Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38370741

RESUMEN

The limited proliferative capacity of erythroid precursors is a major obstacle to generate sufficient numbers of in vitro-derived red blood cells (RBC) for clinical purposes. We and others have determined that BMI1, a member of the polycomb repressive complex 1 (PRC1), is both necessary and sufficient to drive extensive proliferation of self-renewing erythroblasts (SREs). However, the mechanisms of BMI1 action remain poorly understood. BMI1 overexpression led to 10 billion-fold increase BMI1-induced (i)SRE self-renewal. Despite prolonged culture and BMI1 overexpression, human iSREs can terminally mature and agglutinate with typing reagent monoclonal antibodies against conventional RBC antigens. BMI1 and RING1B occupancy, along with repressive histone marks, were identified at known BMI1 target genes, including the INK-ARF locus, consistent with an altered cell cycle following BMI1 inhibition. We also identified upregulated BMI1 target genes with low repressive histone modifications, including key regulator of cholesterol homeostasis. Functional studies suggest that both cholesterol import and synthesis are essential for BMI1-associated self-renewal. These findings support the hypothesis that BMI1 regulates erythroid self-renewal not only through gene repression but also through gene activation and offer a strategy to expand the pool of immature erythroid precursors for eventual clinical uses.

2.
Blood Adv ; 6(10): 3072-3089, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35139174

RESUMEN

Primitive erythropoiesis is a critical component of the fetal cardiovascular network and is essential for the growth and survival of the mammalian embryo. The need to rapidly establish a functional cardiovascular system is met, in part, by the intravascular circulation of primitive erythroid precursors that mature as a single semisynchronous cohort. To better understand the processes that regulate erythroid precursor maturation, we analyzed the proteome, metabolome, and lipidome of primitive erythroblasts isolated from embryonic day (E) 10.5 and E12.5 of mouse gestation, representing their transition from basophilic erythroblast to orthochromatic erythroblast (OrthoE) stages of maturation. Previous transcriptional and biomechanical characterizations of these precursors have highlighted a transition toward the expression of protein elements characteristic of mature red blood cell structure and function. Our analysis confirmed a loss of organelle-specific protein components involved in messenger RNA processing, proteostasis, and metabolism. In parallel, we observed metabolic rewiring toward the pentose phosphate pathway, glycolysis, and the Rapoport-Luebering shunt. Activation of the pentose phosphate pathway in particular may have stemmed from increased expression of hemoglobin chains and band 3, which together control oxygen-dependent metabolic modulation. Increased expression of several antioxidant enzymes also indicated modification to redox homeostasis. In addition, accumulation of oxylipins and cholesteryl esters in primitive OrthoE cells was paralleled by increased transcript levels of the p53-regulated cholesterol transporter (ABCA1) and decreased transcript levels of cholesterol synthetic enzymes. The present study characterizes the extensive metabolic rewiring that occurs in primary embryonic erythroid precursors as they prepare to enucleate and continue circulating without internal organelles.


Asunto(s)
Eritroblastos , Proteómica , Animales , Embrión de Mamíferos/metabolismo , Eritroblastos/metabolismo , Eritropoyesis/genética , Hemoglobinas/metabolismo , Mamíferos , Ratones
3.
Cell Rep ; 36(7): 109562, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407416

RESUMEN

Hematopoietic ontogeny consists of two broad programs: an initial hematopoietic stem cell (HSC)-independent program followed by HSC-dependent hematopoiesis that sequentially seed the fetal liver and generate blood cells. However, the transition from HSC-independent to HSC-derived hematopoiesis remains poorly characterized. To help resolve this question, we developed Mds1CreERT2 mice, which inducibly express Cre-recombinase in emerging HSCs in the aorta and label long-term adult HSCs, but not HSC-independent yolk-sac-derived primitive or definitive erythromyeloid (EMP) hematopoiesis. Our lineage-tracing studies indicate that HSC-derived erythroid, myeloid, and lymphoid progeny significantly expand in the liver and blood stream between E14.5 and E16.5. Additionally, we find that HSCs contribute the majority of F4/80+ macrophages in adult spleen and marrow, in contrast to their limited contribution to macrophage populations in brain, liver, and lungs. The Mds1CreERT2 mouse model will be useful to deconvolute the complexity of hematopoiesis as it unfolds in the embryo and functions postnatally.


Asunto(s)
Envejecimiento/metabolismo , Alelos , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Animales , Linaje de la Célula/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Feto/citología , Hemangioblastos/metabolismo , Hematopoyesis/efectos de los fármacos , Hígado/embriología , Proteína del Locus del Complejo MDS1 y EV11 , Ratones Endogámicos C57BL , Ratones Transgénicos , Tamoxifeno/farmacología
4.
Epigenetics Chromatin ; 14(1): 37, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330317

RESUMEN

BACKGROUND: Condensation of chromatin prior to enucleation is an essential component of terminal erythroid maturation, and defects in this process are associated with inefficient erythropoiesis and anemia. However, the mechanisms involved in this phenomenon are not well understood. Here, we describe a potential role for the histone variant H2A.X in erythropoiesis. RESULTS: We find in multiple model systems that this histone is essential for normal maturation, and that the loss of H2A.X in erythroid cells results in dysregulation in expression of erythroid-specific genes as well as a nuclear condensation defect. In addition, we demonstrate that erythroid maturation is characterized by phosphorylation at both S139 and Y142 on the C-terminal tail of H2A.X during late-stage erythropoiesis. Knockout of the kinase BAZ1B/WSTF results in loss of Y142 phosphorylation and a defect in nuclear condensation, but does not replicate extensive transcriptional changes to erythroid-specific genes observed in the absence of H2A.X. CONCLUSIONS: We relate these findings to Caspase-Initiated Chromatin Condensation (CICC) in terminal erythroid maturation, where aspects of the apoptotic pathway are invoked while apoptosis is specifically suppressed.


Asunto(s)
Cromatina , Eritropoyesis , Caspasas , Histonas/metabolismo , Fosforilación
5.
Science ; 370(6518)2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33184181

RESUMEN

The gene expression program underlying the specification of human cell types is of fundamental interest. We generated human cell atlases of gene expression and chromatin accessibility in fetal tissues. For gene expression, we applied three-level combinatorial indexing to >110 samples representing 15 organs, ultimately profiling ~4 million single cells. We leveraged the literature and other atlases to identify and annotate hundreds of cell types and subtypes, both within and across tissues. Our analyses focused on organ-specific specializations of broadly distributed cell types (such as blood, endothelial, and epithelial), sites of fetal erythropoiesis (which notably included the adrenal gland), and integration with mouse developmental atlases (such as conserved specification of blood cells). These data represent a rich resource for the exploration of in vivo human gene expression in diverse tissues and cell types.


Asunto(s)
Cromatina/metabolismo , Feto/citología , Feto/metabolismo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Análisis de la Célula Individual , Atlas como Asunto , Humanos , Neuronas/metabolismo , Factores de Transcripción/metabolismo
6.
Nat Commun ; 11(1): 4544, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917861

RESUMEN

Stratification of enhancers by signal strength in ChIP-seq assays has resulted in the establishment of super-enhancers as a widespread and useful tool for identifying cell type-specific, highly expressed genes and associated pathways. We examine a distinct method of stratification that focuses on peak breadth, termed hyperacetylated chromatin domains (HCDs), which classifies broad regions exhibiting histone modifications associated with gene activation. We find that this analysis serves to identify genes that are both more highly expressed and more closely aligned to cell identity than super-enhancer analysis does using multiple data sets. Moreover, genetic manipulations of selected gene loci suggest that some enhancers located within HCDs work at least in part via a distinct mechanism involving the modulation of histone modifications across domains and that this activity can be imported into a heterologous gene locus. In addition, such genetic dissection reveals that the super-enhancer concept can obscure important functions of constituent elements.


Asunto(s)
Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Sitios Genéticos/genética , Activación Transcripcional , Acetilación , Animales , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Conjuntos de Datos como Asunto , Embrión de Mamíferos , Eritroblastos , Femenino , Feto , Código de Histonas/genética , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Regiones Promotoras Genéticas/genética , RNA-Seq
7.
Dev Cell ; 53(2): 229-239.e7, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32197069

RESUMEN

Natural killer (NK) cells are a critical component of the innate immune system. However, their ontogenic origin has remained unclear. Here, we report that NK cell potential first arises from Hoxaneg/low Kit+CD41+CD16/32+ hematopoietic-stem-cell (HSC)-independent erythro-myeloid progenitors (EMPs) present in the murine yolk sac. EMP-derived NK cells and primary fetal NK cells, unlike their adult counterparts, exhibit robust degranulation in response to stimulation. Parallel studies using human pluripotent stem cells (hPSCs) revealed that HOXAneg/low CD34+ progenitors give rise to NK cells that, similar to murine EMP-derived NK cells, harbor a potent cytotoxic degranulation bias. In contrast, hPSC-derived HOXA+ CD34+ progenitors, as well as human cord blood CD34+ cells, give rise to NK cells that exhibit an attenuated degranulation response but robustly produce inflammatory cytokines. Collectively, our studies identify an extra-embryonic origin of potently cytotoxic NK cells, suggesting that ontogenic origin is a relevant factor in designing hPSC-derived adoptive immunotherapies.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Células Precursoras Eritroides/citología , Células Madre Hematopoyéticas/citología , Células Asesinas Naturales/patología , Células Progenitoras Mieloides/citología , Animales , Células Madre Embrionarias/metabolismo , Células Precursoras Eritroides/metabolismo , Femenino , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Células Progenitoras Mieloides/metabolismo , Saco Vitelino
8.
Int J Radiat Biol ; 95(11): 1447-1461, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31329495

RESUMEN

Purpose: Incidents, such as nuclear facility accidents and the release of a 'dirty bomb', might result in not only external irradiation of personnel, but additional internal exposures through concomitant inhalation and/or ingestion of radioactive particulates. The purpose of this study was to define the impact of such a combination of radiation injuries on the hematopoietic niche.Material and methods: To assess changes in the murine hematopoietic system, we used a combined exposure of total body irradiation (TBI, 6 Gy) followed immediately by an internal (intraperitoneal) administration of 100 µCi of soluble 137Cs. We then evaluated acute survival in combined versus single modality exposure groups, as well as assessing hematopoietic function at 12 and 26 week time points.Results: Acutely, the combination of external and internal exposures led to an unexpected delay in excretion of 137Cs, increasing the absorbed dose in the combined exposure group and leading to mortality from an acute hematopoietic syndrome. At 12 weeks, all exposure paradigms resulted in decreased numbers of phenotypic hematopoietic stem cells (HSCs), particularly the short-term HSCs (ST-HSC); long-term HSCs (LT-HSC) were depleted only in the internal and combined exposure groups. At 26 weeks, there was significant anemia in both the TBI alone and combined exposure groups. There were decreased numbers in both the LT- and ST-HSCs and decreased functionality, as measured by competitive repopulation, was seen in all radiation groups, with the greatest effects seen in the internal and combined exposure groups.Conclusions: Our data indicate that a combined injury of sublethal external irradiation with internal contamination induces significant and persistent changes in the hematopoietic system, as may have been predicted from the literature and our own group's findings. However, a novel observation was that the combined exposure led to an alteration in the excretion kinetics of the internal contamination, increasing the acute effects beyond those anticipated. As a result, we believe that a combined exposure poses a unique challenge to the medical community during both the acute and, possibly, delayed recovery stages.


Asunto(s)
Médula Ósea/efectos de la radiación , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Irradiación Corporal Total , Animales , Células Cultivadas , Radioisótopos de Cesio , Femenino , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos C57BL , Fenotipo , Traumatismos Experimentales por Radiación/fisiopatología , Especies Reactivas de Oxígeno/metabolismo
9.
Blood Adv ; 3(1): 72-82, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622145

RESUMEN

Platelets are essential for hemostasis; however, several studies have identified age-dependent differences in platelet function. To better understand the origins of fetal platelet function, we have evaluated the contribution of the fetal-specific RNA binding protein Lin28b in the megakaryocyte/platelet lineage. Because activated fetal platelets have very low levels of P-selectin, we hypothesized that the expression of platelet P-selectin is part of a fetal-specific hematopoietic program conferred by Lin28b. Using the mouse as a model, we find that activated fetal platelets have low levels of P-selectin and do not readily associate with granulocytes in vitro and in vivo, relative to adult controls. Transcriptional analysis revealed high levels of Lin28b and Hmga2 in fetal, but not adult, megakaryocytes. Overexpression of LIN28B in adult mice significantly reduces the expression of P-selectin in platelets, and therefore identifies Lin28b as a negative regulator of P-selectin expression. Transplantation of fetal hematopoietic progenitors resulted in the production of platelets with low levels of P-selectin, suggesting that the developmental regulation of P-selectin is intrinsic and independent of differences between fetal and adult microenvironments. Last, we observe that the upregulation of P-selectin expression occurs postnatally, and the temporal kinetics of this upregulation are recapitulated by transplantation of fetal hematopoietic stem and progenitor cells into adult recipients. Taken together, these studies identify Lin28b as a new intrinsic regulator of fetal platelet function.


Asunto(s)
Plaquetas/metabolismo , Regulación de la Expresión Génica , Proteínas de Unión al ARN/genética , Factores de Edad , Animales , Biomarcadores , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Selectina-P/genética , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria/genética , Pruebas de Función Plaquetaria , Proteínas de Unión al ARN/metabolismo
10.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30366982

RESUMEN

Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyetina/metabolismo , Ferroquelatasa/metabolismo , Hemo/biosíntesis , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Células Eritroides/citología , Eritropoyesis , Células HEK293 , Humanos , Proteínas de la Membrana/química , Ratones , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Transporte de Proteínas
11.
Biomicrofluidics ; 11(5): 054112, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29085523

RESUMEN

Primitive erythroblasts (precursors of red blood cells) enter vascular circulation during the embryonic period and mature while circulating. As a result, primitive erythroblasts constantly experience significant hemodynamic shear stress. Shear-induced deformation of primitive erythroblasts however, is poorly studied. In this work, we examined the deformability of primitive erythroblasts at physiologically relevant flow conditions in microfluidic channels and identified the regulatory roles of the maturation stage of primitive erythroblasts and cytoskeletal protein 4.1 R in shear-induced cell deformation. The results showed that the maturation stage affected the deformability of primitive erythroblasts significantly and that primitive erythroblasts at later maturational stages exhibited a better deformability due to a matured cytoskeletal structure in the cell membrane.

12.
Sci Rep ; 7(1): 5164, 2017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28701737

RESUMEN

Hematopoietic ontogeny is characterized by distinct primitive and definitive erythroid lineages. Definitive erythroblasts mature and enucleate extravascularly and form a unique membrane skeleton, composed of spectrin, 4.1R-complex, and ankyrinR-complex components, to survive the vicissitudes of the adult circulation. However, little is known about the formation and composition of the membrane skeleton in primitive erythroblasts, which progressively mature while circulating in the embryonic bloodstream. We found that primary primitive erythroblasts express the major membrane skeleton genes present in similarly staged definitive erythroblasts, suggesting that the composition and formation of this membrane network is conserved in maturing primitive and definitive erythroblasts despite their respective intravascular and extravascular locations. Membrane deformability and stability of primitive erythroblasts, assayed by microfluidic studies and fluorescence imaged microdeformation, respectively, significantly increase prior to enucleation. These functional changes coincide with protein 4.1 R isoform switching and protein 4.1R-null primitive erythroblasts fail to establish normal membrane stability and deformability. We conclude that maturing primitive erythroblasts initially navigate the embryonic vasculature prior to establishing a deformable cytoskeleton, which is ultimately formed prior to enucleation. Formation of an erythroid-specific, protein 4.1R-dependent membrane skeleton is an important feature not only of definitive, but also of primitive, erythropoiesis in mammals.


Asunto(s)
Diferenciación Celular , Eritroblastos/metabolismo , Eritropoyesis , Proteínas de Microfilamentos/metabolismo , Empalme Alternativo , Animales , Diferenciación Celular/genética , Línea Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Eritroblastos/citología , Membrana Eritrocítica/metabolismo , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética
13.
Exp Hematol ; 44(10): 947-63, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27373493

RESUMEN

Adenosine deaminases that act on RNA (ADARs) convert adenosine residues to inosine in double-stranded RNA. In vivo, ADAR1 is essential for the maintenance of hematopoietic stem/progenitors. Whether other hematopoietic cell types also require ADAR1 has not been assessed. Using erythroid- and myeloid-restricted deletion of Adar1, we demonstrate that ADAR1 is dispensable for myelopoiesis but is essential for normal erythropoiesis. Adar1-deficient erythroid cells display a profound activation of innate immune signaling and high levels of cell death. No changes in microRNA levels were found in ADAR1-deficient erythroid cells. Using an editing-deficient allele, we demonstrate that RNA editing is the essential function of ADAR1 during erythropoiesis. Mapping of adenosine-to-inosine editing in purified erythroid cells identified clusters of hyperedited adenosines located in long 3'-untranslated regions of erythroid-specific transcripts and these are ADAR1-specific editing events. ADAR1-mediated RNA editing is essential for normal erythropoiesis.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina/genética , Eritropoyesis , Inosina/genética , Edición de ARN , Adenosina Desaminasa/genética , Animales , Análisis por Conglomerados , Índices de Eritrocitos , Células Eritroides/metabolismo , Eritropoyesis/genética , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Granulocitos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Interferones/metabolismo , Ratones , MicroARNs/genética , Mielopoyesis/genética , Especificidad de Órganos , Fenotipo , Proteínas de Unión al ARN/genética , Receptores de Interferón/metabolismo , Retroelementos , Transducción de Señal , Transcripción Genética
14.
Stem Cell Reports ; 4(6): 995-1003, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26028528

RESUMEN

Red blood cells (RBCs), responsible for oxygen delivery and carbon dioxide exchange, are essential for our well-being. Alternative RBC sources are needed to meet the increased demand for RBC transfusions projected to occur as our population ages. We previously have discovered that erythroblasts derived from the early mouse embryo can self-renew extensively ex vivo for many months. To better understand the mechanisms regulating extensive erythroid self-renewal, global gene expression data sets from self-renewing and differentiating erythroblasts were analyzed and revealed the differential expression of Bmi-1. Bmi-1 overexpression conferred extensive self-renewal capacity upon adult bone-marrow-derived self-renewing erythroblasts, which normally have limited proliferative potential. Importantly, Bmi-1 transduction did not interfere with the ability of extensively self-renewing erythroblasts (ESREs) to terminally mature either in vitro or in vivo. Bmi-1-induced ESREs can serve to generate in vitro models of erythroid-intrinsic disorders and ultimately may serve as a source of cultured RBCs for transfusion therapy.


Asunto(s)
Eritroblastos/citología , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Dexametasona/farmacología , Eritroblastos/metabolismo , Eritroblastos/trasplante , Eritropoyetina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Complejo Represivo Polycomb 1/antagonistas & inhibidores , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Células Madre/farmacología , Irradiación Corporal Total
15.
Cell Rep ; 11(12): 1892-904, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26095363

RESUMEN

Hematopoietic potential arises in mammalian embryos before adult-repopulating hematopoietic stem cells (HSCs). At embryonic day 9.5 (E9.5), we show the first murine definitive erythro-myeloid progenitors (EMPs) have an immunophenotype distinct from primitive hematopoietic progenitors, maturing megakaryocytes and macrophages, and rare B cell potential. EMPs emerge in the yolk sac with erythroid and broad myeloid, but not lymphoid, potential. EMPs migrate to the fetal liver and rapidly differentiate, including production of circulating neutrophils by E11.5. Although the surface markers, transcription factors, and lineage potential associated with EMPs overlap with those found in adult definitive hematopoiesis, they are present in unique combinations or proportions that result in a specialized definitive embryonic progenitor. Furthermore, we find that embryonic stem cell (ESC)-derived hematopoiesis recapitulates early yolk sac hematopoiesis, including primitive, EMP, and rare B cell potential. EMPs do not have long-term potential when transplanted in immunocompromised adults, but they can provide transient adult-like RBC reconstitution.


Asunto(s)
Desarrollo Embrionario/genética , Células Madre Embrionarias , Hematopoyesis , Células Madre Hematopoyéticas , Animales , Células Sanguíneas/citología , Linaje de la Célula , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Ratones , Saco Vitelino/citología , Saco Vitelino/crecimiento & desarrollo
16.
Sci Signal ; 8(372): ra34, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25872869

RESUMEN

In multicellular organisms, the mechanisms by which diverse cell types acquire distinct amino acids and how cellular function adapts to their availability are fundamental questions in biology. We found that increased neutral essential amino acid (NEAA) uptake was a critical component of erythropoiesis. As red blood cells matured, expression of the amino acid transporter gene Lat3 increased, which increased NEAA import. Inadequate NEAA uptake by pharmacologic inhibition or RNAi-mediated knockdown of LAT3 triggered a specific reduction in hemoglobin production in zebrafish embryos and murine erythroid cells through the mTORC1 (mammalian target of rapamycin complex 1)/4E-BP (eukaryotic translation initiation factor 4E-binding protein) pathway. CRISPR-mediated deletion of members of the 4E-BP family in murine erythroid cells rendered them resistant to mTORC1 and LAT3 inhibition and restored hemoglobin production. These results identify a developmental role for LAT3 in red blood cells and demonstrate that mTORC1 serves as a homeostatic sensor that couples hemoglobin production at the translational level to sufficient uptake of NEAAs, particularly L-leucine.


Asunto(s)
Proteínas Portadoras/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Hemoglobinas/metabolismo , Leucina/metabolismo , Complejos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Proteínas Portadoras/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Células Cultivadas , Embrión de Mamíferos/irrigación sanguínea , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoyesis/genética , Factores Eucarióticos de Iniciación/genética , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Hemoglobinas/genética , Humanos , Immunoblotting , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Microscopía Confocal , Complejos Multiproteicos/genética , Fosfoproteínas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Pez Cebra
17.
J Clin Invest ; 124(10): 4294-304, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25157825

RESUMEN

The transport and intracellular trafficking of heme biosynthesis intermediates are crucial for hemoglobin production, which is a critical process in developing red cells. Here, we profiled gene expression in terminally differentiating murine fetal liver-derived erythroid cells to identify regulators of heme metabolism. We determined that TMEM14C, an inner mitochondrial membrane protein that is enriched in vertebrate hematopoietic tissues, is essential for erythropoiesis and heme synthesis in vivo and in cultured erythroid cells. In mice, TMEM14C deficiency resulted in porphyrin accumulation in the fetal liver, erythroid maturation arrest, and embryonic lethality due to profound anemia. Protoporphyrin IX synthesis in TMEM14C-deficient erythroid cells was blocked, leading to an accumulation of porphyrin precursors. The heme synthesis defect in TMEM14C-deficient cells was ameliorated with a protoporphyrin IX analog, indicating that TMEM14C primarily functions in the terminal steps of the heme synthesis pathway. Together, our data demonstrate that TMEM14C facilitates the import of protoporphyrinogen IX into the mitochondrial matrix for heme synthesis and subsequent hemoglobin production. Furthermore, the identification of TMEM14C as a protoporphyrinogen IX importer provides a genetic tool for further exploring erythropoiesis and congenital anemias.


Asunto(s)
Eritropoyesis/genética , Hemo/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Anemia/metabolismo , Animales , Línea Celular , Células Eritroides/metabolismo , Regulación de la Expresión Génica , Hemoglobinas/metabolismo , Hígado/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Transporte de Membrana Mitocondrial/genética , Membranas Mitocondriales/metabolismo , Porfirinas/metabolismo , Protoporfirinas/metabolismo , ARN Interferente Pequeño/metabolismo
18.
Blood ; 124(2): 277-86, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24735964

RESUMEN

Megakaryocyte (MK) development in the bone marrow progresses spatially from the endosteal niche, which promotes MK progenitor proliferation, to the sinusoidal vascular niche, the site of terminal maturation and thrombopoiesis. The chemokine stromal cell-derived factor-1 (SDF-1), signaling through CXCR4, is implicated in the maturational chemotaxis of MKs toward sinusoidal vessels. Here, we demonstrate that both IV administration of SDF-1 and stabilization of endogenous SDF-1 acutely increase MK-vasculature association and thrombopoiesis with no change in MK number. In the setting of radiation injury, we find dynamic fluctuations in marrow SDF-1 distribution that spatially and temporally correlate with variations in MK niche occupancy. Stabilization of altered SDF-1 gradients directly affects MK location. Importantly, these SDF-1-mediated changes have functional consequences for platelet production, as the movement of MKs away from the vasculature decreases circulating platelets, while MK association with the vasculature increases circulating platelets. Finally, we demonstrate that manipulation of SDF-1 gradients can improve radiation-induced thrombocytopenia in a manner additive with earlier TPO treatment. Taken together, our data support the concept that SDF-1 regulates the spatial distribution of MKs in the marrow and consequently circulating platelet numbers. This knowledge of the microenvironmental regulation of the MK lineage could lead to improved therapeutic strategies for thrombocytopenia.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/fisiología , Megacariocitos/citología , Megacariocitos/fisiología , Traumatismos Experimentales por Radiación , Nicho de Células Madre/genética , Trombopoyesis/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/efectos de la radiación , Células Cultivadas , Quimiocina CXCL12/administración & dosificación , Femenino , Células Progenitoras de Megacariocitos/citología , Células Progenitoras de Megacariocitos/efectos de los fármacos , Células Progenitoras de Megacariocitos/fisiología , Células Progenitoras de Megacariocitos/efectos de la radiación , Megacariocitos/efectos de los fármacos , Megacariocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/patología , Receptores CXCR4/administración & dosificación , Receptores CXCR4/metabolismo , Nicho de Células Madre/efectos de los fármacos , Nicho de Células Madre/efectos de la radiación , Trombopoyesis/efectos de los fármacos , Trombopoyesis/efectos de la radiación
19.
Cell Metab ; 17(3): 343-52, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23416069

RESUMEN

Sorting of endocytic ligands and receptors is critical for diverse cellular processes. The physiological significance of endosomal sorting proteins in vertebrates, however, remains largely unknown. Here we report that sorting nexin 3 (Snx3) facilitates the recycling of transferrin receptor (Tfrc) and thus is required for the proper delivery of iron to erythroid progenitors. Snx3 is highly expressed in vertebrate hematopoietic tissues. Silencing of Snx3 results in anemia and hemoglobin defects in vertebrates due to impaired transferrin (Tf)-mediated iron uptake and its accumulation in early endosomes. This impaired iron assimilation can be complemented with non-Tf iron chelates. We show that Snx3 and Vps35, a component of the retromer, interact with Tfrc to sort it to the recycling endosomes. Our findings uncover a role of Snx3 in regulating Tfrc recycling, iron homeostasis, and erythropoiesis. Thus, the identification of Snx3 provides a genetic tool for exploring erythropoiesis and disorders of iron metabolism.


Asunto(s)
Anemia/genética , Hierro/metabolismo , Receptores de Transferrina/metabolismo , Nexinas de Clasificación/metabolismo , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Ratones , Nexinas de Clasificación/genética , Pez Cebra
20.
Blood ; 121(6): e5-e13, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23243273

RESUMEN

Erythroid ontogeny is characterized by overlapping waves of primitive and definitive erythroid lineages that share many morphologic features during terminal maturation but have marked differences in cell size and globin expression. In the present study, we compared global gene expression in primitive, fetal definitive, and adult definitive erythroid cells at morphologically equivalent stages of maturation purified from embryonic, fetal, and adult mice. Surprisingly, most transcriptional complexity in erythroid precursors is already present by the proerythroblast stage. Transcript levels are markedly modulated during terminal erythroid maturation, but housekeeping genes are not preferentially lost. Although primitive and definitive erythroid lineages share a large set of nonhousekeeping genes, annotation of lineage-restricted genes shows that alternate gene usage occurs within shared functional categories, as exemplified by the selective expression of aquaporins 3 and 8 in primitive erythroblasts and aquaporins 1 and 9 in adult definitive erythroblasts. Consistent with the known functions of Aqp3 and Aqp8 as H2O2 transporters, primitive, but not definitive, erythroblasts preferentially accumulate reactive oxygen species after exogenous H2O2 exposure. We have created a user-friendly Web site (http://www.cbil.upenn.edu/ErythronDB) to make these global expression data readily accessible and amenable to complex search strategies by the scientific community.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyesis/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Animales , Acuaporina 1/genética , Acuaporina 3/genética , Acuaporinas/genética , Linaje de la Célula/genética , Células Cultivadas , Eritroblastos/metabolismo , Eritrocitos/metabolismo , Femenino , Sistema Hematopoyético/citología , Sistema Hematopoyético/embriología , Sistema Hematopoyético/crecimiento & desarrollo , Ratones , Ratones Endogámicos ICR , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA