Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.185
Filter
1.
Blood ; 143(22): 2300-2313, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38447046

ABSTRACT

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.


Subject(s)
Chromatin , Erythropoiesis , GATA2 Transcription Factor , RNA, Long Noncoding , Humans , Erythropoiesis/genetics , RNA, Long Noncoding/genetics , Chromatin/metabolism , Chromatin/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Cell Differentiation/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/cytology
2.
Exp Hematol ; 106: 19-30, 2022 02.
Article in English | MEDLINE | ID: mdl-34879257

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Erythropoiesis , Animals , Cations, Divalent/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans
3.
Cell Rep ; 36(6): 109507, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380040

ABSTRACT

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.


Subject(s)
Cytoplasm/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Calibration , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Computer Simulation , Erythropoietin/pharmacology , Mice, Inbred BALB C , Models, Biological , Phosphorylation/drug effects , Signal Transduction/drug effects
4.
Genes (Basel) ; 12(8)2021 07 27.
Article in English | MEDLINE | ID: mdl-34440315

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Erythroid Precursor Cells/drug effects , Hydroxyurea/pharmacology , Nitric Oxide Synthase/metabolism , Animals , Erythroid Precursor Cells/cytology , Humans , K562 Cells , Mice , Nitric Oxide Donors/pharmacology
5.
Genes (Basel) ; 12(7)2021 06 30.
Article in English | MEDLINE | ID: mdl-34208866

ABSTRACT

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.


Subject(s)
Cell Differentiation , Cell Lineage , Chromatin Assembly and Disassembly , Erythroid Precursor Cells/cytology , Erythropoiesis , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Genome , Humans , Transcription Factors/genetics
6.
Am J Hematol ; 96(9): 1064-1076, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34021930

ABSTRACT

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.


Subject(s)
Erythroid Cells/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis , Antigens, CD/analysis , Antigens, CD34/analysis , Bone Marrow Cells/cytology , Cells, Cultured , Endoglin/analysis , Flow Cytometry/methods , Humans , Immunophenotyping/methods
7.
Exp Hematol ; 97: 32-46.e35, 2021 05.
Article in English | MEDLINE | ID: mdl-33675821

ABSTRACT

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.


Subject(s)
Erythroblasts/cytology , Erythroid Cells/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Erythroblasts/metabolism , Erythroid Cells/metabolism , Erythroid Precursor Cells/metabolism , Humans , Metabolome
8.
Am J Hum Genet ; 108(4): 709-721, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33735615

ABSTRACT

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.


Subject(s)
Epigenesis, Genetic , Gene Expression Profiling , Repressor Proteins/deficiency , Repressor Proteins/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics , Animals , Antigens, CD34/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Child , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Female , Fetal Hemoglobin/genetics , Gene Editing , Humans , Male , Mice , Promoter Regions, Genetic/genetics , Reproducibility of Results , Sulfites , Whole Genome Sequencing , beta-Thalassemia/pathology
9.
PLoS One ; 16(3): e0247858, 2021.
Article in English | MEDLINE | ID: mdl-33661998

ABSTRACT

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.


Subject(s)
Gene Editing , Myeloproliferative Disorders/genetics , CRISPR-Cas Systems , Cell Line , Coculture Techniques , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/genetics
10.
J Cell Mol Med ; 25(5): 2377-2389, 2021 03.
Article in English | MEDLINE | ID: mdl-33496386

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , Erythroid Precursor Cells/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/genetics , RNA Interference , Serine Endopeptidases/genetics , 3' Untranslated Regions , Animals , Biomarkers , Cell Line , Cells, Cultured , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Immunophenotyping , Mice
11.
Blood Rev ; 46: 100740, 2021 03.
Article in English | MEDLINE | ID: mdl-32798012

ABSTRACT

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.


Subject(s)
Cell Differentiation , Erythrocytes , Erythropoiesis/physiology , Animals , Cell Nucleus , Chromatin/genetics , Chromatin/metabolism , Erythroblasts/cytology , Erythroblasts/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Gene Expression Regulation , Humans , Reticulocytes/cytology , Reticulocytes/metabolism
12.
Blood Cells Mol Dis ; 87: 102533, 2021 03.
Article in English | MEDLINE | ID: mdl-33352376

ABSTRACT

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.


Subject(s)
Erythroid Cells/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis , Kruppel-Like Transcription Factors/genetics , alpha-Globins/genetics , Cell Line , Epigenesis, Genetic , Erythroid Cells/metabolism , Erythroid Precursor Cells/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Promoter Regions, Genetic , Transcriptional Activation
13.
Mol Cell ; 81(2): 239-254.e8, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33301730

ABSTRACT

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.


Subject(s)
DNA/genetics , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Transcription Factors/genetics , Animals , Binding Sites , COS Cells , CRISPR-Cas Systems , Chlorocebus aethiops , DNA/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/transplantation , Fetal Blood/cytology , Fetal Blood/metabolism , Fetal Hemoglobin/metabolism , Fetus , Gene Editing , HEK293 Cells , Heterografts , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Molecular , Mouse Embryonic Stem Cells/cytology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcriptional Activation
15.
Exp Hematol ; 89: 43-54.e2, 2020 09.
Article in English | MEDLINE | ID: mdl-32750404

ABSTRACT

Steady-state erythropoiesis generates new erythrocytes at a constant rate, and it has enormous productive capacity. This production is balanced by the removal of senescent erythrocytes by macrophages in the spleen and liver. Erythroid homeostasis is highly regulated to maintain sufficient erythrocytes for efficient oxygen delivery to the tissues, while avoiding viscosity problems associated with overproduction. However, there are times when this constant production of erythrocytes is inhibited or is inadequate; at these times, erythroid output is increased to compensate for the loss of production. In some cases, increased steady-state erythropoiesis can offset the loss of erythrocytes but, in response to inflammation caused by infection or tissue damage, steady-state erythropoiesis is inhibited. To maintain homeostasis under these conditions, an alternative stress erythropoiesis pathway is activated. Emerging data suggest that the bone morphogenetic protein 4 (BMP4)-dependent stress erythropoiesis pathway is integrated into the inflammatory response and generates a bolus of new erythrocytes that maintain homeostasis until steady-state erythropoiesis can resume. In this perspective, we define the mechanisms that generate new erythrocytes when steady-state erythropoiesis is impaired and discuss experimental models to study human stress erythropoiesis.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Erythrocytes/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Macrophages/cytology , Stress, Physiological/genetics , Animals , Bone Morphogenetic Protein 4/immunology , Cellular Senescence/immunology , Cytokines/genetics , Cytokines/immunology , Erythrocytes/immunology , Erythroid Precursor Cells/immunology , Erythropoiesis/immunology , Gene Expression Regulation , Humans , Inflammation , Liver/cytology , Liver/immunology , Macrophages/immunology , Mice , Models, Biological , Phagocytosis , Spleen/cytology , Spleen/immunology , Stress, Physiological/immunology
16.
Circ Res ; 127(10): 1221-1232, 2020 10 23.
Article in English | MEDLINE | ID: mdl-32791884

ABSTRACT

RATIONALE: Endothelial cells are thought to emerge de novo from the mesoderm to form the entire circulatory system. Recently, erythro-myeloid progenitors (EMPs) have been proposed to be another remarkable developmental origin for blood vessels in multiple organs, including the hindbrain, liver, lung, and heart, as demonstrated by lineage tracing studies using different genetic tools. These observations challenge the current consensus that intraembryonic vessels are thought to expand solely by the proliferation of preexisting endothelial cells. Resolution of this controversy over the developmental origin of endothelial cells is crucial for developing future therapeutics for vessel-dependent organ repair and regeneration. OBJECTIVE: To examine the contribution of EMPs to intraembryonic endothelial cells. METHODS AND RESULTS: We first used a transgenic mouse expressing a tamoxifen-inducible Mer-iCre fusion protein driven by the Csf1r (colony stimulating factor 1 receptor) promoter. Genetic lineage tracing based on Csf1r-Mer-iCre-Mer showed no contribution of EMPs to brain endothelial cells identified by several markers. We also generated a knock-in mouse line by inserting an internal ribosome entry site-iCre cassette into the 3' untranslated region of Csf1r gene to further investigate the cellular fates of EMPs. Similarly, we did not find any Csf1r-ires-iCre traced endothelial cells in brain, liver, lung, or heart in development either. Additionally, we found that Kit (KIT proto-oncogene receptor tyrosine kinase) was expressed not only in EMPs but also in embryonic hindbrain endothelial cells. Therefore, Kit promoter-driven recombinase, such as Kit-CreER, is a flawed tool for lineage tracing when examining the contribution of EMPs to hindbrain endothelial cells. We also traced CD45 (protein tyrosine phosphatase receptor type C; Ptprc)+ circulating EMPs and did not find any CD45 lineage-derived endothelial cells during development. CONCLUSIONS: Our study suggested that EMPs are not the origin of intraembryonic endothelial cells.


Subject(s)
Cell Lineage , Endothelial Cells/cytology , Erythroid Precursor Cells/cytology , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Fetal Heart/cytology , Liver/cytology , Liver/embryology , Lung/cytology , Lung/embryology , Macrophages/cytology , Mesoderm/cytology , Mice , Rhombencephalon/cytology , Rhombencephalon/embryology
17.
Exp Hematol ; 89: 1-12, 2020 09.
Article in English | MEDLINE | ID: mdl-32798645

ABSTRACT

I am deeply honored to receive the International Society for Experimental Hematology (ISEH) 2020 Donald Metcalf Lecture Award. Although I am not a physician and have had no formal training in hematology, I have had the privilege of working with some of the top hematologists in the world, beginning in 1970 when Dr. David Nathan was a sabbatical visitor in my laboratory and introduced me to hematological diseases.  And I take this award to be given not just to me but to an exceptional group of MD and PhD trainees and visitors in my laboratory who have cloned and characterized many proteins and RNAs important for red cell development and function. Many of these projects involved taking exceptionally large risks in developing and employing novel experimental technologies. Unsurprisingly, all of these trainees have gone on to become leaders in hematology and, more broadly, in molecular cell biology and molecular medicine. To illustrate some of the challenges we have faced and the technologies we had to develop, I have chosen several of our multiyear projects to describe in some detail: elucidating the regulation of translation of α- and ß-globin mRNAs and the defect in beta thalassemia in the 1970s; cloning the Epo receptor and several red cell membrane proteins in the 1980s and 1990s; and more recently, determining the function of many microRNAs and long noncoding RNAs in red cell development. I summarize how we are currently utilizing single-cell transcriptomics (scRNAseq) to understand how dividing transit-amplifying burst-forming unit erythroid progenitors balance the need for more progenitor cells with the need for terminally differentiated erythroid cells, and to identify drugs potentially useful in treating Epo-resistant anemias such as Diamond Blackfan anemia. I hope that the lessons I learned in managing these diverse fellows and projects, initially without having grants to support them, will be helpful to others who would like to undertake ambitious and important lines of research in hematology.


Subject(s)
Erythroid Precursor Cells/metabolism , Hematology/history , Molecular Biology/history , Receptors, Erythropoietin/history , beta-Thalassemia/genetics , Cloning, Molecular , Erythrocytes/metabolism , Erythrocytes/pathology , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Gene Expression , History, 20th Century , History, 21st Century , Humans , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
18.
Sci Rep ; 10(1): 11806, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678227

ABSTRACT

The molecular mechanisms involved in the terminal differentiation of erythroblasts have been elucidated by comparing enucleation and cell division. Although various similarities and differences between erythroblast enucleation and cytokinesis have been reported, the mechanisms that control enucleation remain unclear. We previously reported that dynein and microtubule-organizing centers mediated the polarization of nuclei in human erythroblasts. Moreover, the accumulation of F-actin was noted during the enucleation of erythroblasts. Therefore, during enucleation, upstream effectors in the signal transduction pathway regulating dynein or actin, such as cell division control protein 42 homolog (Cdc42), may be crucial. We herein investigated the effects of the Cdc42 inhibitor, CASIN, on cytokinesis and enucleation in colony-forming units-erythroid (CFU-Es) and mature erythroblasts (day 10). CASIN blocked the proliferation of CFU-Es and their enucleation in a dose-dependent manner. Dynein adopted an island-like distribution in the cytoplasm of non-treated CFU-Es, but was concentrated near the nucleus as a dot and co-localized with γ-tubulin in CASIN-treated cells. CASIN blocked the accumulation of F-actin in CFU-Es and day 10 cells. These results demonstrated that Cdc42 plays an important role in cytokinesis, nuclear polarization and nuclear extrusion through a relationship with dynein and actin filament organization during the terminal differentiation of erythroblasts.


Subject(s)
Actomyosin/metabolism , Cell Differentiation , Erythroblasts/cytology , Erythroblasts/metabolism , cdc42 GTP-Binding Protein/genetics , Biomarkers , Cell Differentiation/genetics , Cell Nucleus/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Fluorescent Antibody Technique , Gene Expression , Humans , Immunohistochemistry , cdc42 GTP-Binding Protein/metabolism
19.
Blood ; 136(9): 1044-1054, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32548608

ABSTRACT

Erythropoietin (EPO) provides the major survival signal to maturing erythroid precursors (EPs) and is essential for terminal erythropoiesis. Nonetheless, progenitor cells can irreversibly commit to an erythroid fate well before EPO acts, risking inefficiency if these progenitors are unneeded to maintain red blood cell (RBC) counts. We identified a new modular organization of erythropoiesis and, for the first time, demonstrate that the pre-EPO module is coupled to late EPO-dependent erythropoiesis by megakaryocyte (Mk) signals. Disrupting megakaryocytic transforming growth factor ß1 (Tgfb1) disorganized hematopoiesis by expanding the pre-EPO pool of progenitor cells and consequently triggering significant apoptosis of EPO-dependent EPs. Similarly, pharmacologic blockade of TGFß signaling in normal mice boosted the pre-EPO module, leading to apoptosis of EPO-sensitive EPs. Subsequent treatment with low-dose EPO triggered robust RBC production in both models. This work reveals modular regulation of erythropoiesis and offers a new strategy for overcoming chronic anemias.


Subject(s)
Erythroid Precursor Cells/cytology , Erythropoiesis/physiology , Megakaryocytes/cytology , Transforming Growth Factor beta1/physiology , Animals , Apoptosis/drug effects , Bone Marrow/pathology , Erythroid Precursor Cells/metabolism , Erythropoietin/pharmacology , Gene Knockout Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Megakaryocyte-Erythroid Progenitor Cells/cytology , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiation Chimera , Recombinant Proteins/pharmacology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology
20.
Exp Hematol ; 86: 53-66.e1, 2020 06.
Article in English | MEDLINE | ID: mdl-32450207

ABSTRACT

Red blood cells are generated daily to replenish dying cells and maintain erythrocyte homeostasis. Erythropoiesis is driven by erythropoietin and supported by specialized red pulp macrophages that facilitate enucleation. Here we show that the leukocyte-specific tyrosine phosphatase CD45 is downregulated in late erythroid development, yet it regulates the CD71+TER119+ progenitor pool, which includes the Pro E, Ery A, and Ery B populations. The CD71+TER119+ progenitors are a major splenic population in neonates required for extramedullary erythropoiesis, to meet the high demand for red blood cells during growth. This population decreases as the mice mature, but this was not the case in CD45-deficient mice, which maintained a high level of these progenitors in the spleen into adulthood. Despite these increased erythroid progenitors, CD45-deficient mice had normal numbers of mature red blood cells. This was attributed to the increased proliferation of the Pro E and Ery A populations and the increased apoptosis of the CD71+TER119+ population, as well as an increased turnover of circulating red blood cells. The expansion of the CD71+TER119+ population in the absence of CD45 was attributed to increased numbers of red pulp macrophages producing erythropoietin in the spleen. Thus, CD45 regulates extramedullary erythropoiesis in the spleen.


Subject(s)
Antigens, CD/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis , Hematopoiesis, Extramedullary , Leukocyte Common Antigens/metabolism , Receptors, Transferrin/metabolism , Animals , Antigens, CD/genetics , Erythroid Precursor Cells/cytology , Leukocyte Common Antigens/genetics , Mice , Mice, Knockout , Receptors, Transferrin/genetics , Spleen/cytology , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...