Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.288
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(12): 2042-2052, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37919525

ABSTRACT

Tumor-derived factors are thought to regulate thrombocytosis and erythrocytopenia in individuals with cancer; however, such factors have not yet been identified. Here we show that tumor cell-released kynurenine (Kyn) biases megakaryocytic-erythroid progenitor cell (MEP) differentiation into megakaryocytes in individuals with cancer by activating the aryl hydrocarbon receptor-Runt-related transcription factor 1 (AhR-RUNX1) axis. During tumor growth, large amounts of Kyn from tumor cells are released into the periphery, where they are taken up by MEPs via the transporter SLC7A8. In the cytosol, Kyn binds to and activates AhR, leading to its translocation into the nucleus where AhR transactivates RUNX1, thus regulating MEP differentiation into megakaryocytes. In addition, activated AhR upregulates SLC7A8 in MEPs to induce positive feedback. Importantly, Kyn-AhR-RUNX1-regulated MEP differentiation was demonstrated in both humanized mice and individuals with cancer, providing potential strategies for the prevention of thrombocytosis and erythrocytopenia.


Subject(s)
Neoplasms , Thrombocytosis , Animals , Mice , Kynurenine/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Megakaryocytes/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Erythroid Precursor Cells/metabolism , Cell Differentiation/physiology , Neoplasms/metabolism , Thrombocytosis/metabolism , Bias
2.
Nat Immunol ; 23(7): 1109-1120, 2022 07.
Article in English | MEDLINE | ID: mdl-35761081

ABSTRACT

Nonimmune cells can have immunomodulatory roles that contribute to healthy development. However, the molecular and cellular mechanisms underlying the immunomodulatory functions of erythroid cells during human ontogenesis remain elusive. Here, integrated, single-cell transcriptomic studies of erythroid cells from the human yolk sac, fetal liver, preterm umbilical cord blood (UCB), term UCB and adult bone marrow (BM) identified classical and immune subsets of erythroid precursors with divergent differentiation trajectories. Immune-erythroid cells were present from the yolk sac to the adult BM throughout human ontogenesis but failed to be generated in vitro from human embryonic stem cells. Compared with classical-erythroid precursors, these immune-erythroid cells possessed dual erythroid and immune regulatory networks, showed immunomodulatory functions and interacted more frequently with various innate and adaptive immune cells. Our findings provide important insights into the nature of immune-erythroid cells and their roles during development and diseases.


Subject(s)
Erythroid Precursor Cells , Transcriptome , Adult , Cell Differentiation/genetics , Erythroid Cells , Fetal Blood , Humans , Infant, Newborn , Yolk Sac
3.
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
4.
Cell ; 149(1): 63-74, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464323

ABSTRACT

Osteoblasts are an important component of the hematopoietic microenvironment in bone. However, the mechanisms by which osteoblasts control hematopoiesis remain unknown. We show that augmented HIF signaling in osteoprogenitors results in HSC niche expansion associated with selective expansion of the erythroid lineage. Increased red blood cell production occurred in an EPO-dependent manner with increased EPO expression in bone and suppressed EPO expression in the kidney. In contrast, inactivation of HIF in osteoprogenitors reduced EPO expression in bone. Importantly, augmented HIF activity in osteoprogenitors protected mice from stress-induced anemia. Pharmacologic or genetic inhibition of prolyl hydroxylases1/2/3 in osteoprogenitors elevated EPO expression in bone and increased hematocrit. These data reveal an unexpected role for osteoblasts in the production of EPO and modulation of erythropoiesis. Furthermore, these studies demonstrate a molecular role for osteoblastic PHD/VHL/HIF signaling that can be targeted to elevate both HSCs and erythroid progenitors in the local hematopoietic microenvironment.


Subject(s)
Erythropoiesis , Erythropoietin/metabolism , Osteoblasts/metabolism , Signal Transduction , Anemia/prevention & control , Animals , Erythroid Precursor Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/metabolism , Mice , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
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
6.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Article in English | MEDLINE | ID: mdl-38605277

ABSTRACT

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Erythropoiesis , Mice, Knockout , Microcephaly , Tumor Suppressor Protein p53 , Animals , Mice , Anemia, Macrocytic/genetics , Anemia, Macrocytic/pathology , Anemia, Macrocytic/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Microcephaly/genetics , Microcephaly/pathology , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Curr Opin Hematol ; 31(3): 96-103, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38415760

ABSTRACT

PURPOSE OF REVIEW: Recent work reveals that cell cycle duration and structure are remodeled in lock-step with distinct stages of erythroid differentiation. These cell cycle features have regulatory roles in differentiation, beyond the generic function of increasing cell number. RECENT FINDINGS: Developmental progression through the early erythroid progenitor stage (known as colony-forming-erythroid, or 'CFU-e') is characterized by gradual shortening of G1 phase of the cycle. This process culminates in a key transcriptional switch to erythroid terminal differentiation (ETD) that is synchronized with, and dependent on, S phase progression. Further, the CFU-e/ETD switch takes place during an unusually short S phase, part of an exceptionally short cell cycle that is characterized by globally fast replication fork speeds. Cell cycle and S phase speed can alter developmental events during erythroid differentiation, through pathways that are targeted by glucocorticoid and erythropoietin signaling during the erythroid stress response. SUMMARY: There is close inter-dependence between cell cycle structure and duration, S phase and replication fork speeds, and erythroid differentiation stage. Further, modulation of cell cycle structure and speed cycle impacts developmental progression and cell fate decisions during erythroid differentiation. These pathways may offer novel mechanistic insights and potential therapeutic targets.


Subject(s)
Erythroid Precursor Cells , Signal Transduction , Humans , Cell Cycle/physiology , Cell Differentiation , S Phase , Erythropoiesis/physiology
8.
Int J Cancer ; 155(11): 1928-1938, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39039820

ABSTRACT

Immunotherapy, especially immune checkpoint blockade therapy, represents a major milestone in the history of cancer therapy. However, the current response rate to immunotherapy among cancer patients must be improved; thus, new strategies for sensitizing patients to immunotherapy are urgently needed. Erythroid progenitor cells (EPCs), a population of immature erythroid cells, exert potent immunosuppressive functions. As a newly recognized immunosuppressive population, EPCs have not yet been effectively targeted. In this review, we summarize the immunoregulatory mechanisms of EPCs, especially for CD45+ EPCs. Moreover, in view of the regulatory effects of EPCs on the tumor microenvironment, we propose the concept of EPC-immunity, present existing strategies for targeting EPCs, and discuss the challenges encountered in both basic research and clinical applications. In particular, the impact of existing cancer treatments on EPCs is discussed, laying the foundation for combination therapies. The aim of this review is to provide new avenues for improving the efficacy of cancer immunotherapy by targeting EPCs.


Subject(s)
Erythroid Precursor Cells , Immunotherapy , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/pathology , Immunotherapy/methods , Tumor Microenvironment/immunology , Erythroid Precursor Cells/immunology , Animals , Leukocyte Common Antigens/metabolism
9.
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
10.
Blood Cells Mol Dis ; 106: 102829, 2024 May.
Article in English | MEDLINE | ID: mdl-38278056

ABSTRACT

BACKGROUND: Erythropoiesis is a complex developmental process in which a hematopoietic stem cell undergoes serial divisions and differentiates through well-defined stages to give rise to red blood cells. Over the last decades, several protocols have been developed to perform ex vivo erythroid differentiation, allowing investigation into erythropoiesis and red cell production in health and disease. RESULTS: In the current study, we compared the two commonly used protocols by assessing the differentiation kinetics, synchronisation, and cellular yield, using molecular and cellular approaches. Peripheral blood CD34+ cells were cultured in a two-phase (2P) or a four-phase (4P) liquid culture (LC) and monitored for 20 days. Both protocols could recapitulate all stages of erythropoiesis and generate reticulocytes, although to different extents. Higher proliferation and viability rates were achieved in the 4P-LC, with a higher degree of terminal differentiation and enucleation, associated with higher levels of the erythroid-specific transcription factors GATA-1, KLF-1, and TAL-1. Although the 2P-LC protocol was less efficient regarding terminal erythroid differentiation and maturation, it showed a higher yield of erythroid progenitors in the erythropoietin (EPO)-free expansion phase. CONCLUSIONS: We provide data supporting the use of one protocol or the other to study the biological processes occurring in the early or late stages of erythroid differentiation, depending on the physiological process or pathological defect under investigation in a given study.


Subject(s)
Erythropoietin , Hematopoietic Stem Cells , Humans , Cell Differentiation , Erythrocytes , Erythropoiesis/physiology , Antigens, CD34 , Erythroid Precursor Cells
11.
Blood Cells Mol Dis ; 106: 102838, 2024 May.
Article in English | MEDLINE | ID: mdl-38413287

ABSTRACT

Diamond-Blackfan anemia (DBA) was the first ribosomopathy described in humans. DBA is a congenital hypoplastic anemia, characterized by macrocytic aregenerative anemia, manifesting by differentiation blockage between the BFU-e/CFU-e developmental erythroid progenitor stages. In 50 % of the DBA cases, various malformations are noted. Strikingly, for a hematological disease with a relative erythroid tropism, DBA is due to ribosomal haploinsufficiency in 24 different ribosomal protein (RP) genes. A few other genes have been described in DBA-like disorders, but they do not fit into the classical DBA phenotype (Sankaran et al., 2012; van Dooijeweert et al., 2022; Toki et al., 2018; Kim et al., 2017 [1-4]). Haploinsufficiency in a RP gene leads to defective ribosomal RNA (rRNA) maturation, which is a hallmark of DBA. However, the mechanistic understandings of the erythroid tropism defect in DBA are still to be fully defined. Erythroid defect in DBA has been recently been linked in a non-exclusive manner to a number of mechanisms that include: 1) a defect in translation, in particular for the GATA1 erythroid gene; 2) a deficit of HSP70, the GATA1 chaperone, and 3) free heme toxicity. In addition, p53 activation in response to ribosomal stress is involved in DBA pathophysiology. The DBA phenotype may thus result from the combined contributions of various actors, which may explain the heterogenous phenotypes observed in DBA patients, even within the same family.


Subject(s)
Anemia, Diamond-Blackfan , Anemia, Dyserythropoietic, Congenital , Anemia, Macrocytic , Humans , Anemia, Diamond-Blackfan/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Erythroid Precursor Cells/metabolism , Mutation
12.
Blood ; 140(22): 2371-2384, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36054916

ABSTRACT

We found that in regenerative erythropoiesis, the erythroid progenitor landscape is reshaped, and a previously undescribed progenitor population with colony-forming unit-erythroid (CFU-E) activity (stress CFU-E [sCFU-E]) is expanded markedly to restore the erythron. sCFU-E cells are targets of erythropoietin (Epo), and sCFU-E expansion requires signaling from the Epo receptor (EpoR) cytoplasmic tyrosines. Molecularly, Epo promotes sCFU-E expansion via JAK2- and STAT5-dependent expression of IRS2, thus engaging the progrowth signaling from the IGF1 receptor (IGF1R). Inhibition of IGF1R and IRS2 signaling impairs sCFU-E cell growth, whereas exogenous IRS2 expression rescues cell growth in sCFU-E expressing truncated EpoR-lacking cytoplasmic tyrosines. This sCFU-E pathway is the major pathway involved in erythrocytosis driven by the oncogenic JAK2 mutant JAK2(V617F) in myeloproliferative neoplasm. Inability to expand sCFU-E cells by truncated EpoR protects against JAK2(V617F)-driven erythrocytosis. In samples from patients with myeloproliferative neoplasm, the number of sCFU-E-like cells increases, and inhibition of IGR1R and IRS2 signaling blocks Epo-hypersensitive erythroid cell colony formation. In summary, we identified a new stress-specific erythroid progenitor cell population that links regenerative erythropoiesis to pathogenic erythrocytosis.


Subject(s)
Erythropoietin , Myeloproliferative Disorders , Neoplasms , Polycythemia , Humans , Erythropoiesis/physiology , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Polycythemia/metabolism , Erythropoietin/metabolism , Myeloproliferative Disorders/metabolism , Erythroid Precursor Cells/metabolism , Neoplasms/metabolism , Receptor, IGF Type 1/metabolism
13.
Eur J Haematol ; 113(4): 416-425, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38853593

ABSTRACT

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.


Subject(s)
AMP-Activated Protein Kinases , Erythroid Precursor Cells , Galectins , TOR Serine-Threonine Kinases , Animals , Galectins/metabolism , Galectins/genetics , Mice , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/cytology , Mice, Inbred C57BL , Gene Expression , Immunophenotyping , Biomarkers , Bone Marrow Cells/metabolism
14.
Am J Hematol ; 99(1): 99-112, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37929634

ABSTRACT

Human erythropoiesis is a complex process leading to the production of 2.5 million red blood cells per second. Following commitment of hematopoietic stem cells to the erythroid lineage, this process can be divided into three distinct stages: erythroid progenitor differentiation, terminal erythropoiesis, and reticulocyte maturation. We recently resolved the heterogeneity of erythroid progenitors into four different subpopulations termed EP1-EP4. Here, we characterized the growth factor(s) responsiveness of these four progenitor populations in terms of proliferation and differentiation. Using mass spectrometry-based proteomics on sorted erythroid progenitors, we quantified the absolute expression of ~5500 proteins from EP1 to EP4. Further functional analyses highlighted dynamic changes in cell cycle in these populations with an acceleration of the cell cycle during erythroid progenitor differentiation. The finding that E2F4 expression was increased from EP1 to EP4 is consistent with the noted changes in cell cycle. Finally, our proteomic data suggest that the protein machinery necessary for both oxidative phosphorylation and glycolysis is present in these progenitor cells. Together, our data provide comprehensive insights into growth factor-dependence of erythroid progenitor proliferation and the proteome of four distinct populations of human erythroid progenitors which will be a useful framework for the study of erythroid disorders.


Subject(s)
Hematopoietic Stem Cells , Proteomics , Humans , Cell Differentiation , Cell Cycle , Erythropoiesis , Metabolic Networks and Pathways , Intercellular Signaling Peptides and Proteins/metabolism , Erythroid Precursor Cells
15.
Nature ; 555(7694): 54-60, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29466336

ABSTRACT

The formation of red blood cells begins with the differentiation of multipotent haematopoietic progenitors. Reconstructing the steps of this differentiation represents a general challenge in stem-cell biology. Here we used single-cell transcriptomics, fate assays and a theory that allows the prediction of cell fates from population snapshots to demonstrate that mouse haematopoietic progenitors differentiate through a continuous, hierarchical structure into seven blood lineages. We uncovered coupling between the erythroid and the basophil or mast cell fates, a global haematopoietic response to erythroid stress and novel growth factor receptors that regulate erythropoiesis. We defined a flow cytometry sorting strategy to purify early stages of erythroid differentiation, completely isolating classically defined burst-forming and colony-forming progenitors. We also found that the cell cycle is progressively remodelled during erythroid development and during a sharp transcriptional switch that ends the colony-forming progenitor stage and activates terminal differentiation. Our work showcases the utility of linking transcriptomic data to predictive fate models, and provides insights into lineage development in vivo.


Subject(s)
Erythrocytes/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis , Animals , Basophils/cytology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Lineage/drug effects , Cell Lineage/genetics , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Female , Flow Cytometry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mast Cells/cytology , Mice , Proto-Oncogene Proteins c-kit/metabolism , RNA, Small Cytoplasmic/analysis , RNA, Small Cytoplasmic/genetics , Single-Cell Analysis , Transcriptome
16.
Rinsho Ketsueki ; 65(3): 183-187, 2024.
Article in Japanese | MEDLINE | ID: mdl-38569864

ABSTRACT

The transcription factor GATA-1 is essential for erythroid differentiation. Recently, FAM210B, which encodes a mitochondrial inner membrane protein, has been identified as a novel target of GATA-1. To clarify the role of FAM210B, we depleted endogenous FAM210B in human iPS-derived erythroid progenitor (HiDEP-1) cells, and found that erythroid differentiation was more pronounced in the FAM210B depleted cells. Comprehensive metabolite analysis revealed a decline in mitochondrial function accompanied by increased lactate production, indicative of anaerobic glycolysis. Mass spectrometry revealed that FAM210B could interact with multiple subunits of mitochondrial ATP synthases, such as subunit alpha (ATP5A) and beta (ATP5B). Our results suggested that FAM210B contributes prominently to erythroid differentiation by regulating mitochondrial energy metabolism. This review will discuss the potential association between mitochondrial metabolism and erythropoiesis.


Subject(s)
GATA1 Transcription Factor , Mitochondria , Humans , Mitochondria/metabolism , Erythroid Precursor Cells/metabolism , Cell Differentiation/physiology , Erythropoiesis/physiology
17.
Curr Opin Hematol ; 30(4): 137-143, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37052294

ABSTRACT

PURPOSE OF REVIEW: It is well described that tumor-directed aberrant myelopoiesis contributes to the generation of various myeloid populations with tumor-promoting properties. A growing number of recent studies have revealed the importance of the previously unappreciated roles of erythroid progenitor cells (EPCs) in the context of cancer, bringing the updated concept that altered erythropoiesis also facilitates tumor growth and progression. Better characterization of EPCs may provide attractive therapeutic opportunities. RECENT FINDINGS: EPCs represent a heterogeneous population. They exhibit crucial pro-tumor activities by secreting growth factors and modulating the immune response. Cancers induce potent EPC expansion and suppress their differentiation. Recent single-cell transcriptome and lineage tracking analyses have provided novel insight that tumor-induced EPCs are able to be transdifferentiated into immunosuppressive myeloid cells to limit T-cell function and immunotherapy. Therapeutic strategies targeting key factors of EPC-driven immunosuppression, reducing the amount of EPCs, and promoting EPC differentiation and maturation have been extensively investigated. SUMMARY: This review summarizes the current state of knowledge as to the fascinating biology of EPCs, highlights mechanisms by which they exert the tumor promoting activities, as well as the perspectives on future directions and strategies to target these cells for potential therapeutic benefit.


Subject(s)
Erythroid Precursor Cells , Neoplasms , Humans , Cell Differentiation , Neoplasms/therapy
18.
J Virol ; 96(2): e0132621, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34669461

ABSTRACT

Parvovirus B19 (B19V) infection can cause hematological disorders and fetal hydrops during pregnancy. Currently, no antivirals or vaccines are available for the treatment or prevention of B19V infection. To identify novel small-molecule antivirals against B19V replication, we developed a high-throughput screening (HTS) assay, which is based on an in vitro nicking assay using recombinant N-terminal amino acids 1 to 176 of the viral large nonstructural protein (NS1N) and a fluorescently labeled DNA probe (OriQ) that spans the nicking site of the viral DNA replication origin. We collectively screened 17,040 compounds and identified 2,178 (12.78%) hits that possess >10% inhibition of the NS1 nicking activity, among which 84 hits were confirmed to inhibit nicking in a dose-dependent manner. Using ex vivo-expanded primary human erythroid progenitor cells (EPCs) infected by B19V, we validated 24 compounds that demonstrated >50% in vivo inhibition of B19V infection at 10 µM, which can be categorized into 7 structure scaffolds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, the top 4 compounds were chosen to examine their inhibitions of B19V infection in EPCs at two times of the 90% maximal effective concentration (EC90). A purine derivative (P7) demonstrated an antiviral effect (EC50 = 1.46 µM) without prominent cytotoxicity (CC50 = 71.8 µM) in EPCs and exhibited 92% inhibition of B19V infection in EPCs at 3.32 µM, which can be used as the lead compound in future studies for the treatment of B19V infection-caused hematological disorders. IMPORTANCE B19V encodes a large nonstructural protein, NS1. Its N-terminal domain (NS1N) consisting of amino acids 1 to 176 binds to viral DNA and serves as an endonuclease to nick the viral DNA replication origins, which is a pivotal step in rolling-hairpin-dependent B19V DNA replication. For high-throughput screening (HTS) of anti-B19V antivirals, we miniaturized a fluorescence-based in vitro nicking assay, which employs a fluorophore-labeled probe spanning the terminal resolution site (trs) and the NS1N protein, into a 384-well-plate format. The HTS assay showed high reliability and capability in screening 17,040 compounds. Based on the therapeutic index (half-maximal cytotoxic concentration [CC50]/half-maximal effective concentration [EC50] ratio) in EPCs, a purine derivative demonstrated an antiviral effect of 92% inhibition of B19V infection in EPCs at 3.32 µM (two times the EC90). Our study demonstrated a robust HTS assay for screening antivirals against B19V infection.


Subject(s)
Antiviral Agents/pharmacology , Erythroid Precursor Cells/virology , High-Throughput Screening Assays/methods , Parvovirus B19, Human/drug effects , Antiviral Agents/chemistry , Cell Survival/drug effects , DNA Replication/drug effects , DNA, Viral/metabolism , Erythroid Precursor Cells/drug effects , Fluorescent Dyes , Humans , Parvovirus B19, Human/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Origin , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
19.
Transfusion ; 63(6): 1122-1128, 2023 06.
Article in English | MEDLINE | ID: mdl-37154531

ABSTRACT

BACKGROUND: The generation of immortalized erythroid progenitor cell lines capable of producing enough red blood cells (RBCs) for blood transfusion typically requires the overexpression of oncogenes in stem cells or progenitor cells to permanently proliferate immature cells. It is essential that any live oncogene-expressing cells are eliminated from the final RBC products for clinical use. STUDY DESIGN AND METHODS: It is believed that safety issues may be resolved by using a leukoreduction filter or by irradiating the final products, as is conventionally done in blood banks; however, this has never been proven to be effective. Therefore, to investigate whether immortalized erythroblasts can be completely removed using γ-ray irradiation, we irradiated the erythroblast cell line, HiDEP, and the erythroleukemic cell line, K562 that overexpress HPV16 E6/E7. We then analyzed the extent of cell death using flow cytometry and polymerase chain reaction (PCR). The cells were also subjected to leukoreduction filters. RESULTS: Using γ-ray irradiation at 25 Gy, 90.4% of HiDEP cells, 91.6% of K562-HPV16 E6/E7 cells, and 93.5% of non-transduced K562 cells were dead. In addition, 5.58 × 107 HiDEP cells were passed through a leukoreduction filter, and 38 intact cells were harvested, revealing a filter removal efficiency of 99.9999%. However, both intact cells and oncogene DNA were still detected. DISCUSSION: Irradiation cannot induce total cell death of oncogene-expressing erythroblasts and leukocyte filter efficiency is not 100%. Therefore, our findings imply that for clinical applications, safer methods should be developed to completely remove residual nucleated cells from cell line-derived RBC products.


Subject(s)
Erythrocytes , Erythroid Cells , Humans , Erythrocytes/metabolism , Erythroid Precursor Cells , K562 Cells , Flow Cytometry
20.
Gastric Cancer ; 26(6): 918-933, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37676622

ABSTRACT

BACKGROUND AND AIMS: Specific mechanisms of lymph node (LN) metastasis in early-stage gastric cancer (GC) have not been elucidated. The role of anemia, a vital clinical feature of GC, in LN metastasis is also unclear. Since the number of erythroid progenitor cells (EPCs) is increased in chronic anemia, we investigated its association with LN metastasis in GC. METHODS: Flow cytometry and immunofluorescence analyses were performed to sort and study EPCs from the circulation and tumors of patients with stage I-III GC. The effect of these EPCs on the activation of T and B cells and on the functions of lymphatic endothelial cells (LECs) was determined, and their ability to promote LN metastasis was evaluated using a footpad-popliteal LN metastasis model based on two human adenocarcinoma GC cell lines in nude mice. The prognostic value of EPCs was also analyzed. RESULTS: The proportion of CD45- EPCs was higher in the mononuclear cells in the circulation, tumors, and LNs of GC patients with LN metastasis (N+) than in those of GC patients without LN metastasis (N0). In N+ patients, CD45- EPCs were more abundant in metastatic LNs than in non-metastatic LNs. Lymphatic vessel endothelial hyaluronan receptor 1 immunoreactivity in tumors revealed that CD45- EPCs were positively associated with nodal stages and lymph vessel density. Furthermore, CD45- EPCs increased LEC proliferation and migration through their S100A8/A9 heterodimer-induced hybrid epithelial/mesenchymal (E/M) state; however, they did not influence the invasion and tubulogenesis of LECs or T and B cell proliferation. CD45- EPCs promoted LN metastasis in vivo; the S100A8/A9 heterodimer mimicked this phenomenon. Finally, CD45- EPCs predicted the overall and disease-free survival of stage I-III GC patients after radical resection. CONCLUSIONS: The CD45- EPCs accumulated in GC tissues and metastatic LNs and promoted LN metastasis via the S100A8/9-induced hybrid E/M state of LECs, which was the specific mechanism of LN metastasis in the early stages of GC.


Subject(s)
Anemia , Stomach Neoplasms , Mice , Animals , Humans , Lymphatic Metastasis/pathology , Stomach Neoplasms/pathology , Endothelial Cells/metabolism , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Mice, Nude , Lymph Nodes/pathology , Anemia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL