología , Factores de Transcripción
4.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34353901

RESUMEN

Dyskeratosis congenita (DC) is a rare inherited bone marrow failure and cancer predisposition syndrome caused by mutations in telomerase or telomeric proteins. Here, we report that zebrafish telomerase RNA (terc) binds to specific DNA sequences of master myeloid genes and controls their expression by recruiting RNA Polymerase II (Pol II). Zebrafish terc harboring the CR4-CR5 domain mutation found in DC patients hardly interacted with Pol II and failed to regulate myeloid gene expression in vivo and to increase their transcription rates in vitro. Similarly, TERC regulated myeloid gene expression and Pol II promoter occupancy in human myeloid progenitor cells. Strikingly, induced pluripotent stem cells derived from DC patients with a TERC mutation in the CR4-CR5 domain showed impaired myelopoiesis, while those with mutated telomerase catalytic subunit differentiated normally. Our findings show that TERC acts as a transcription factor, revealing a target for therapeutic intervention in DC patients.


Asunto(s)
Disqueratosis Congénita/genética , Mielopoyesis/fisiología , ARN Polimerasa II/genética , ARN/metabolismo , Telomerasa/metabolismo , Animales , Animales Modificados Genéticamente , Sitios de Unión , Células Cultivadas , Disqueratosis Congénita/patología , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/patología , Larva/genética , Mutación , Mielopoyesis/genética , Regiones Promotoras Genéticas , Dominios Proteicos , ARN/genética , ARN Polimerasa II/metabolismo , Telomerasa/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
J Immunol ; 207(4): 1078-1086, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34341172

RESUMEN

Emergency granulopoiesis, also known as demand-adapted granulopoiesis, is defined as the response of an organism to systemic bacterial infections, and it results in neutrophil mobilization from reservoir pools and increased myelopoiesis in the bone marrow. Indirect and direct initiating mechanisms of emergency granulopoiesis have been hypothesized. However, the detailed mechanism of hyperactive myelopoiesis in the bone marrow, which leads to granulocyte left shift, remains unknown. In this study, we report that TLR4 is expressed on granulo-monocytic progenitors, as well as mobilized human peripheral blood CD34+ cells, which account for 0.2% of monocytes in peripheral blood, and ∼ 10% in bone marrow. LPS, a component of Gram-negative bacteria that results in a systemic bacterial infection, induces the differentiation of peripheral blood CD34+ cells into myelocytes and monocytes in vitro via the TLR4 signaling pathway. Moreover, CD34+ cells directly responded to LPS stimulation by activating the MAPK and NF-κB signaling pathways, and they produced IL-6 that promotes emergency granulopoiesis by phosphorylating C/EBPα and C/EBPß, and this effect was suppressed by the action of an IL-6 receptor inhibitor. This work supports the finding that TLR is expressed on human hematopoietic stem and progenitor cells, and it provides evidence that human hematopoietic stem and progenitor cells can directly sense pathogens and produce cytokines exerting autocrine and/or paracrine effects, thereby promoting differentiation.


Asunto(s)
Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-6/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Receptor Toll-Like 4/metabolismo , Adaptación Fisiológica/fisiología , Antígenos CD34/metabolismo , Médula Ósea/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/fisiología , Citocinas/metabolismo , Regulación de la Expresión Génica/fisiología , Células Precursoras de Granulocitos/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Monocitos/metabolismo , Mielopoyesis/fisiología
6.
J Exp Med ; 218(7)2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34129018

RESUMEN

Inflammation exerts multiple effects on the early hematopoietic compartment. Best studied is the role of proinflammatory cytokines in activating adult hematopoietic stem and progenitor cells to dynamically replenish myeloid lineage cells in a process known as emergency myelopoiesis. However, it is increasingly appreciated that the same proinflammatory signaling pathways are used in diverse hematopoietic scenarios. This review focuses on inflammatory signaling in the emergence of the definitive hematopoietic compartment during embryonic life, and tonic inflammatory signals derived from commensal microbiota in shaping the adult hematopoietic compartment in the absence of pathogenic insults. Insights into the unique and shared aspects of inflammatory signaling that regulate hematopoietic stem and progenitor cell function across the lifespan and health span of an individual will enable better diagnostic and therapeutic approaches to hematopoietic dysregulation and malignancies.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Inflamación/fisiopatología , Transducción de Señal/fisiología , Células Madre/fisiología , Animales , Homeostasis/fisiología , Humanos , Células Mieloides/fisiología , Mielopoyesis/fisiología
7.
Pharmacol Res ; 169: 105663, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33979688

RESUMEN

Recruitment of innate immune cells and their accumulation in the arterial wall and infarcted myocardium has been recognized as a central feature of atherosclerosis and cardiac ischemic injury, respectively. In both, steady state and under pathological conditions, majority of these cells have a finite life span and are continuously replenished from haematopoietic stem/progenitor cell pool residing in the bone marrow and extramedullary sites. While having a crucial role in the cardiovascular disease development, proliferation and differentiation of innate immune cells within haematopoietic compartments is greatly affected by the ongoing cardiovascular pathology. In the current review, we summarize key cells, processes and tissue compartments that are involved in myelopoiesis under the steady state, during atherosclerosis development and in myocardial infarction.


Asunto(s)
Médula Ósea/fisiopatología , Enfermedades Cardiovasculares/fisiopatología , Hematopoyesis Extramedular/fisiología , Mielopoyesis/fisiología , Animales , Aterosclerosis/fisiopatología , Humanos , Células Mieloides/fisiología
8.
Leuk Res ; 105: 106570, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33838549

RESUMEN

Transient abnormal myelopoiesis (TAM) in neonates with Down syndrome, which spontaneously resolves within several weeks or months after birth, may represent a special form of leukemia developing in the fetal liver (FL). To explore the role of hepatoblasts, one of the major constituents of the FL hematopoietic microenvironment, in the pathogenesis of TAM, we investigated the influence of a human hepatoblastoma cell line, HUH-6, on the in vitro growth and differentiation of TAM blasts. In a coculture system with membrane filters, which hinders cell-to-cell contact between TAM blasts and HUH-6 cells, the growth and megakaryocytic differentiation of TAM blast progenitors were increased in the presence of HUH-6 cells. The culture supernatant of HUH-6 cells contained hematopoietic growth factors, including stem cell factor (SCF) and thrombopoietin (TPO). The neutralizing antibody against SCF abrogated the growth-stimulating activity of the culture supernatant of HUH-6 cells, demonstrating that, among the growth factors produced by HUH-6 cells, SCF may be the major growth stimulator and that TPO may be involved in megakaryocytic differentiation, rather than growth, of TAM blasts. This suggests that hepatoblasts function in the regulation of the growth and differentiation of TAM blasts in the FL through the production of hematopoietic growth factors, including SCF and TPO, and are involved in the leukemogenesis of TAM.


Asunto(s)
Síndrome de Down/patología , Hepatoblastoma , Hepatocitos , Reacción Leucemoide/patología , Células Progenitoras de Megacariocitos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/fisiología , Técnicas de Cocultivo , Hepatoblastoma/metabolismo , Hepatoblastoma/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia Megacarioblástica Aguda/patología , Células Progenitoras de Megacariocitos/metabolismo , Células Progenitoras de Megacariocitos/patología , Mielopoyesis/fisiología , Células Madre/metabolismo , Células Madre/patología
9.
Blood ; 137(21): 2958-2969, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33598715

RESUMEN

Eosinophils are white blood cells that contribute to the regulation of immunity and are involved in the pathogenesis of numerous inflammatory diseases. In contrast to other cells of the immune system, no information is available regarding the role of autophagy in eosinophil differentiation and functions. To study the autophagic pathway in eosinophils, we generated conditional knockout mice in which Atg5 is deleted within the eosinophil lineage only (designated Atg5eoΔ mice). Eosinophilia was provoked by crossbreeding Atg5eoΔ mice with Il5 (IL-5) overexpressing transgenic mice (designated Atg5eoΔIl5tg mice). Deletion of Atg5 in eosinophils resulted in a dramatic reduction in the number of mature eosinophils in blood and an increase of immature eosinophils in the bone marrow. Atg5-knockout eosinophil precursors exhibited reduced proliferation under both in vitro and in vivo conditions but no increased cell death. Moreover, reduced differentiation of eosinophils in the absence of Atg5 was also observed in mouse and human models of chronic eosinophilic leukemia. Atg5-knockout blood eosinophils exhibited augmented levels of degranulation and bacterial killing in vitro. Moreover, in an experimental in vivo model, we observed that Atg5eoΔ mice achieve better clearance of the local and systemic bacterial infection with Citrobacter rodentium. Evidence for increased degranulation of ATG5low-expressing human eosinophils was also obtained in both tissues and blood. Taken together, mouse and human eosinophil hematopoiesis and effector functions are regulated by ATG5, which controls the amplitude of overall antibacterial eosinophil immune responses.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/fisiología , Eosinófilos/fisiología , Mielopoyesis/fisiología , Animales , Proteína 5 Relacionada con la Autofagia/biosíntesis , Proteína 5 Relacionada con la Autofagia/deficiencia , Proteína 5 Relacionada con la Autofagia/genética , Médula Ósea/patología , Sistemas CRISPR-Cas , Degranulación de la Célula , Línea Celular Tumoral , Células Cultivadas , Citrobacter rodentium , Ensayo de Unidades Formadoras de Colonias , Infecciones por Enterobacteriaceae/inmunología , Eosinófilos/citología , Eosinófilos/inmunología , Humanos , Síndrome Hipereosinofílico/sangre , Síndrome Hipereosinofílico/patología , Interleucina-5/genética , Recuento de Leucocitos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Escisión y Poliadenilación de ARNm/genética
10.
Front Immunol ; 11: 2120, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042124

RESUMEN

Streptococcus pneumoniae is the main cause of bacterial pneumonia, a condition that currently produces significant global morbidity and mortality. The initial immune response to this bacterium occurs when the innate system recognizes common motifs expressed by many pathogens, events driven by pattern recognition receptors like the Toll-like family receptors (TLRs). In this study, lung myeloid-cell populations responsible for the innate immune response (IIR) against S. pneumoniae, and their dependence on the TLR4-signaling axis, were analyzed in TLR4-/- and Myeloid-Differentiation factor-88 deficient (MyD88-/-) mice. Neutrophils and monocyte-derived cells were recruited in infected mice 3-days post-infection. Compared to wild-type mice, there was an increased bacterial load in both these deficient mouse strains and an altered IIR, although TLR4-/- mice were more susceptible to bacterial infection. These mice also developed fewer alveolar macrophages, weaker neutrophil infiltration, less Ly6Chigh monocyte differentiation and a disrupted classical and non-classical monocyte profile. The pro-inflammatory cytokine profile (CXCL1, TNF-α, IL-6, and IL-1ß) was also severely affected by the lack of TLR4 and no induction of Th1 was observed in these mice. The respiratory burst (ROS production) after infection was profoundly dampened in TLR4-/- and MyD88-/- mice. These data demonstrate the complex dynamics of myeloid populations and a key role of the TLR4-signaling axis in the IIR to S. pneumoniae, which involves both the MyD88 and TRIF (Toll/IL-1R domain-containing adaptor-inducing IFN-ß) dependent pathways.


Asunto(s)
Pulmón/inmunología , Monocitos/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Mielopoyesis/fisiología , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/patología , Transducción de Señal/fisiología , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 4/fisiología , Administración Intranasal , Animales , Carga Bacteriana , Citocinas/biosíntesis , Inmunidad Innata , Pulmón/patología , Macrófagos Alveolares/inmunología , Ratones , Monocitos/patología , Factor 88 de Diferenciación Mieloide/deficiencia , Infiltración Neutrófila , Especies Reactivas de Oxígeno/metabolismo , Células TH1/inmunología , Receptor Toll-Like 4/deficiencia
11.
Immunity ; 53(2): 303-318.e5, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32579887

RESUMEN

Granulocyte-monocyte progenitors (GMPs) have been previously defined for their potential to generate various myeloid progenies such as neutrophils and monocytes. Although studies have proposed lineage heterogeneity within GMPs, it is unclear if committed progenitors already exist among these progenitors and how they may behave differently during inflammation. By combining single-cell transcriptomic and proteomic analyses, we identified the early committed progenitor within the GMPs responsible for the strict production of neutrophils, which we designate as proNeu1. Our dissection of the GMP hierarchy led us to further identify a previously unknown intermediate proNeu2 population. Similar populations could be detected in human samples. proNeu1s, but not proNeu2s, selectively expanded during the early phase of sepsis at the expense of monocytes. Collectively, our findings help shape the neutrophil maturation trajectory roadmap and challenge the current definition of GMPs.


Asunto(s)
Células Precursoras de Granulocitos/citología , Monocitos/citología , Mielopoyesis/fisiología , Neutrófilos/citología , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de la Célula Individual
12.
Nat Genet ; 52(7): 655-661, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32514124

RESUMEN

Three-dimensional organization of the genome is important for transcriptional regulation1-7. In mammals, CTCF and the cohesin complex create submegabase structures with elevated internal chromatin contact frequencies, called topologically associating domains (TADs)8-12. Although TADs can contribute to transcriptional regulation, ablation of TAD organization by disrupting CTCF or the cohesin complex causes modest gene expression changes13-16. In contrast, CTCF is required for cell cycle regulation17, embryonic development and formation of various adult cell types18. To uncouple the role of CTCF in cell-state transitions and cell proliferation, we studied the effect of CTCF depletion during the conversion of human leukemic B cells into macrophages with minimal cell division. CTCF depletion disrupts TAD organization but not cell transdifferentiation. In contrast, CTCF depletion in induced macrophages impairs the full-blown upregulation of inflammatory genes after exposure to endotoxin. Our results demonstrate that CTCF-dependent genome topology is not strictly required for a functional cell-fate conversion but facilitates a rapid and efficient response to an external stimulus.


Asunto(s)
Linfocitos B/fisiología , Factor de Unión a CCCTC/fisiología , Macrófagos/fisiología , Mielopoyesis/fisiología , Antígenos de Diferenciación/metabolismo , Factor de Unión a CCCTC/genética , Línea Celular Tumoral , Proliferación Celular/fisiología , Cromatina/fisiología , Regulación de la Expresión Génica , Humanos , Conformación Molecular , Mielopoyesis/genética , Conformación Proteica
13.
FASEB J ; 34(8): 10191-10211, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32557809

RESUMEN

Osteocytes, the bone cells embedded in the mineralized matrix, control bone modeling, and remodeling through direct contact with adjacent cells and via paracrine and endocrine factors that affect cells in the bone marrow microenvironment or distant organs. Osteocytes express numerous G protein-coupled receptors (GPCRs) and thus mice lacking the stimulatory subunit of G-protein (Gsα) in osteocytes (Dmp1-GsαKO mice) have abnormal myelopoiesis, osteopenia, and reduced adipose tissue. We previously reported that the severe osteopenia and the changes in adipose tissue present in these mice were mediated by increased sclerostin, which suppress osteoblast functions and promote browning of white adipocytes. Inversely, the myeloproliferation was driven by granulocyte colony-stimulating factor (G-CSF) and administration of neutralizing antibodies against G-CSF only partially restored the myeloproliferation, suggesting that additional osteocyte-derived factors might be involved. We hypothesized that osteocytes secrete Gsα-dependent factor(s) which regulate the myeloid cells proliferation. To identify osteocyte-secreted proteins, we used the osteocytic cell line Ocy454 expressing or lacking Gsα expression (Ocy454-Gsαcont and Ocy454-GsαKO ) to delineate the osteocyte "secretome" and its regulation by Gsα. Here we reported that factors secreted by osteocytes increased the number of myeloid colonies and promoted macrophage proliferation. The proliferation of myeloid cells was further promoted by osteocytes lacking Gsα expression. Myeloid cells can differentiate into bone-resorbing osteoclasts, therefore, we hypothesized that osteocyte-secreted factors might also regulate osteoclastogenesis in a Gsα-dependent manner. Conditioned medium (CM) from Ocy454 (both Gsαcont and GsαKO ) significanlty increased the proliferation of bone marrow mononuclear cells (BMNC) and, at the same time, inhibited their differentiation into mature osteoclasts via a Gsα-dependent mechanism. Proteomics analysis of CM from Ocy454 Gsαcont and GsαKO cells identified neuropilin-1 (Nrp-1) and granulin (Grn) as osteocytic-secreted proteins upregulated in Ocy454-GsαKO cells compared to Ocy454-Gsαcont , whereas semaphorin3A was significantly suppressed. Treatment of Ocy454-Gsαcont cells with recombinant proteins or knockdown of Nrp-1 and Grn in Ocy454-GsαKO cells partially rescued the inhibition of osteoclasts, demonstrating that osteocytes control osteoclasts differentiation through Nrp-1 and Grn which are regulated by Gsα signaling.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células Mieloides/metabolismo , Células Mieloides/fisiología , Osteocitos/metabolismo , Osteocitos/fisiología , Animales , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/fisiopatología , Médula Ósea/metabolismo , Médula Ósea/fisiología , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Línea Celular , Medios de Cultivo Condicionados/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Mielopoyesis/fisiología , Osteoclastos/metabolismo , Osteoclastos/fisiología , Osteogénesis/fisiología , Transducción de Señal/fisiología
14.
Mol Immunol ; 123: 1-6, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32380279

RESUMEN

The repertoire of dendritic cells (DCs), monocytes and macrophages in adult humans is diverse and we are appreciating this to a greater extent as high throughput methods, such a single-cell RNA sequencing, become widely adopted and scalable. This powerful lens of analysis is also beginning to shed light on prenatal immunology, allowing us to chart the emergence, tissue distribution and developmental regulation of DCs, monocytes and macrophages during early human life. In this review, we will integrate recent insights from studies of the developing immune system into our understanding of adult DC, monocyte and macrophage organization, illustrating where insights from early life both affirm and challenge current understanding.


Asunto(s)
Células Dendríticas/citología , Desarrollo Fetal/fisiología , Macrófagos/citología , Monocitos/citología , Mielopoyesis/fisiología , Análisis de la Célula Individual/métodos , Adulto , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Cultivadas , Células Dendríticas/fisiología , Femenino , Humanos , Macrófagos/fisiología , Monocitos/fisiología , Embarazo
15.
Blood ; 135(24): 2146-2158, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32219443

RESUMEN

Immunomodulatory drugs (IMiDs) are key agents for the treatment of multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the recruitment of neosubstrates to cereblon, a substrate receptor of the E3 ubiquitin ligase complex; therefore, identification of cell-specific neosubstrates is important to understand the effects of IMiDs. In clinical practice, IMiDs induce thrombocytopenia, which frequently results in the discontinuation of IMiD treatment. In the current study, we sought to identify the molecular mechanism underlying thrombocytopenia induced by IMiD treatment. We found that IMiDs strongly impaired proplatelet formation, a critical step in functional platelet production, through the inhibition of autocrine estradiol signaling in human megakaryocytes. Furthermore, we identified aromatase, an indispensable enzyme for estradiol biosynthesis, as a novel neosubstrate of cereblon. IMiDs promoted the recruitment of aromatase to cereblon, resulting in the degradation of aromatase in a proteasome-dependent manner. Finally, aromatase was significantly degraded in the bone marrow of patients with multiple myeloma who developed thrombocytopenia with IMiD treatment. These data suggest that aromatase is a neosubstrate of cereblon that is responsible for IMiD-induced thrombocytopenia.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aromatasa/metabolismo , Factores Inmunológicos/efectos adversos , Trombocitopenia/inducido químicamente , Trombocitopenia/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Anciano , Anciano de 80 o más Años , Femenino , Células HEK293 , Humanos , Células K562 , Masculino , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Síndromes Mielodisplásicos/tratamiento farmacológico , Mielopoyesis/efectos de los fármacos , Mielopoyesis/fisiología , Especificidad por Sustrato , Trombocitopenia/patología
16.
Nat Commun ; 11(1): 155, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31919358

RESUMEN

Dysregulated hematopoiesis occurs in several chronic inflammatory diseases, but it remains unclear how hematopoietic stem cells (HSCs) in the bone marrow (BM) sense peripheral inflammation and contribute to tissue damage in arthritis. Here, we show the HSC gene expression program is biased toward myelopoiesis and differentiation skewed toward granulocyte-monocyte progenitors (GMP) during joint and intestinal inflammation in experimental spondyloarthritis (SpA). GM-CSF-receptor is increased on HSCs and multipotent progenitors, favoring a striking increase in myelopoiesis at the earliest hematopoietic stages. GMP accumulate in the BM in SpA and, unexpectedly, at extramedullary sites: in the inflamed joints and spleen. Furthermore, we show that GM-CSF promotes extramedullary myelopoiesis, tissue-toxic neutrophil accumulation in target organs, and GM-CSF prophylactic or therapeutic blockade substantially decreases SpA severity. Surprisingly, besides CD4+ T cells and innate lymphoid cells, mast cells are a source of GM-CSF in this model, and its pathogenic production is promoted by the alarmin IL-33.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Hematopoyesis Extramedular/fisiología , Células Madre Hematopoyéticas/metabolismo , Mielopoyesis/fisiología , Espondiloartritis/patología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Células Cultivadas , Femenino , Interleucina-33/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Espondiloartritis/inmunología
17.
Nat Rev Immunol ; 20(3): 196-202, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31740804

RESUMEN

Genetic defects that accumulate in haematopoietic stem cells (HSCs) are thought to be responsible for age-related changes in haematopoiesis that include a decline in lymphopoiesis and skewing towards the myeloid lineage. This HSC-centric view is based largely on studies showing that HSCs from aged mice exhibit these lineage biases following transplantation into irradiated young recipient mice. In this Opinion article, we make the case that the reliance on this approach has led to inaccurate conclusions regarding the effects of ageing on blood-forming stem cells; we suggest instead that changes in the environment contribute to haematopoietic system ageing. We propose that a complete understanding of how ageing affects haematopoiesis depends on the analysis of blood cell production in unperturbed mice. We describe how this can be achieved using in situ fate mapping. This approach indicates that changes in downstream progenitors, in addition to any HSC defects, may explain the reduced lymphopoiesis and sustained myelopoiesis that occur during ageing.


Asunto(s)
Envejecimiento/fisiología , Senescencia Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Hematopoyesis/fisiología , Humanos , Mielopoyesis/fisiología
18.
Nat Commun ; 10(1): 5046, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31695038

RESUMEN

Abdominal aortic aneurysm (AAA) is a prevalent life-threatening disease, where aortic wall degradation is mediated by accumulated immune cells. Although cytokines regulate inflammation within the aorta, their contribution to AAA via distant alterations, particularly in the control of hematopoietic stem cell (HSC) differentiation, remains poorly defined. Here we report a pathogenic role for the interleukin-27 receptor (IL-27R) in AAA, as genetic ablation of IL-27R protects mice from the disease development. Mitigation of AAA is associated with a blunted accumulation of myeloid cells in the aorta due to the attenuation of Angiotensin II (Ang II)-induced HSC expansion. IL-27R signaling is required to induce transcriptional programming to overcome HSC quiescence and increase differentiation and output of mature myeloid cells in response to stress stimuli to promote their accumulation in the diseased aorta. Overall, our studies illuminate how a prominent vascular disease can be distantly driven by a cytokine-dependent regulation of bone marrow precursors.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Interleucina-27/metabolismo , Mielopoyesis/fisiología , Receptores de Interleucina/metabolismo , Aneurisma/metabolismo , Angiotensina II/metabolismo , Animales , Aorta/patología , Aneurisma de la Aorta Abdominal/patología , Presión Sanguínea , Diferenciación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Noqueados para ApoE , Células Mieloides/patología , Receptores de Interleucina/genética , Transducción de Señal
19.
Immunity ; 51(2): 351-366.e6, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31303400

RESUMEN

Aging results in increased myelopoiesis, which is linked to the increased incidence of myeloid leukemias and production of myeloid-derived suppressor cells. Here, we examined the contribution of plasma cells (PCs) to age-related increases in myelopoiesis, as PCs exhibit immune regulatory function and sequester in bone marrow (BM). PC number was increased in old BM, and they exhibited high expression of genes encoding inflammatory cytokines and pathogen sensors. Antibody-mediated depletion of PCs from old mice reduced the number of myeloid-biased hematopoietic stem cells and mature myeloid cells to levels in young animals, but lymphopoiesis was not rejuvenated, indicating that redundant mechanisms inhibit that process. PCs also regulated the production of inflammatory factors from BM stromal cells, and disruption of the PC-stromal cell circuitry with inhibitors of the cytokines IL-1 and TNF-α attenuated myelopoiesis in old mice. Thus, the age-related increase in myelopoiesis is driven by an inflammatory network orchestrated by PCs.


Asunto(s)
Envejecimiento/fisiología , Médula Ósea/fisiología , Células Madre Hematopoyéticas/patología , Inflamación/metabolismo , Mielopoyesis/fisiología , Células Plasmáticas/fisiología , Animales , Células Cultivadas , Humanos , Interleucina-1/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/metabolismo
20.
Exp Cell Res ; 382(1): 111445, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31152707

RESUMEN

MicroRNAs (miRNA) are small noncoding RNAs that regulate gene expression by targeting mRNAs in a sequence specific manner, thereby determining their degradation or inhibiting translation. They are involved in processes such as proliferation, differentiation and apoptosis by fine-tuning the expression of genes underlying such events. The expression of specific miRNAs is involved in hematopoietic differentiation and their deregulation contributes to the development of hematopoietic malignancies such as acute myeloid leukemia (AML). miR-130a is over-expressed in AML. Here we show that miR-130a is physiologically expressed in myeloblasts and down-regulated during monocyte differentiation. Gain- and loss-of-function experiments performed on CD34+ human hematopoietic stem cells confirmed that expression of miR-130a inhibits monocyte differentiation by interfering with the expression of key transcription factors HOXA10, IRF8, KLF4, MAFB and PU-1. The data obtained in this study highlight that the correct modulation of miR-130a is necessary for normal differentiation to occur and confirming that deregulation of this miRNA might underlie the differentiation block occurring in AML.


Asunto(s)
Regulación de la Expresión Génica , Células Precursoras de Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , MicroARNs/fisiología , Monocitos/citología , Mielopoyesis/fisiología , Proteínas de Neoplasias/fisiología , Antígenos CD34/análisis , Línea Celular Tumoral , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Mutación con Ganancia de Función , Células Precursoras de Granulocitos/citología , Células Madre Hematopoyéticas/citología , Humanos , Factor 4 Similar a Kruppel , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación con Pérdida de Función , MicroARNs/antagonistas & inhibidores , MicroARNs/biosíntesis , MicroARNs/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Ácidos Nucleicos de Péptidos/farmacología , ARN Neoplásico/genética , ARN Neoplásico/fisiología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...