Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.190
Filtrar
1.
Methods Mol Biol ; 2835: 99-110, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39105909

RESUMEN

Induced pluripotent stem cells (iPSCs) are generated through the reprogramming of somatic cells to an embryonic-like state by activating specific genes. They closely resemble embryonic stem cells (ESCs), in various aspects, including the expression of key stem cell genes, potency, and differentiation capabilities. iPSCs can be derived from various cell types such as fibroblasts, keratinocytes, and peripheral blood mononuclear cells (PBMCs). The ease of obtaining origin cells through non-invasive methods simplifies the generation of human iPSCs. Therefore, PBMCs are commonly preferred, with erythroid progenitor cells (EPCs) obtained through EPC enrichment being used as origin cells in this protocol. The EPC enrichment performed in this protocol not only reduces costs but also increases efficiency by enhancing the percentage of reprogrammable cells with progenitor characteristics. Human iPSCs are incredibly valuable for in vitro research, cell therapy, drug discovery, and tissue engineering. The outlined procedures below provide a general framework for inducing iPSCs from erythroid progenitor cells, pluripotency confirmation experiments, and cultivating them for downstream experiments.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Células Precursoras Eritroides , Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Técnicas de Cultivo de Célula/métodos , Reprogramación Celular/genética , Células Cultivadas , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo
2.
Exp Hematol ; 136: 104283, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39048071

RESUMEN

Red blood cells (RBCs) comprise a critical component of the cardiovascular network, which constitutes the first functional organ system of the developing mammalian embryo. Examination of circulating blood cells in mammalian embryos revealed two distinct types of erythroid cells: large, nucleated "primitive" erythroblasts followed by smaller, enucleated "definitive" erythrocytes. This review describes the current understanding of primitive and definitive erythropoiesis gleaned from studies of mouse and human embryos and induced pluripotent stem cells (iPSCs). Primitive erythropoiesis in the mouse embryo comprises a transient wave of committed primitive erythroid progenitors (primitive erythroid colony-forming cells, EryP-CFC) in the early yolk sac that generates a robust cohort of precursors that mature in the bloodstream and enucleate. In contrast, definitive erythropoiesis has two distinct developmental origins. The first comprises a transient wave of definitive erythroid progenitors (burst-forming units erythroid, BFU-E) that emerge in the yolk sac and seed the fetal liver where they terminally mature to provide the first definitive RBCs. The second comprises hematopoietic stem cell (HSC)-derived BFU-E that terminally mature at sites colonized by HSCs particularly the fetal liver and subsequently the bone marrow. Primitive and definitive erythropoiesis are derived from endothelial identity precursors with distinct developmental origins. Although they share prototypical transcriptional regulation, primitive and definitive erythropoiesis are also characterized by distinct lineage-specific factors. The exquisitely timed, sequential production of primitive and definitive erythroid cells is necessary for the survival and growth of the mammalian embryo.


Asunto(s)
Embrión de Mamíferos , Eritropoyesis , Eritropoyesis/fisiología , Animales , Humanos , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Ratones , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Saco Vitelino/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo
3.
Cells ; 13(13)2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38995000

RESUMEN

Erythropoiesis occurs first in the yolk sac as a transit "primitive" form, then is gradually replaced by the "definitive" form in the fetal liver (FL) during fetal development and in the bone marrow (BM) postnatally. While it is well known that differences exist between primitive and definitive erythropoiesis, the similarities and differences between FL and BM definitive erythropoiesis have not been studied. Here we performed comprehensive comparisons of erythroid progenitors and precursors at all maturational stages sorted from E16.5 FL and adult BM. We found that FL cells at all maturational stages were larger than their BM counterparts. We further found that FL BFU-E cells divided at a faster rate and underwent more cell divisions than BM BFU-E. Transcriptome comparison revealed that genes with increased expression in FL BFU-Es were enriched in cell division. Interestingly, the expression levels of glucocorticoid receptor Nr3c1, Myc and Myc downstream target Ccna2 were significantly higher in FL BFU-Es, indicating the role of the Nr3c1-Myc-Ccna2 axis in the enhanced proliferation/cell division of FL BFU-E cells. At the CFU-E stage, the expression of genes associated with hemoglobin biosynthesis were much higher in FL CFU-Es, indicating more hemoglobin production. During terminal erythropoiesis, overall temporal patterns in gene expression were conserved between the FL and BM. While biological processes related to translation, the tricarboxylic acid cycle and hypoxia response were upregulated in FL erythroblasts, those related to antiviral signal pathway were upregulated in BM erythroblasts. Our findings uncovered previously unrecognized differences between FL and BM definitive erythropoiesis and provide novel insights into erythropoiesis.


Asunto(s)
Médula Ósea , Eritropoyesis , Feto , Hígado , Transcriptoma , Animales , Eritropoyesis/genética , Ratones , Hígado/metabolismo , Hígado/embriología , Hígado/citología , Transcriptoma/genética , Feto/metabolismo , Feto/citología , Médula Ósea/metabolismo , Ratones Endogámicos C57BL , Regulación del Desarrollo de la Expresión Génica , Femenino , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología
4.
Cytotherapy ; 26(11): 1362-1373, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39001769

RESUMEN

BACKGROUND AIMS: Ex vivo production of red blood cells (RBCs) represents a promising alternative for transfusion medicine. Several strategies have been described to generate erythroid cell lines from different sources, including embryonic, induced pluripotent, and hematopoietic stem cells. All these approaches have in common that they require elaborate differentiation cultures whereas the yield of enucleated RBCs is inefficient. METHODS: We generated a human immortalized adult erythroid progenitor cell line derived from bone marrow CD71-positive erythroid progenitor cells (immortalized bone marrow erythroid progenitor adult, or imBMEP-A) by an inducible expression system, to shorten differentiation culture necessary for terminal erythroid differentiation. It is the first erythroid cell line that is generated from direct reticulocyte progenitors and demonstrates robust hemoglobin production in the immortalized state. RESULTS: Morphologic analysis of the immortalized cells showed that the preferred cell type of the imBMEP-A line corresponds to hemoglobin-producing basophilic erythroblasts. In addition, we were able to generate a stable cell line from a single cell clone with the triple knockout of RhAG, RhDCE and KELL. After removal of doxycycline, part of the cells differentiated into normoblasts and reticulocytes within 5-7 days. CONCLUSIONS: Our results demonstrate that the imBMEP-A cell line can serve as a stable and straightforward modifiable platform for RBC engineering in the future.


Asunto(s)
Antígenos CD , Diferenciación Celular , Células Precursoras Eritroides , Receptores de Transferrina , Humanos , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Receptores de Transferrina/metabolismo , Antígenos CD/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Eritropoyesis , Línea Celular , Eritrocitos/citología , Eritrocitos/metabolismo , Reticulocitos/citología , Reticulocitos/metabolismo
5.
Eur J Haematol ; 113(4): 416-425, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38853593

RESUMEN

OBJECTIVES: Galectin-9 (Gal-9) is an immune checkpoint ligand for T-cell immunoglobulin and mucin domain 3. Although the roles of Gal-9 in regulating immune responses have been well investigated, their biological roles have yet to be fully documented. This study aimed to analyse the expression of Gal-9 bone marrow (BM) cells in C57BL/6J (B6) mice. Furthermore, the co-expression of Gal-9 with the mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) was investigated. METHODS: The BM cells in adult C57BL/6J (B6) mice were collected and analysed in vitro. RESULTS: In a flow cytometric analysis of BM cells, Gal-9 was highly expressed in c-KithiSca-1-CD34-CD71+ erythroid progenitors (EPs), whereas it was downregulated in more differentiated c-KitloCD71+TER119+ cells. Subsequently, a negative selection of CD3-B220-Sca-1-CD34-CD41-CD16/32- EPs was performed. This resulted in substantial enrichment of KithiCD71+Gal-9+ cells and erythroid colony-forming units (CFU-Es), suggesting that the colony-forming subset of EPs are included in the KithiCD71+Gal-9+ population. Furthermore, we found that EPs had lower mTOR and AMPK expression levels in Gal-9 knockout B6 mice than in wild-type B6 mice. CONCLUSIONS: These results may stimulate further investigation of the role of Gal-9 in haematopoiesis.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Células Precursoras Eritroides , Galectinas , Serina-Treonina Quinasas TOR , Animales , Galectinas/metabolismo , Galectinas/genética , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología , Ratones Endogámicos C57BL , Expresión Génica , Inmunofenotipificación , Biomarcadores , Células de la Médula Ósea/metabolismo
6.
Blood ; 143(22): 2300-2313, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38447046

RESUMEN

ABSTRACT: Long noncoding RNAs (lncRNAs) are extensively expressed in eukaryotic cells and have been revealed to be important for regulating cell differentiation. Many lncRNAs have been found to regulate erythroid differentiation in the mouse. However, given the low sequence conservation of lncRNAs between mouse and human, our understanding of lncRNAs in human erythroid differentiation remains incomplete. lncRNAs are often transcribed opposite to protein coding genes and regulate their expression. Here, we characterized a human erythrocyte-expressed lncRNA, GATA2AS, which is transcribed opposite to erythroid transcription regulator GATA2. GATA2AS is a 2080-bp long, primarily nucleus-localized noncoding RNA that is expressed in erythroid progenitor cells and decreases during differentiation. Knockout of GATA2AS in human HUDEP2 erythroid progenitor cells using CRISPR-Cas9 genome editing to remove the transcription start site accelerated erythroid differentiation and dysregulated erythroblast gene expression. We identified GATA2AS as a novel GATA2 and HBG activator. Chromatin isolation by RNA purification showed that GATA2AS binds to thousands of genomic sites and colocalizes at a subset of sites with erythroid transcription factors including LRF and KLF1. RNA pulldown and RNA immunoprecipitation confirmed interaction between GATA2AS and LRF and KLF1. Chromatin immunoprecipitation sequencing (ChIP-seq) showed that knockout of GATA2AS reduces binding of these transcription factors genome wide. Assay for transposase-accessible chromatin sequencing (ATAC-seq) and H3K27ac ChIP-seq showed that GATA2AS is essential to maintain the chromatin regulatory landscape during erythroid differentiation. Knockdown of GATA2AS in human primary CD34+ cells mimicked results in HUDEP2 cells. Overall, our results implicate human-specific lncRNA GATA2AS as a regulator of erythroid differentiation by influencing erythroid transcription factor binding and the chromatin regulatory landscape.


Asunto(s)
Cromatina , Eritropoyesis , Factor de Transcripción GATA2 , ARN Largo no Codificante , Humanos , Eritropoyesis/genética , ARN Largo no Codificante/genética , Cromatina/metabolismo , Cromatina/genética , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología
7.
Exp Hematol ; 135: 104191, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38493949

RESUMEN

Erythropoiesis in the adult bone marrow relies on mitochondrial membrane transporters to facilitate heme and hemoglobin production. Erythrocytes in the bone marrow are produced although the differentiation of erythroid progenitor cells that originate from hematopoietic stem cells (HSCs). Whether and how mitochondria transporters potentiate HSCs and affect their differentiation toward erythroid lineage remains unclear. Here, we show that the ATP-binding cassette (ABC) transporter 10 (Abcb10), located on the inner mitochondrial membrane, is essential for HSC maintenance and erythroid-lineage differentiation. Induced deletion of Abcb10 in adult mice significantly increased erythroid progenitor cell and decreased HSC number within the bone marrow (BM). Functionally, Abcb10-deficient HSCs exhibited significant decreases in stem cell potential but with a skew toward erythroid-lineage differentiation. Mechanistically, deletion of Abcb10 rendered HSCs with excess mitochondrial iron accumulation and oxidative stress yet without alteration in mitochondrial bioenergetic function. However, impaired hematopoiesis could not be rescued through the in vivo administration of a mitochondrial iron chelator or antioxidant to Abcb10-deficient mice. Abcb10-mediated mitochondrial iron transfer is thus pivotal for the regulation of physiologic HSC potential and erythroid-lineage differentiation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Diferenciación Celular , Eritropoyesis , Células Madre Hematopoyéticas , Ratones Noqueados , Mitocondrias , Animales , Ratones , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Mitocondrias/metabolismo , Eritropoyesis/genética , Hierro/metabolismo , Células Eritroides/citología , Células Eritroides/metabolismo , Estrés Oxidativo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citología , Ratones Endogámicos C57BL
8.
Exp Hematol ; 106: 19-30, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34879257

RESUMEN

Calcium (Ca2+) is an important second messenger molecule in the body, regulating cell cycle and fate. There is growing evidence that intracellular Ca2+ levels play functional roles in the total physiological process of erythroid differentiation, including the proliferation and differentiation of erythroid progenitor cells, terminal enucleation, and mature red blood cell aging and clearance. Moreover, recent research on the pathology of erythroid disorders has made great progress in the past decades, indicating that calcium ion hemostasis is closely related to ineffective erythropoiesis and increased sensitivity to stress factors. In this review, we summarized what is known about the functional roles of intracellular Ca2+ in erythropoiesis and erythrocyte-related diseases, with an emphasis on the regulation of the intracellular Ca2+ homeostasis during erythroid differentiation. An understanding of the regulation roles of Ca2+ homeostasis in erythroid differentiation will facilitate further studies and eventually molecular identification of the pathways involved in the pathological process of erythroid disorders, providing new therapeutic opportunities in erythrocyte-related disease.


Asunto(s)
Calcio/metabolismo , Eritropoyesis , Animales , Cationes Bivalentes/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Humanos
9.
Cell Rep ; 36(6): 109507, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34380040

RESUMEN

Survival or apoptosis is a binary decision in individual cells. However, at the cell-population level, a graded increase in survival of colony-forming unit-erythroid (CFU-E) cells is observed upon stimulation with erythropoietin (Epo). To identify components of Janus kinase 2/signal transducer and activator of transcription 5 (JAK2/STAT5) signal transduction that contribute to the graded population response, we extended a cell-population-level model calibrated with experimental data to study the behavior in single cells. The single-cell model shows that the high cell-to-cell variability in nuclear phosphorylated STAT5 is caused by variability in the amount of Epo receptor (EpoR):JAK2 complexes and of SHP1, as well as the extent of nuclear import because of the large variance in the cytoplasmic volume of CFU-E cells. 24-118 pSTAT5 molecules in the nucleus for 120 min are sufficient to ensure cell survival. Thus, variability in membrane-associated processes is sufficient to convert a switch-like behavior at the single-cell level to a graded population-level response.


Asunto(s)
Citoplasma/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Janus Quinasa 2/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Animales , Calibración , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Simulación por Computador , Eritropoyetina/farmacología , Ratones Endogámicos BALB C , Modelos Biológicos , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Genes (Basel) ; 12(8)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34440315

RESUMEN

Hydroxyurea (HU) causes nitric oxide (NO) bioactivation, acting as both a NO donor and a stimulator of NO synthase (NOS). To examine whether HU effects are NO mediated by chemical degradation or enzymatic induction, we studied human and mouse erythroid cells during proliferation, apoptosis, and differentiation. The HU and NO donor demonstrated persisted versus temporary inhibition of erythroid cell growth during differentiation, as observed by γ- and ß-globin gene expression. HU decreased the percentage of erythroleukemic K562 cells in the G2/M phase that was reversed by N-nitro l-arginine methyl ester hydrochloride (L-NAME). Besides activation of endothelial NOS, HU significantly increased apoptosis of K562 cells, again demonstrating NOS dependence. Administration of HU to mice significantly inhibited colony-forming unit-erythroid (CFU-E), mediated by NOS. Moreover, burst-forming-units-erythroid (BFU-E) and CFU-E ex vivo growth was inhibited by the administration of nitrate or nitrite to mice. Chronic in vivo NOS inhibition with L-NAME protected the bone marrow cellularity despite HU treatment of mice. NO metabolites and HU reduced the frequency of NOS-positive cells from CFU-E and BFU-E colonies that was reverted by NOS inhibition. HU regulation of the G2/M phase, apoptosis, differentiation, cellularity, and NOS immunoreactive cells was NOS dependent. Inhalation of NO therapy as well as strategies to increase endogenous NO production could replace or enhance HU activity.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Hidroxiurea/farmacología , Óxido Nítrico Sintasa/metabolismo , Animales , Células Precursoras Eritroides/citología , Humanos , Células K562 , Ratones , Donantes de Óxido Nítrico/farmacología
11.
Genes (Basel) ; 12(7)2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208866

RESUMEN

Hematopoiesis is a convenient model to study how chromatin dynamics plays a decisive role in regulation of cell fate. During erythropoiesis a population of stem and progenitor cells becomes increasingly lineage restricted, giving rise to terminally differentiated progeny. The concerted action of transcription factors and epigenetic modifiers leads to a silencing of the multipotent transcriptome and activation of the transcriptional program that controls terminal differentiation. This article reviews some aspects of the biology of red blood cells production with the focus on the extensive chromatin reorganization during differentiation.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Células Precursoras Eritroides/citología , Eritropoyesis , Regulación de la Expresión Génica , Factores de Transcripción/metabolismo , Animales , Genoma , Humanos , Factores de Transcripción/genética
12.
Am J Hematol ; 96(9): 1064-1076, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34021930

RESUMEN

Identification of stage-specific erythroid cells is critical for studies of normal and disordered human erythropoiesis. While immunophenotypic strategies have previously been developed to identify cells at each stage of terminal erythroid differentiation, erythroid progenitors are currently defined very broadly. Refined strategies to identify and characterize BFU-E and CFU-E subsets are critically needed. To address this unmet need, a flow cytometry-based technique was developed that combines the established surface markers CD34 and CD36 with CD117, CD71, and CD105. This combination allowed for the separation of erythroid progenitor cells into four discrete populations along a continuum of progressive maturation, with increasing cell size and decreasing nuclear/cytoplasmic ratio, proliferative capacity and stem cell factor responsiveness. This strategy was validated in uncultured, primary erythroid cells isolated from bone marrow of healthy individuals. Functional colony assays of these progenitor populations revealed enrichment of BFU-E only in the earliest population, transitioning to cells yielding BFU-E and CFU-E, then CFU-E only. Utilizing CD34/CD105 and GPA/CD105 profiles, all four progenitor stages and all five stages of terminal erythroid differentiation could be identified. Applying this immunophenotyping strategy to primary bone marrow cells from patients with myelodysplastic syndrome, identified defects in erythroid progenitors and in terminal erythroid differentiation. This novel immunophenotyping technique will be a valuable tool for studies of normal and perturbed human erythropoiesis. It will allow for the discovery of stage-specific molecular and functional insights into normal erythropoiesis as well as for identification and characterization of stage-specific defects in inherited and acquired disorders of erythropoiesis.


Asunto(s)
Células Eritroides/citología , Células Precursoras Eritroides/citología , Eritropoyesis , Antígenos CD/análisis , Antígenos CD34/análisis , Células de la Médula Ósea/citología , Células Cultivadas , Endoglina/análisis , Citometría de Flujo/métodos , Humanos , Inmunofenotipificación/métodos
13.
PLoS One ; 16(3): e0247858, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33661998

RESUMEN

Myeloproliferative neoplasms (MPNs) cause the over-production of blood cells such as erythrocytes (polycythemia vera) or platelets (essential thrombocytosis). JAK2 V617F is the most prevalent somatic mutation in many MPNs, but previous modeling of this mutation in mice relied on transgenic overexpression and resulted in diverse phenotypes that were in some cases attributed to expression level. CRISPR-Cas9 engineering offers new possibilities to model and potentially cure genetically encoded disorders via precise modification of the endogenous locus in primary cells. Here we develop "scarless" Cas9-based reagents to create and reverse the JAK2 V617F mutation in an immortalized human erythroid progenitor cell line (HUDEP-2), CD34+ adult human hematopoietic stem and progenitor cells (HSPCs), and immunophenotypic long-term hematopoietic stem cells (LT-HSCs). We find no overt in vitro increase in proliferation associated with an endogenous JAK2 V617F allele, but co-culture with wild type cells unmasks a competitive growth advantage provided by the mutation. Acquisition of the V617F allele also promotes terminal differentiation of erythroid progenitors, even in the absence of hematopoietic cytokine signaling. Taken together, these data are consistent with the gradually progressive manifestation of MPNs and reveals that endogenously acquired JAK2 V617F mutations may yield more subtle phenotypes as compared to transgenic overexpression models.


Asunto(s)
Edición Génica , Trastornos Mieloproliferativos/genética , Sistemas CRISPR-Cas , Línea Celular , Técnicas de Cocultivo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Janus Quinasa 2/genética
14.
Am J Hum Genet ; 108(4): 709-721, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33735615

RESUMEN

The fetal-to-adult hemoglobin switch is regulated in a developmental stage-specific manner and reactivation of fetal hemoglobin (HbF) has therapeutic implications for treatment of ß-thalassemia and sickle cell anemia, two major global health problems. Although significant progress has been made in our understanding of the molecular mechanism of the fetal-to-adult hemoglobin switch, the mechanism of epigenetic regulation of HbF silencing remains to be fully defined. Here, we performed whole-genome bisulfite sequencing and RNA sequencing analysis of the bone marrow-derived GYPA+ erythroid cells from ß-thalassemia-affected individuals with widely varying levels of HbF groups (HbF ≥ 95th percentile or HbF ≤ 5th percentile) to screen epigenetic modulators of HbF and phenotypic diversity of ß-thalassemia. We identified an ETS2 repressor factor encoded by ERF, whose promoter hypermethylation and mRNA downregulation are associated with high HbF levels in ß-thalassemia. We further observed that hypermethylation of the ERF promoter mediated by enrichment of DNMT3A leads to demethylation of γ-globin genes and attenuation of binding of ERF on the HBG promoter and eventually re-activation of HbF in ß-thalassemia. We demonstrated that ERF depletion markedly increased HbF production in human CD34+ erythroid progenitor cells, HUDEP-2 cell lines, and transplanted NCG-Kit-V831M mice. ERF represses γ-globin expression by directly binding to two consensus motifs regulating γ-globin gene expression. Importantly, ERF depletion did not affect maturation of erythroid cells. Identification of alterations in DNA methylation of ERF as a modulator of HbF synthesis opens up therapeutic targets for ß-hemoglobinopathies.


Asunto(s)
Epigénesis Genética , Perfilación de la Expresión Génica , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Talasemia beta/genética , gamma-Globinas/genética , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Línea Celular , Niño , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Femenino , Hemoglobina Fetal/genética , Edición Génica , Humanos , Masculino , Ratones , Regiones Promotoras Genéticas/genética , Reproducibilidad de los Resultados , Sulfitos , Secuenciación Completa del Genoma , Talasemia beta/patología
15.
Exp Hematol ; 97: 32-46.e35, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33675821

RESUMEN

Oxygen is a critical noncellular component of the bone marrow microenvironment that plays an important role in the development of hematopoietic cell lineages. In this study, we investigated the impact of low oxygen (hypoxia) on ex vivo myeloerythroid differentiation of human cord blood-derived CD34+ hematopoietic stem and progenitor cells. We characterized the culture conditions to demonstrate that low oxygen inhibits cell proliferation and causes a metabolic shift in the stem and progenitor populations. We found that hypoxia promotes erythroid differentiation by supporting the development of progenitor populations. Hypoxia also increases the megakaryoerythroid potential of the common myeloid progenitors and the erythroid potential of megakaryoerythroid progenitors and significantly accelerates maturation of erythroid cells. Specifically, we determined that hypoxia promotes the loss of CD71 and the appearance of the erythroid markers CD235a and CD239. Further, evaluation of erythroid populations revealed a hypoxia-induced increase in proerythroblasts and in enucleation of CD235a+ cells. These results reveal the extensive role of hypoxia at multiple steps during erythroid development. Overall, our work establishes a valuable model for further investigations into the relationship between erythroid progenitors and/or erythroblast populations and their hypoxic microenvironment.


Asunto(s)
Eritroblastos/citología , Células Eritroides/citología , Células Precursoras Eritroides/citología , Eritropoyesis , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Eritroblastos/metabolismo , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Humanos , Metaboloma
16.
J Cell Mol Med ; 25(5): 2377-2389, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33496386

RESUMEN

The exact molecular mechanism underlying erythroblast enucleation has been a fundamental biological question for decades. In this study, we found that miR-144/451 critically regulated erythroid differentiation and enucleation. We further identified CAP1, a G-actin-binding protein, as a direct target of miR-144/451 in these processes. During terminal erythropoiesis, CAP1 expression declines along with gradually increased miR-144/451 levels. Enforced CAP1 up-regulation inhibits the formation of contractile actin rings in erythroblasts and prevents their terminal differentiation and enucleation. Our findings reveal a negative regulatory role of CAP1 in miR-144/451-mediated erythropoiesis and thus shed light on how microRNAs fine-tune terminal erythroid development through regulating actin dynamics.


Asunto(s)
Diferenciación Celular/genética , Células Precursoras Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Interferencia de ARN , Serina Endopeptidasas/genética , Regiones no Traducidas 3' , Animales , Biomarcadores , Línea Celular , Células Cultivadas , Células Precursoras Eritroides/citología , Eritropoyesis/genética , Inmunofenotipificación , Ratones
17.
Blood Rev ; 46: 100740, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32798012

RESUMEN

A characteristic feature of terminal erythropoiesis in mammals is extrusion of the highly condensed nucleus out of the cytoplasm. Other vertebrates, including fish, reptiles, amphibians, and birds, undergo nuclear condensation but do not enucleate. Enucleation provides mammals evolutionary advantages by gaining extra space for hemoglobin and being more flexible to migrate through capillaries. Nascent reticulocytes further mature into red blood cells through membrane and proteome remodeling and organelle clearance. Over the past decade, novel molecular mechanisms and signaling pathways have been uncovered that play important roles in chromatin condensation, enucleation, and reticulocyte maturation. These advances not only increase understanding of the physiology of erythropoiesis, but also facilitate efforts in generating in vitro red blood cells for various translational application. In the present review, recent studies in epigenetic modification and release of histones during chromatin condensation are highlighted. New insights in enucleation, including protein sorting, vesicle trafficking, transcriptional regulation, noncoding RNA, cytoskeleton remodeling, erythroblastic islands, and cytokinesis, are summarized. Moreover, organelle clearance and proteolysis mediated by ubiquitin-proteasome degradation during reticulocytes maturation is also examined. Perspectives for future directions in this rapidly evolving research area are also provided.


Asunto(s)
Diferenciación Celular , Eritrocitos , Eritropoyesis/fisiología , Animales , Núcleo Celular , Cromatina/genética , Cromatina/metabolismo , Eritroblastos/citología , Eritroblastos/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Regulación de la Expresión Génica , Humanos , Reticulocitos/citología , Reticulocitos/metabolismo
18.
Blood Cells Mol Dis ; 87: 102533, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33352376

RESUMEN

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). The article has been retracted at the request of the editor. The journal was informed by Dr Xiangmin Xu and Dr Yongzhong Zhao that they were not involved in the study or research and that the article was submitted without their knowledge. As such this article represents a misuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process. All authors were informed of the article retraction however Dr Li and Dr Zeng did not respond to the enquiries.


Asunto(s)
Células Eritroides/citología , Células Precursoras Eritroides/citología , Eritropoyesis , Factores de Transcripción de Tipo Kruppel/genética , Globinas alfa/genética , Línea Celular , Epigénesis Genética , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional
19.
Mol Cell ; 81(2): 239-254.e8, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33301730

RESUMEN

Metazoan transcription factors typically regulate large numbers of genes. Here we identify via a CRISPR-Cas9 genetic screen ZNF410, a pentadactyl DNA-binding protein that in human erythroid cells directly activates only a single gene, the NuRD component CHD4. Specificity is conveyed by two highly evolutionarily conserved clusters of ZNF410 binding sites near the CHD4 gene with no counterparts elsewhere in the genome. Loss of ZNF410 in adult-type human erythroid cell culture systems and xenotransplantation settings diminishes CHD4 levels and derepresses the fetal hemoglobin genes. While previously known to be silenced by CHD4, the fetal globin genes are exposed here as among the most sensitive to reduced CHD4 levels.. In vitro DNA binding assays and crystallographic studies reveal the ZNF410-DNA binding mode. ZNF410 is a remarkably selective transcriptional activator in erythroid cells, and its perturbation might offer new opportunities for treatment of hemoglobinopathies.


Asunto(s)
ADN/genética , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Factores de Transcripción/genética , Animales , Sitios de Unión , Células COS , Sistemas CRISPR-Cas , Chlorocebus aethiops , ADN/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/trasplante , Sangre Fetal/citología , Sangre Fetal/metabolismo , Hemoglobina Fetal/metabolismo , Feto , Edición Génica , Células HEK293 , Xenoinjertos , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/química , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones , Modelos Moleculares , Células Madre Embrionarias de Ratones/citología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...