Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
bioRxiv ; 2023 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-37693628

RESUMEN

Tropomyosins coat actin filaments and impact actin-related signaling and cell morphogenesis. Genome-wide association studies have linked Tropomyosin 1 (TPM1) with human blood trait variation. Prior work suggested that TPM1 regulated blood cell formation in vitro, but it was unclear how or when TPM1 affected hematopoiesis. Using gene-edited induced pluripotent stem cell (iPSC) model systems, TPM1 knockout was found to augment developmental cell state transitions, as well as TNFα and GTPase signaling pathways, to promote hemogenic endothelial (HE) cell specification and hematopoietic progenitor cell (HPC) production. Single-cell analyses showed decreased TPM1 expression during human HE specification, suggesting that TPM1 regulated in vivo hematopoiesis via similar mechanisms. Indeed, analyses of a TPM1 gene trap mouse model showed that TPM1 deficiency enhanced the formation of HE during embryogenesis. These findings illuminate novel effects of TPM1 on developmental hematopoiesis.

2.
Dev Cell ; 57(23): 2652-2660.e3, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36473461

RESUMEN

Placental fetal macrophages (fMacs) are the only immune cells on the fetal side of the placental barrier. Mouse models have not been used to test their function because they have previously been found to have distinct cellular origins and functions in mice and humans. Here, we test the ontogeny of mouse placental fMacs. Using a new Hoxa13Cre allele that labels all placental endothelial cells (ECs), we demonstrate that mouse placenta fMacs do not arise from placental endothelium. Instead, lineage tracing studies using Tie2-Cre and Cx3cr1CreERT2 alleles demonstrate that mouse placental fMacs arise from yolk sac endothelium. Administration of blocking antibodies against CSF1R at E6.5 and E7.5 results in depletion of placental fMacs throughout pregnancy, and this suggests a yolk sac origin, similar to that in human fMacs. This Matters Arising paper is in response to Liang et al., published in Developmental Cell. A response by Liang and Liu is published in this issue.


Asunto(s)
Células Endoteliales , Placenta , Embarazo , Femenino , Animales , Humanos , Ratones
3.
Blood ; 139(19): 2942-2957, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35245372

RESUMEN

The hematopoietic stem cells (HSCs) that produce blood for the lifetime of an animal arise from RUNX1+ hemogenic endothelial cells (HECs) in the embryonic vasculature through a process of endothelial-to-hematopoietic transition (EHT). Studies have identified inflammatory mediators and fluid shear forces as critical environmental stimuli for EHT, raising the question of how such diverse inputs are integrated to drive HEC specification. Endothelial cell MEKK3-KLF2/4 signaling can be activated by both fluid shear forces and inflammatory mediators, and it plays roles in cardiovascular development and disease that have been linked to both stimuli. Here we demonstrate that MEKK3 and KLF2/4 are required in endothelial cells for the specification of RUNX1+ HECs in both the yolk sac and dorsal aorta of the mouse embryo and for their transition to intraaortic hematopoietic cluster (IAHC) cells. The inflammatory mediators lipopolysaccharide and interferon-γ increase RUNX1+ HECs in an MEKK3-dependent manner. Maternal administration of catecholamines that stimulate embryo cardiac function and accelerate yolk sac vascular remodeling increases EHT by wild-type but not MEKK3-deficient endothelium. These findings identify MEKK-KLF2/4 signaling as an essential pathway for EHT and provide a molecular basis for the integration of diverse environmental inputs, such as inflammatory mediators and hemodynamic forces, during definitive hematopoiesis.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Hemangioblastos , Hematopoyesis , Animales , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Endotelio/metabolismo , Hemangioblastos/citología , Hemangioblastos/metabolismo , Hemodinámica , Mediadores de Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , MAP Quinasa Quinasa Quinasa 3/metabolismo , Ratones
4.
Proc Natl Acad Sci U S A ; 117(38): 23626-23635, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32883883

RESUMEN

Hematopoietic stem and progenitor cell (HSPC) formation and lineage differentiation involve gene expression programs orchestrated by transcription factors and epigenetic regulators. Genetic disruption of the chromatin remodeler chromodomain-helicase-DNA-binding protein 7 (CHD7) expanded phenotypic HSPCs, erythroid, and myeloid lineages in zebrafish and mouse embryos. CHD7 acts to suppress hematopoietic differentiation. Binding motifs for RUNX and other hematopoietic transcription factors are enriched at sites occupied by CHD7, and decreased RUNX1 occupancy correlated with loss of CHD7 localization. CHD7 physically interacts with RUNX1 and suppresses RUNX1-induced expansion of HSPCs during development through modulation of RUNX1 activity. Consequently, the RUNX1:CHD7 axis provides proper timing and function of HSPCs as they emerge during hematopoietic development or mature in adults, representing a distinct and evolutionarily conserved control mechanism to ensure accurate hematopoietic lineage differentiation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Proteínas de Unión al ADN , Hematopoyesis , Animales , Diferenciación Celular , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Células Madre Hematopoyéticas , Humanos , Masculino , Ratones , Bazo/citología , Pez Cebra
5.
Genes Dev ; 34(13-14): 950-964, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32499402

RESUMEN

Hematopoietic stem cell (HSC) ontogeny is accompanied by dynamic changes in gene regulatory networks. We performed RNA-seq and histone mark ChIP-seq to define the transcriptomes and epigenomes of cells representing key developmental stages of HSC ontogeny in mice. The five populations analyzed were embryonic day 10.5 (E10.5) endothelium and hemogenic endothelium from the major arteries, an enriched population of prehematopoietic stem cells (pre-HSCs), fetal liver HSCs, and adult bone marrow HSCs. Using epigenetic signatures, we identified enhancers for each developmental stage. Only 12% of enhancers are primed, and 78% are active, suggesting the vast majority of enhancers are established de novo without prior priming in earlier stages. We constructed developmental stage-specific transcriptional regulatory networks by linking enhancers and predicted bound transcription factors to their target promoters using a novel computational algorithm, target inference via physical connection (TIPC). TIPC predicted known transcriptional regulators for the endothelial-to-hematopoietic transition, validating our overall approach, and identified putative novel transcription factors, including the broadly expressed transcription factors SP3 and MAZ. Finally, we validated a role for SP3 and MAZ in the formation of hemogenic endothelium. Our data and computational analyses provide a useful resource for uncovering regulators of HSC formation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Algoritmos , Animales , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Edición Génica , Ratones , Factor de Transcripción Sp3/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma
6.
Blood ; 136(7): 845-856, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32392346

RESUMEN

Hematopoietic stem and progenitor cells (HSPCs) in the bone marrow are derived from a small population of hemogenic endothelial (HE) cells located in the major arteries of the mammalian embryo. HE cells undergo an endothelial to hematopoietic cell transition, giving rise to HSPCs that accumulate in intra-arterial clusters (IAC) before colonizing the fetal liver. To examine the cell and molecular transitions between endothelial (E), HE, and IAC cells, and the heterogeneity of HSPCs within IACs, we profiled ∼40 000 cells from the caudal arteries (dorsal aorta, umbilical, vitelline) of 9.5 days post coitus (dpc) to 11.5 dpc mouse embryos by single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing. We identified a continuous developmental trajectory from E to HE to IAC cells, with identifiable intermediate stages. The intermediate stage most proximal to HE, which we term pre-HE, is characterized by increased accessibility of chromatin enriched for SOX, FOX, GATA, and SMAD motifs. A developmental bottleneck separates pre-HE from HE, with RUNX1 dosage regulating the efficiency of the pre-HE to HE transition. A distal candidate Runx1 enhancer exhibits high chromatin accessibility specifically in pre-HE cells at the bottleneck, but loses accessibility thereafter. Distinct developmental trajectories within IAC cells result in 2 populations of CD45+ HSPCs; an initial wave of lymphomyeloid-biased progenitors, followed by precursors of hematopoietic stem cells (pre-HSCs). This multiomics single-cell atlas significantly expands our understanding of pre-HSC ontogeny.


Asunto(s)
Diferenciación Celular , Endotelio/embriología , Hemangioblastos/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Animales , Diferenciación Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Embrión de Mamíferos , Endotelio/citología , Endotelio/metabolismo , Femenino , Dosificación de Gen/fisiología , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos/citología , Hematopoyesis/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , RNA-Seq/métodos
7.
Cell Rep ; 29(12): 4200-4211.e7, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31851943

RESUMEN

Fetal hematopoietic stem cells (HSCs) undergo a developmental switch to become adult HSCs with distinct functional properties. To better understand the molecular mechanisms underlying the developmental switch, we have conducted deep sequencing of the 3D genome, epigenome, and transcriptome of fetal and adult HSCs in mouse. We find that chromosomal compartments and topologically associating domains (TADs) are largely conserved between fetal and adult HSCs. However, there is a global trend of increased compartmentalization and TAD boundary strength in adult HSCs. In contrast, intra-TAD chromatin interactions are much more dynamic and widespread, involving over a thousand gene promoters and distal enhancers. These developmental-stage-specific enhancer-promoter interactions are mediated by different sets of transcription factors, such as TCF3 and MAFB in fetal HSCs, versus NR4A1 and GATA3 in adult HSCs. Loss-of-function studies of TCF3 confirm the role of TCF3 in mediating condition-specific enhancer-promoter interactions and gene regulation in fetal HSCs.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Elementos de Facilitación Genéticos/genética , Femenino , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Masculino , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Regiones Promotoras Genéticas/genética
8.
Exp Hematol ; 68: 2-9, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30391350

RESUMEN

The transcription factor RUNX1 is required in the embryo for formation of the adult hematopoietic system. Here, we describe the seminal findings that led to the discovery of RUNX1 and of its critical role in blood cell formation in the embryo from hemogenic endothelium (HE). We also present RNA-sequencing data demonstrating that HE cells in different anatomic sites, which produce hematopoietic progenitors with dissimilar differentiation potentials, are molecularly distinct. Hemogenic and non-HE cells in the yolk sac are more closely related to each other than either is to hemogenic or non-HE cells in the major arteries. Therefore, a major driver of the different lineage potentials of the committed erythro-myeloid progenitors that emerge in the yolk sac versus hematopoietic stem cells that originate in the major arteries is likely to be the distinct molecular properties of the HE cells from which they are derived. We used bioinformatics analyses to predict signaling pathways active in arterial HE, which include the functionally validated pathways Notch, Wnt, and Hedgehog. We also used a novel bioinformatics approach to assemble transcriptional regulatory networks and predict transcription factors that may be specifically involved in hematopoietic cell formation from arterial HE, which is the origin of the adult hematopoietic system.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Hemangioblastos/fisiología , Hematopoyesis/fisiología , Animales , Arterias/citología , Arterias/embriología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/deficiencia , Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/fisiología , Proteínas de Drosophila/genética , Sangre Fetal/fisiología , Regulación del Desarrollo de la Expresión Génica , Humanos , Leucemia Experimental/genética , Leucemia Experimental/virología , Leucemia Mieloide Aguda/genética , Ratones , Ratones Noqueados , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/fisiología , Transcripción Genética , Saco Vitelino/citología
9.
J Exp Med ; 215(2): 645-659, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29282253

RESUMEN

Hematopoietic stem cells (HSCs) mature from pre-HSCs that originate in the major arteries of the embryo. To identify HSCs from in vitro sources, it will be necessary to refine markers of HSCs matured ex vivo. We purified and compared the transcriptomes of pre-HSCs, HSCs matured ex vivo, and fetal liver HSCs. We found that HSC maturation in vivo or ex vivo is accompanied by the down-regulation of genes involved in embryonic development and vasculogenesis, and up-regulation of genes involved in hematopoietic organ development, lymphoid development, and immune responses. Ex vivo matured HSCs more closely resemble fetal liver HSCs than pre-HSCs, but are not their molecular equivalents. We show that ex vivo-matured and fetal liver HSCs express programmed death ligand 1 (PD-L1). PD-L1 does not mark all pre-HSCs, but cell surface PD-L1 was present on HSCs matured ex vivo. PD-L1 signaling is not required for engraftment of embryonic HSCs. Hence, up-regulation of PD-L1 is a correlate of, but not a requirement for, HSC maturation.


Asunto(s)
Antígeno B7-H1/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Antígeno B7-H1/deficiencia , Antígeno B7-H1/genética , Diferenciación Celular , Femenino , Células Madre Fetales/citología , Células Madre Fetales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Hígado/citología , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Embarazo , Regulación hacia Arriba
10.
Nat Cell Biol ; 17(5): 580-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25915127

RESUMEN

The generation of haematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) will depend on the accurate recapitulation of embryonic haematopoiesis. In the early embryo, HSCs develop from the haemogenic endothelium (HE) and are specified in a Notch-dependent manner through a process named endothelial-to-haematopoietic transition (EHT). As HE is associated with arteries, it is assumed that it represents a subpopulation of arterial vascular endothelium (VE). Here we demonstrate at a clonal level that hPSC-derived HE and VE represent separate lineages. HE is restricted to the CD34(+)CD73(-)CD184(-) fraction of day 8 embryoid bodies and it undergoes a NOTCH-dependent EHT to generate RUNX1C(+) cells with multilineage potential. Arterial and venous VE progenitors, in contrast, segregate to the CD34(+)CD73(med)CD184(+) and CD34(+)CD73(hi)CD184(-) fractions, respectively. Together, these findings identify HE as distinct from VE and provide a platform for defining the signalling pathways that regulate their specification to functional HSCs.


Asunto(s)
Arterias/fisiología , Diferenciación Celular , Linaje de la Célula , Células Progenitoras Endoteliales/fisiología , Células Madre Hematopoyéticas/fisiología , Células Madre Multipotentes/fisiología , Células Madre Pluripotentes/fisiología , 5'-Nucleotidasa/deficiencia , Antígenos CD34/metabolismo , Arterias/citología , Arterias/metabolismo , Biomarcadores/metabolismo , Línea Celular , Separación Celular/métodos , Técnicas de Cocultivo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Progenitoras Endoteliales/metabolismo , Proteínas Ligadas a GPI/deficiencia , Células Madre Hematopoyéticas/metabolismo , Humanos , Microscopía por Video , Células Madre Multipotentes/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Células Precursoras de Linfocitos T/fisiología , Receptores CXCR5/deficiencia , Receptores Notch/metabolismo , Transducción de Señal , Factores de Tiempo , Venas/citología , Venas/fisiología
11.
Genes Dev ; 28(23): 2597-612, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25395663

RESUMEN

Identifying signaling pathways that regulate hematopoietic stem and progenitor cell (HSPC) formation in the embryo will guide efforts to produce and expand HSPCs ex vivo. Here we show that sterile tonic inflammatory signaling regulates embryonic HSPC formation. Expression profiling of progenitors with lymphoid potential and hematopoietic stem cells (HSCs) from aorta/gonad/mesonephros (AGM) regions of midgestation mouse embryos revealed a robust innate immune/inflammatory signature. Mouse embryos lacking interferon γ (IFN-γ) or IFN-α signaling and zebrafish morphants lacking IFN-γ and IFN-ϕ activity had significantly fewer AGM HSPCs. Conversely, knockdown of IFN regulatory factor 2 (IRF2), a negative regulator of IFN signaling, increased expression of IFN target genes and HSPC production in zebrafish. Chromatin immunoprecipitation (ChIP) combined with sequencing (ChIP-seq) and expression analyses demonstrated that IRF2-occupied genes identified in human fetal liver CD34(+) HSPCs are actively transcribed in human and mouse HSPCs. Furthermore, we demonstrate that the primitive myeloid population contributes to the local inflammatory response to impact the scale of HSPC production in the AGM region. Thus, sterile inflammatory signaling is an evolutionarily conserved pathway regulating the production of HSPCs during embryonic development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/citología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Transducción de Señal , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Proliferación Celular/genética , Células Cultivadas , Citocinas/inmunología , Embrión de Mamíferos , Embrión no Mamífero , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/genética , Inflamación/inmunología , Interferones/genética , Interferones/metabolismo , Ratones , Pez Cebra/embriología
12.
Development ; 140(18): 3765-76, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23924635

RESUMEN

The transcription factor Runx1 is essential for the formation of yolk sac-derived erythroid/myeloid progenitors (EMPs) and hematopoietic stem cells (HSCs) from hemogenic endothelium during embryogenesis. However, long-term repopulating HSCs (LT-HSCs) persist when Runx1 is conditionally deleted in fetal liver cells, demonstrating that the requirement for Runx1 changes over time. To define more precisely when Runx1 transitions from an essential factor to a homeostatic regulator of EMPs and HSCs, and whether that transition requires fetal liver colonization, we performed conditional, timed deletions of Runx1 between E7.5 and E13.5. We determined that Runx1 loss reduces the formation or function of EMPs up through E10.5. The Runx1 requirement in HSCs ends later, as deletion up to E11.5 eliminates HSCs. At E11.5, there is an abrupt transition to Runx1 independence in at least a subset of HSCs that does not require fetal liver colonization. The transition to Runx1 independence in EMPs is not mediated by other core binding factors (Runx2 and/or Runx3); however, deleting the common non-DNA-binding ß subunit (CBFß) severely compromises LT-HSC function. Hence, the requirements for Runx1 in EMP and HSC formation are temporally distinct, and LT-HSC function is highly reliant on continued core binding factor activity.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Animales , Aorta/citología , Aorta/embriología , Diferenciación Celular , Pollos , Ensayo de Unidades Formadoras de Colonias , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Endotelio/embriología , Endotelio/metabolismo , Epigénesis Genética , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Feto/embriología , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Integrasas/metabolismo , Hígado/citología , Hígado/embriología , Ratones , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Factores de Tiempo , Saco Vitelino/citología
13.
Proc Natl Acad Sci U S A ; 109(43): 17573-8, 2012 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-23045704

RESUMEN

Patients with Down syndrome (trisomy 21, T21) have hematologic abnormalities throughout life. Newborns frequently exhibit abnormal blood counts and a clonal preleukemia. Human T21 fetal livers contain expanded erythro-megakaryocytic precursors with enhanced proliferative capacity. The impact of T21 on the earliest stages of embryonic hematopoiesis is unknown and nearly impossible to examine in human subjects. We modeled T21 yolk sac hematopoiesis using human induced pluripotent stem cells (iPSCs). Blood progenitor populations generated from T21 iPSCs were present at normal frequency and proliferated normally. However, their developmental potential was altered with enhanced erythropoiesis and reduced myelopoiesis, but normal megakaryocyte production. These abnormalities overlap with those of T21 fetal livers, but also reflect important differences. Our studies show that T21 confers distinct developmental stage- and species-specific hematopoietic defects. More generally, we illustrate how iPSCs can provide insight into early stages of normal and pathological human development.


Asunto(s)
Síndrome de Down , Hematopoyesis/genética , Células Madre Pluripotentes/citología , Diferenciación Celular , Perfilación de la Expresión Génica , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Blood ; 112(10): 4051-60, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18755987

RESUMEN

Thrombocytopenia is a critical problem that occurs in many hematologic diseases, as well as after cancer therapy and radiation exposure. Platelet transfusion is the most commonly used therapy but has limitations of alloimmunization, availability, and expense. Thus, the development of safe, small, molecules to enhance platelet production would be advantageous for the treatment of thrombocytopenia. Herein, we report that an important lipid mediator and a peroxisome proliferator-activated receptor gamma (PPARgamma) ligand called 15-deoxy-Delta(12,14) prostaglandin J(2) (15d-PGJ(2)), increases Meg-01 maturation and platelet production. 15d-PGJ(2) also promotes platelet formation from culture-derived mouse and human megakaryocytes and accelerates platelet recovery after in vivo radiation-induced bone marrow injury. Interestingly, the platelet-enhancing effects of 15d-PGJ(2) in Meg-01 cells are independent of PPARgamma, but dependent on reactive oxygen species (ROS) accumulation; treatment with antioxidants such as glutathione ethyl ester (GSH-EE); or N-acetylcysteine (NAC) attenuate 15d-PGJ(2)-induced platelet production. Collectively, these data support the concept that megakaryocyte redox status plays an important role in platelet generation and that small electrophilic molecules may have clinical efficacy for improving platelet numbers in thrombocytopenic patients.


Asunto(s)
Antineoplásicos/farmacología , Plaquetas/metabolismo , Megacariocitos/metabolismo , Prostaglandina D2/análogos & derivados , Trombocitopenia/metabolismo , Acetilcisteína/farmacología , Animales , Antineoplásicos/uso terapéutico , Antioxidantes/farmacología , Médula Ósea/metabolismo , Línea Celular , Femenino , Glutatión/análogos & derivados , Glutatión/farmacología , Humanos , Masculino , Ratones , PPAR gamma/metabolismo , Transfusión de Plaquetas , Prostaglandina D2/farmacología , Prostaglandina D2/uso terapéutico , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/terapia , Especies Reactivas de Oxígeno/metabolismo , Trombocitopenia/terapia
15.
Blood ; 111(5): 2636-9, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18174377

RESUMEN

Hematopoiesis initiates within the yolk sac of mammalian embryos in overlapping primitive and definitive waves, each containing erythroid and megakaryocyte progenitors. c-myb-null mouse fetuses lack definitive erythrocytes but contain primitive erythroblasts and hepatic megakaryocytes. However, it is unclear if c-myb-null embryos harbor definitive erythroid or any megakaryocyte progenitors. We determined that c-myb was not expressed in primitive erythroid precursors and that c-myb-null embryos had normal primitive erythroid and megakaryocyte progenitor numbers and kinetics between embryonic day (E) 7.0 and E9.0. While primitive hematopoiesis is c-myb-independent, no definitive erythroid potential was detected in c-myb-null embryos, confirming that definitive erythropoiesis, beginning at E8.25 in the yolk sac, is completely c-myb-dependent. In contrast, reduced numbers of megakaryocyte progenitors with restricted proliferative capacity persist in E10.5 yolk sac and E11.5 liver. Despite this impaired megakaryocyte potential, c-myb-null fetuses had normal platelet numbers at E12.5 but became thrombocytopenic by E15.5, suggesting that c-myb is required for sustained thrombopoiesis.


Asunto(s)
Eritropoyesis , Megacariocitos/citología , Saco Vitelino/citología , Animales , Recuento de Células , Linaje de la Célula , Embrión de Mamíferos/citología , Células Eritroides/citología , Heterocigoto , Ratones , Proteínas Proto-Oncogénicas c-myb/deficiencia , Proteínas Proto-Oncogénicas c-myb/metabolismo , Trombopoyesis
16.
Blood ; 109(4): 1433-41, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17062726

RESUMEN

In the adult, platelets are derived from unipotential megakaryocyte colony-forming cells (Meg-CFCs) that arise from bipotential megakaryocyte/erythroid progenitors (MEPs). To better define the developmental origin of the megakaryocyte lineage, several aspects of megakaryopoiesis, including progenitors, maturing megakaryocytes, and circulating platelets, were examined in the murine embryo. We found that a majority of hemangioblast precursors during early gastrulation contains megakaryocyte potential. Combining progenitor assays with immunohistochemical analysis, we identified 2 waves of MEPs in the yolk sac associated with the primitive and definitive erythroid lineages. Primitive MEPs emerge at E7.25 along with megakaryocyte and primitive erythroid progenitors, indicating that primitive hematopoiesis is bilineage in nature. Subsequently, definitive MEPs expand in the yolk sac with Meg-CFCs and definitive erythroid progenitors. The first GP1bbeta-positive cells in the conceptus were identified in the yolk sac at E9.5, while large, highly reticulated platelets were detected in the embryonic bloodstream beginning at E10.5. At this time, the number of megakaryocyte progenitors begins to decline in the yolk sac and expand in the fetal liver. We conclude that the megakaryocyte lineage initially originates from hemangioblast precursors during early gastrulation and is closely associated both with primitive and with definitive erythroid lineages in the yolk sac prior to the transition of hematopoiesis to intraembryonic sites.


Asunto(s)
Linaje de la Célula/fisiología , Gástrula/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Megacariocitos/citología , Animales , Plaquetas/citología , Células Precursoras Eritroides/citología , Ratones , Ratones Endogámicos ICR , Células Madre Multipotentes/citología , Saco Vitelino/irrigación sanguínea
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA