Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell ; 160(1-2): 241-52, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25594182

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) can reconstitute and sustain the entire blood system. We generated a highly specific transgenic reporter of HSPCs in zebrafish. This allowed us to perform high-resolution live imaging on endogenous HSPCs not currently possible in mammalian bone marrow. Using this system, we have uncovered distinct interactions between single HSPCs and their niche. When an HSPC arrives in the perivascular niche, a group of endothelial cells remodel to form a surrounding pocket. This structure appears conserved in mouse fetal liver. Correlative light and electron microscopy revealed that endothelial cells surround a single HSPC attached to a single mesenchymal stromal cell. Live imaging showed that mesenchymal stromal cells anchor HSPCs and orient their divisions. A chemical genetic screen found that the compound lycorine promotes HSPC-niche interactions during development and ultimately expands the stem cell pool into adulthood. Our studies provide evidence for dynamic niche interactions upon stem cell colonization. PAPERFLICK:


Asunto(s)
Endotelio/fisiología , Células Madre Hematopoyéticas/citología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , División Celular , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/fisiología , Endotelio/citología , Células Madre Hematopoyéticas/fisiología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Nicho de Células Madre , Células del Estroma/citología , Células del Estroma/metabolismo , Pez Cebra/fisiología
2.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34919128

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) are multipotent cells that self-renew or differentiate to establish the entire blood hierarchy. HSPCs arise from the hemogenic endothelium of the dorsal aorta (DA) during development in a process called endothelial-to-hematopoietic transition. The factors and signals that control HSPC fate decisions from the hemogenic endothelium are not fully understood. We found that Vegfc has a role in HSPC emergence from the zebrafish DA. Using time-lapse live imaging, we show that some HSPCs in the DA of vegfc loss-of-function embryos display altered cellular behavior. Instead of typical budding from the DA, emergent HSPCs exhibit crawling behavior similar to myeloid cells. This was confirmed by increased myeloid cell marker expression in the ventral wall of the DA and the caudal hematopoietic tissue. This increase in myeloid cells corresponded with a decrease in HSPCs that persisted into larval stages. Together, our data suggest that Vegfc regulates HSPC emergence in the hemogenic endothelium, in part by suppressing a myeloid cell fate. Our study provides a potential signal for modulation of HSPC fate in stem cell differentiation protocols.


Asunto(s)
Aorta/citología , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Aorta/embriología , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/embriología , Células Madre Hematopoyéticas/citología , Mutación con Pérdida de Función , Células Mieloides/citología , Células Mieloides/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Pez Cebra , Proteínas de Pez Cebra/genética
4.
Blood ; 137(6): 775-787, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32881992

RESUMEN

Hematopoietic and nervous systems are linked via innervation of bone marrow (BM) niche cells. Hematopoietic stem/progenitor cells (HSPCs) express neurotransmitter receptors, such as the γ-aminobutyric acid (GABA) type B receptor subunit 1 (GABBR1), suggesting that HSPCs could be directly regulated by neurotransmitters like GABA that directly bind to GABBR1. We performed imaging mass spectrometry and found that the endogenous GABA molecule is regionally localized and concentrated near the endosteum of the BM niche. To better understand the role of GABBR1 in regulating HSPCs, we generated a constitutive Gabbr1-knockout mouse model. Analysis revealed that HSPC numbers were significantly reduced in the BM compared with wild-type littermates. Moreover, Gabbr1-null hematopoietic stem cells had diminished capacity to reconstitute irradiated recipients in a competitive transplantation model. Gabbr1-null HSPCs were less proliferative under steady-state conditions and upon stress. Colony-forming unit assays demonstrated that almost all Gabbr1-null HSPCs were in a slow or noncycling state. In vitro differentiation of Gabbr1-null HSPCs in cocultures produced fewer overall cell numbers with significant defects in differentiation and expansion of the B-cell lineage. To determine whether a GABBR1 agonist could stimulate human umbilical cord blood (UCB) HSPCs, we performed brief ex vivo treatment prior to transplant into immunodeficient mice, with significant increases in long-term engraftment of HSPCs compared with GABBR1 antagonist or vehicle treatments. Our results indicate a direct role for GABBR1 in HSPC proliferation, and identify a potential target to improve HSPC engraftment in clinical transplantation.


Asunto(s)
Células Madre Hematopoyéticas/citología , Receptores de GABA-B/fisiología , Animales , Linfocitos B/patología , Baclofeno/análogos & derivados , Baclofeno/farmacología , Médula Ósea/inervación , Médula Ósea/metabolismo , Trasplante de Médula Ósea , División Celular , Linaje de la Célula , Femenino , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Linfopenia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Quimera por Radiación , Receptores de GABA-B/deficiencia , Receptores de GABA-B/genética , Nicho de Células Madre
5.
Nature ; 523(7561): 468-71, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26201599

RESUMEN

Haematopoietic stem and progenitor cell (HSPC) transplant is a widely used treatment for life-threatening conditions such as leukaemia; however, the molecular mechanisms regulating HSPC engraftment of the recipient niche remain incompletely understood. Here we develop a competitive HSPC transplant method in adult zebrafish, using in vivo imaging as a non-invasive readout. We use this system to conduct a chemical screen, and identify epoxyeicosatrienoic acids (EETs) as a family of lipids that enhance HSPC engraftment. The pro-haematopoietic effects of EETs were conserved in the developing zebrafish embryo, where 11,12-EET promoted HSPC specification by activating a unique activator protein 1 (AP-1) and runx1 transcription program autonomous to the haemogenic endothelium. This effect required the activation of the phosphatidylinositol-3-OH kinase (PI(3)K) pathway, specifically PI(3)Kγ. In adult HSPCs, 11,12-EET induced transcriptional programs, including AP-1 activation, which modulate several cellular processes, such as migration, to promote engraftment. Furthermore, we demonstrate that the EET effects on enhancing HSPC homing and engraftment are conserved in mammals. Our study establishes a new method to explore the molecular mechanisms of HSPC engraftment, and discovers a previously unrecognized, evolutionarily conserved pathway regulating multiple haematopoietic generation and regeneration processes. EETs may have clinical application in marrow or cord blood transplantation.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Pez Cebra/embriología , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Línea Celular , Movimiento Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Riñón/citología , Masculino , Ratones , Fosfatidilinositol 3-Quinasas , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
6.
Blood ; 138(21): 2012-2014, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34821941
8.
Blood ; 126(26): 2811-20, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26385351

RESUMEN

Rare endothelial cells in the aorta-gonad-mesonephros (AGM) transition into hematopoietic stem cells (HSCs) during embryonic development. Lineage tracing experiments indicate that HSCs emerge from cadherin 5 (Cdh5; vascular endothelial-cadherin)(+) endothelial precursors, and isolated populations of Cdh5(+) cells from mouse embryos and embryonic stem cells can be differentiated into hematopoietic cells. Cdh5 has also been widely implicated as a marker of AGM-derived hemogenic endothelial cells. Because Cdh5(-/-) mice embryos die before the first HSCs emerge, it is unknown whether Cdh5 has a direct role in HSC emergence. Our previous genetic screen yielded malbec (mlb(bw306)), a zebrafish mutant for cdh5, with normal embryonic and definitive blood. Using time-lapse confocal imaging, parabiotic surgical pairing of zebrafish embryos, and blastula transplantation assays, we show that HSCs emerge, migrate, engraft, and differentiate in the absence of cdh5 expression. By tracing Cdh5(-/-)green fluorescent protein (GFP)(+/+) cells in chimeric mice, we demonstrated that Cdh5(-/-)GFP(+/+) HSCs emerging from embryonic day 10.5 and 11.5 (E10.5 and E11.5) AGM or derived from E13.5 fetal liver not only differentiate into hematopoietic colonies but also engraft and reconstitute multilineage adult blood. We also developed a conditional mouse Cdh5 knockout (Cdh5(flox/flox):Scl-Cre-ER(T)) and demonstrated that multipotent hematopoietic colonies form despite the absence of Cdh5. These data establish that Cdh5, a marker of hemogenic endothelium in the AGM, is dispensable for the transition of hemogenic endothelium to HSCs.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Diferenciación Celular/fisiología , Hemangioblastos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Animales , Linaje de la Célula/fisiología , Electroporación , Embrión de Mamíferos , Embrión no Mamífero , Citometría de Flujo , Inmunohistoquímica , Mesonefro/embriología , Ratones , Ratones Noqueados , Microscopía Confocal , Pez Cebra
10.
Dev Biol ; 373(2): 431-41, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22960038

RESUMEN

Growth Factor Independence (Gfi) transcription factors play essential roles in hematopoiesis, differentially activating and repressing transcriptional programs required for hematopoietic stem/progenitor cell (HSPC) development and lineage specification. In mammals, Gfi1a regulates hematopoietic stem cells (HSC), myeloid and lymphoid populations, while its paralog, Gfi1b, regulates HSC, megakaryocyte and erythroid development. In zebrafish, gfi1aa is essential for primitive hematopoiesis; however, little is known about the role of gfi1aa in definitive hematopoiesis or about additional gfi factors in zebrafish. Here, we report the isolation and characterization of an additional hematopoietic gfi factor, gfi1b. We show that gfi1aa and gfi1b are expressed in the primitive and definitive sites of hematopoiesis in zebrafish. Our functional analyses demonstrate that gfi1aa and gfi1b have distinct roles in regulating primitive and definitive hematopoietic progenitors, respectively. Loss of gfi1aa silences markers of early primitive progenitors, scl and gata1. Conversely, loss of gfi1b silences runx-1, c-myb, ikaros and cd41, indicating that gfi1b is required for definitive hematopoiesis. We determine the epistatic relationships between the gfi factors and key hematopoietic transcription factors, demonstrating that gfi1aa and gfi1b join lmo2, scl, runx-1 and c-myb as critical regulators of teleost HSPC. Our studies establish a comparative paradigm for the regulation of hematopoietic lineages by gfi transcription factors.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Secuencia Conservada/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero/metabolismo , Epistasis Genética , Eritropoyesis/genética , Evolución Molecular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/embriología , Sistema Hematopoyético/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Pez Cebra/embriología , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo
11.
Development ; 138(1): 169-77, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21138979

RESUMEN

Molecular genetics approaches in zebrafish research are hampered by the lack of a ubiquitous transgene driver element that is active at all developmental stages. Here, we report the isolation and characterization of the zebrafish ubiquitin (ubi) promoter, which drives constitutive transgene expression during all developmental stages and analyzed adult organs. Notably, ubi expresses in all blood cell lineages, and we demonstrate the application of ubi-driven fluorophore transgenics in hematopoietic transplantation experiments to assess true multilineage potential of engrafted cells. We further generated transgenic zebrafish that express ubiquitous 4-hydroxytamoxifen-controlled Cre recombinase activity from a ubi:cre(ERt2) transgene, as well as ubi:loxP-EGFP-loxP-mCherry (ubi:Switch) transgenics and show their use as a constitutive fluorescent lineage tracing reagent. The ubi promoter and the transgenic lines presented here thus provide a broad resource and important advancement for transgenic applications in zebrafish.


Asunto(s)
Integrasas/metabolismo , Regiones Promotoras Genéticas/genética , Transgenes/genética , Ubiquitina/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Activación Enzimática/efectos de los fármacos , Integrasas/genética , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología
12.
Methods Mol Biol ; 2567: 233-249, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36255705

RESUMEN

The zebrafish as a model organism is well known for its versatile genetics, rapid development, and straightforward live imaging. It is an excellent model to study hematopoiesis because of its highly conserved ontogeny and gene regulatory networks. Recently developed highly specific transgenic reporter lines have allowed direct imaging and tracking of hematopoietic stem and progenitor cells (HSPCs) in live zebrafish. These reporter lines can also be used for fluorescence-activated cell sorting (FACS) of HSPCs. Similar to mammalian models, HSPCs can be transplanted to reconstitute the entire hematopoietic system of zebrafish recipients. However, the zebrafish provides unique advantages to study HSPC biology, such as transplants into embryos and high-throughput chemical screening. This chapter will outline the methods needed to identify, isolate, and transplant HSPCs in zebrafish.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales Modificados Genéticamente , Mamíferos/metabolismo
13.
Dev Biol ; 360(2): 415-25, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22008794

RESUMEN

The node and notochord are key tissues required for patterning of the vertebrate body plan. Understanding the gene regulatory network that drives their formation and function is therefore important. Foxa2 is a key transcription factor at the top of this genetic hierarchy and finding its targets will help us to better understand node and notochord development. We performed an extensive microarray-based gene expression screen using sorted embryonic notochord cells to identify early notochord-enriched genes. We validated their specificity to the node and notochord by whole mount in situ hybridization. This provides the largest available resource of notochord-expressed genes, and therefore candidate Foxa2 target genes in the notochord. Using existing Foxa2 ChIP-seq data from adult liver, we were able to identify a set of genes expressed in the notochord that had associated regions of Foxa2-bound chromatin. Given that Foxa2 is a pioneer transcription factor, we reasoned that these sites might represent notochord-specific enhancers. Candidate Foxa2-bound regions were tested for notochord specific enhancer function in a zebrafish reporter assay and 7 novel notochord enhancers were identified. Importantly, sequence conservation or predictive models could not have readily identified these regions. Mutation of putative Foxa2 binding elements in two of these novel enhancers abrogated reporter expression and confirmed their Foxa2 dependence. The combination of highly specific gene expression profiling and genome-wide ChIP analysis is a powerful means of understanding developmental pathways, even for small cell populations such as the notochord.


Asunto(s)
Cromatina/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Notocorda/embriología , Animales , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Mutación , Notocorda/metabolismo , Análisis por Matrices de Proteínas , Pez Cebra
14.
Dev Cell ; 13(6): 884-96, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18061569

RESUMEN

The node and notochord have been extensively studied as signaling centers in the vertebrate embryo. The morphogenesis of these tissues, particularly in mouse, is not well understood. Using time-lapse live imaging and cell lineage tracking, we show the notochord has distinct morphogenetic origins along the anterior-posterior axis. The anterior head process notochord arises independently of the node by condensation of dispersed cells. The trunk notochord is derived from the node and forms by convergent extension. The tail notochord forms by node-derived progenitors that actively migrate toward the posterior. We also reveal distinct genetic regulation within these different regions. We show that Foxa2 compensates for and genetically interacts with Noto in the trunk notochord, and that Noto has an evolutionarily conserved role in regulating axial versus paraxial cell fate. Therefore, we propose three distinct regions within the mouse notochord, each with unique morphogenetic origins.


Asunto(s)
Factor Nuclear 3-beta del Hepatocito/fisiología , Proteínas de Homeodominio/fisiología , Morfogénesis , Notocorda/fisiología , Animales , Linaje de la Célula , Femenino , Gástrula , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas de Homeodominio/genética , Procesamiento de Imagen Asistido por Computador , Técnicas para Inmunoenzimas , Hibridación in Situ , Ratones/embriología , Ratones Endogámicos ICR , Ratones Transgénicos , Notocorda/embriología , Embarazo , Transgenes/fisiología
15.
Elife ; 112022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35943143

RESUMEN

The blood system is supported by hematopoietic stem and progenitor cells (HSPCs) found in a specialized microenvironment called the niche. Many different niche cell types support HSPCs, however how they interact and their ultrastructure has been difficult to define. Here, we show that single endogenous HSPCs can be tracked by light microscopy, then identified by serial block-face scanning electron microscopy (SBEM) at multiscale levels. Using the zebrafish larval kidney marrow (KM) niche as a model, we followed single fluorescently labeled HSPCs by light sheet microscopy, then confirmed their exact location in a 3D SBEM dataset. We found a variety of different configurations of HSPCs and surrounding niche cells, suggesting there could be functional heterogeneity in sites of HSPC lodgement. Our approach also allowed us to identify dopamine beta-hydroxylase (dbh) positive ganglion cells as a previously uncharacterized functional cell type in the HSPC niche. By integrating multiple imaging modalities, we could resolve the ultrastructure of single rare cells deep in live tissue and define all contacts between an HSPC and its surrounding niche cell types.


Asunto(s)
Nicho de Células Madre , Pez Cebra , Animales , Células Madre Hematopoyéticas/metabolismo , Microscopía Electrónica
16.
BMC Genomics ; 9: 511, 2008 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-18973680

RESUMEN

BACKGROUND: The Spemann/Mangold organizer is a transient tissue critical for patterning the gastrula stage vertebrate embryo and formation of the three germ layers. Despite its important role during development, there are still relatively few genes with specific expression in the organizer and its derivatives. Foxa2 is a forkhead transcription factor that is absolutely required for formation of the mammalian equivalent of the organizer, the node, the axial mesoderm and the definitive endoderm (DE). However, the targets of Foxa2 during embryogenesis, and the molecular impact of organizer loss on the gastrula embryo, have not been well defined. RESULTS: To identify genes specific to the Spemann/Mangold organizer, we performed a microarray-based screen that compared wild-type and Foxa2 mutant embryos at late gastrulation stage (E7.5). We could detect genes that were consistently down-regulated in replicate pools of mutant embryos versus wild-type, and these included a number of known node and DE markers. We selected 314 genes without previously published data at E7.5 and screened for expression by whole mount in situ hybridization. We identified 10 novel expression patterns in the node and 5 in the definitive endoderm. We also found significant reduction of markers expressed in secondary tissues that require interaction with the organizer and its derivatives, such as cardiac mesoderm, vasculature, primitive streak, and anterior neuroectoderm. CONCLUSION: The genes identified in this screen represent novel Spemann/Mangold organizer genes as well as potential Foxa2 targets. Further investigation will be needed to define these genes as novel developmental regulatory factors involved in organizer formation and function. We have placed these genes in a Foxa2-dependent genetic regulatory network and we hypothesize how Foxa2 may regulate a molecular program of Spemann/Mangold organizer development. We have also shown how early loss of the organizer and its inductive properties in an otherwise normal embryo, impacts on the molecular profile of surrounding tissues.


Asunto(s)
Inducción Embrionaria , Gástrula , Regulación de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/fisiología , Organizadores Embrionarios/fisiología , Animales , Embrión de Mamíferos , Desarrollo Embrionario , Redes Reguladoras de Genes , Hibridación in Situ , Ratones , Análisis por Micromatrices , Mutación
18.
Nucleic Acids Res ; 34(17): 4780-90, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16971461

RESUMEN

Understanding transcriptional regulation in early developmental stages is fundamental to understanding mammalian development and embryonic stem (ES) cell properties. Expression surveys suggest that the putative SCAN-Zinc finger transcription factor Zfp206 is expressed specifically in ES cells [Zhang,W., Morris,Q.D., Chang,R., Shai,O., Bakowski,M.A., Mitsakakis,N., Mohammad,N., Robinson,M.D., Zirngibl,R., Somogyi,E. et al., (2004) J. Biol., 3, 21; Brandenberger,R., Wei,H., Zhang,S., Lei,S., Murage,J., Fisk,G.J., Li,Y., Xu,C., Fang,R., Guegler,K. et al., (2004) Nat. Biotechnol., 22, 707-716]. Here, we confirm this observation, and we show that ZFP206 expression decreases rapidly upon differentiation of cultured mouse ES cells, and during development of mouse embryos. We find that there are at least six isoforms of the ZFP206 transcript, the longest being predominant. Overexpression and depletion experiments show that Zfp206 promotes formation of undifferentiated ES cell clones, and positively regulates abundance of a very small set of transcripts whose expression is also specific to ES cells and the two- to four-cell stages of preimplantation embryos. This set includes members of the Zscan4, Thoc4, Tcstv1 and eIF-1A gene families, none of which have been functionally characterized in vivo but whose members include apparent transcription factors, RNA-binding proteins and translation factors. Together, these data indicate that Zfp206 is a regulator of ES cell differentiation that controls a set of genes expressed very early in development, most of which themselves appear to be regulators.


Asunto(s)
Embrión de Mamíferos/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Blastocisto/metabolismo , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Ratones , Isoformas de Proteínas/genética , ARN Mensajero/metabolismo , Células Madre/química , Células Madre/citología , Factores de Transcripción/análisis , Factores de Transcripción/genética
19.
Cell Stem Cell ; 22(1): 5-7, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29304342

RESUMEN

The bone marrow (BM) niche is a complex microenvironment that supports healthy hematopoietic stem cells (HSCs) throughout life. In this issue of Cell Stem Cell, Duarte et al. (2018) reveal the spatio-temporal progress of leukemic cells as they invade and occupy the niche, ultimately outcompeting native HSCs.


Asunto(s)
Médula Ósea , Nicho de Células Madre , Células Madre Hematopoyéticas , Humanos , Leucemia , Masticación
20.
Trends Cell Biol ; 28(12): 987-998, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29857963

RESUMEN

The hematopoietic stem cell (HSC) niche supports steady-state hematopoiesis and responds to changing needs during stress and disease. The nervous system is an important regulator of the niche, and its influence is established early in development when stem cells are specified. Most research has focused on direct innervation of the niche, however recent findings show there are different modes of neural control, including globally by the central nervous system (CNS) and hormone release, locally by neural crest-derived mesenchymal stem cells, and intrinsically by hematopoietic cells that express neural receptors and neurotransmitters. Dysregulation between neural and hematopoietic systems can contribute to disease, however new therapeutic opportunities may be found among neuroregulator drugs repurposed to support hematopoiesis.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Nicho de Células Madre , Animales , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Neurotransmisores/farmacología , Nicho de Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA