Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.084
Filtrar
1.
Rinsho Ketsueki ; 65(3): 183-187, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-38569864

RESUMO

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.


Assuntos
Fator de Transcrição GATA1 , Mitocôndrias , Humanos , Mitocôndrias/metabolismo , Células Precursoras Eritroides/metabolismo , Diferenciação Celular/fisiologia , Eritropoese/fisiologia
2.
Blood Cells Mol Dis ; 106: 102838, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38413287

RESUMO

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.


Assuntos
Anemia de Diamond-Blackfan , Anemia Diseritropoética Congênita , Anemia Macrocítica , Humanos , Anemia de Diamond-Blackfan/genética , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Células Precursoras Eritroides/metabolismo , Mutação
3.
Nat Immunol ; 24(12): 2042-2052, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37919525

RESUMO

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.


Assuntos
Neoplasias , Trombocitose , Animais , Camundongos , Cinurenina/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Megacariócitos/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células Precursoras Eritroides/metabolismo , Diferenciação Celular/fisiologia , Neoplasias/metabolismo , Trombocitose/metabolismo , Viés
4.
Gastric Cancer ; 26(6): 918-933, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37676622

RESUMO

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.


Assuntos
Anemia , Neoplasias Gástricas , Camundongos , Animais , Humanos , Metástase Linfática/patologia , Neoplasias Gástricas/patologia , Células Endoteliais/metabolismo , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Camundongos Nus , Linfonodos/patologia , Anemia/patologia
5.
Rinsho Ketsueki ; 64(6): 482-488, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37407471

RESUMO

Large-scale in vitro red blood cell (RBC) production has been attempted in recent years. Potential cell sources for RBC production include hematopoietic stem/progenitor cells, pluripotent stem cells, and immortalized erythroid progenitor cell lines, which can induce enucleated RBCs with characteristics such as oxygen-carrying capacity and deformability. A phase I clinical study of cultured RBCs produced from hematopoietic stem/progenitor cells has revealed a similar in vivo half-life between cultured and native RBCs. Thus, the application of cultured RBCs in blood transfusion is gradually advancing. However, a single transfusion requires a large number of cells, unlike other cell therapies. Therefore, developing a method to mass-produce RBCs from a small culture volume at a low cost is important in the future. This review summarizes the current status and prospects concerning in vitro RBC production using each cell source, which can improve future transfusion medicine.


Assuntos
Medicina Transfusional , Humanos , Eritrócitos , Células Precursoras Eritroides/metabolismo , Eritropoese , Células-Tronco Hematopoéticas/metabolismo , Diferenciação Celular
6.
Cells ; 12(9)2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37174702

RESUMO

Erythropoiesis is a highly regulated process and undergoes several genotypic and phenotypic changes during differentiation. The phenotypic changes can be evaluated using a combination of cell surface markers expressed at different cellular stages of erythropoiesis using FACS. However, limited studies are available on the in-depth phenotypic characterization of progenitors from human adult hematopoietic stem and progenitor cells (HSPCs) to red blood cells. Therefore, using a set of designed marker panels, in the current study we have kinetically characterized the hematopoietic, erythroid progenitors, and terminally differentiated erythroblasts ex vivo. Furthermore, the progenitor stages were explored for expression of CD117, CD31, CD41a, CD133, and CD45, along with known key markers CD36, CD71, CD105, and GPA. Additionally, we used these marker panels to study the stage-specific phenotypic changes regulated by the epigenetic regulator; Nuclear receptor binding SET Domain protein 1 (NSD1) during erythropoiesis and to study ineffective erythropoiesis in myelodysplastic syndrome (MDS) and pure red cell aplasia (PRCA) patients. Our immunophenotyping strategy can be used to sort and study erythroid-primed hematopoietic and erythroid precursors at specified time points and to study diseases resulting from erythroid dyspoiesis. Overall, the current study explores the in-depth kinetics of phenotypic changes occurring during human erythropoiesis and applies this strategy to study normal and defective erythropoiesis.


Assuntos
Células Precursoras Eritroides , Eritropoese , Adulto , Humanos , Células Precursoras Eritroides/metabolismo , Imunofenotipagem , Eritroblastos/metabolismo , Diferenciação Celular
7.
Exp Hematol ; 123: 1-17, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37172755

RESUMO

Erythropoiesis, the development of erythrocytes from hematopoietic stem cells, occurs through four phases: erythroid progenitor (EP) development, early erythropoiesis, terminal erythroid differentiation (TED), and maturation. According to the classical model that is based on immunophenotypic profiles of cell populations, each of these phases comprises multiple differentiation states that arise in a hierarchical manner. After segregation of lymphoid potential, erythroid priming begins during progenitor development and progresses through progenitor cell types that have multilineage potential. Complete separation of the erythroid lineage is achieved during early erythropoiesis with the formation of unipotent EPs: burst-forming unit-erythroid and colony-forming unit-erythroid. These erythroid-committed progenitors undergo TED and maturation, which involves expulsion of the nucleus and remodeling to form functional biconcave, hemoglobin-filled erythrocytes. In the last decade or so, many studies employing advanced techniques such as single-cell RNA-sequencing (scRNA-seq) as well as the conventional methods, including colony-forming cell assays and immunophenotyping, have revealed heterogeneity within the stem, progenitor, and erythroblast stages, and uncovered alternate paths for segregation of erythroid lineage potential. In this review, we provide an in-depth account of immunophenotypic profiles of all cell types within erythropoiesis, highlight studies that demonstrate heterogeneous erythroid stages, and describe deviations to the classical model of erythropoiesis. Overall, although scRNA-seq approaches have provided new insights, flow cytometry remains relevant and is the primary method for validation of novel immunophenotypes.


Assuntos
Eritropoese , Células-Tronco Hematopoéticas , Humanos , Linhagem da Célula , Diferenciação Celular/genética , Células-Tronco Hematopoéticas/metabolismo , Eritropoese/genética , Células Precursoras Eritroides/metabolismo
8.
Cells ; 12(8)2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-37190030

RESUMO

BACKGROUND: Sickle cell disease (SCD) is a highly prevalent genetic disease caused by a point mutation in the HBB gene, which can lead to chronic hemolytic anemia and vaso-occlusive events. Patient-derived induced pluripotent stem cells (iPSCs) hold promise for the development of novel predictive methods for screening drugs with anti-sickling activity. In this study, we evaluated and compared the efficiency of 2D and 3D erythroid differentiation protocols using a healthy control and SCD-iPSCs. METHODS: iPSCs were subjected to hematopoietic progenitor cell (HSPC) induction, erythroid progenitor cell induction, and terminal erythroid maturation. Differentiation efficiency was confirmed by flow cytometry analysis, colony-forming unit (CFU) assay, morphological analyses, and qPCR-based gene expression analyses of HBB and HBG2. RESULTS: Both 2D and 3D differentiation protocols led to the induction of CD34+/CD43+ HSPCs. The 3D protocol showed good efficiency (>50%) and high productivity (45-fold) for HSPC induction and increased the frequency of BFU-E, CFU-E, CFU-GM, and CFU-GEMM colonies. We also produced CD71+/CD235a+ cells (>65%) with a 630-fold cell expansion relative to that at the beginning of the 3D protocol. After erythroid maturation, we observed 95% CD235a+/DRAQ5- enucleated cells, orthochromatic erythroblasts, and increased expression of fetal HBG2 compared to adult HBB. CONCLUSION: A robust 3D protocol for erythroid differentiation was identified using SCD-iPSCs and comparative analyses; however, the maturation step remains challenging and requires further development.


Assuntos
Anemia Falciforme , Células-Tronco Pluripotentes Induzidas , Adulto , Humanos , Diferenciação Celular , Células-Tronco Hematopoéticas , Células Precursoras Eritroides/metabolismo , Anemia Falciforme/metabolismo
9.
Int J Mol Sci ; 24(1)2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36614221

RESUMO

The human homologue of mouse Ly-1 antibody reactive clone protein (LYAR) is a putative novel regulator of γ-globin gene transcription. The LYAR DNA-binding motif (5'-GGTTAT-3') is located within the 5'-UTR of the Aγ-globin gene. The LYAR rs368698783 (G>A) polymorphism is present in ß-thalassemia patients and decreases the LYAR binding efficiency to the Aγ-globin gene. The objective of this study was to stratify ß-thalassemia patients with respect to the rs368698783 (G>A) polymorphism and to verify whether their erythroid precursor cells (ErPCs) differentially respond in vitro to selected fetal hemoglobin (HbF) inducers. The rs368698783 (G>A) polymorphism was detected by DNA sequencing, hemoglobin production by HPLC, and accumulation of globin mRNAs by RT-qPCR. We found that the LYAR rs368698783 (G>A) polymorphism is associated with high basal and induced production of fetal hemoglobin in ß-thalassemia patients. The most striking association was found using rapamycin as an HbF inducer. The results presented here could be considered important not only for basic biomedicine but also in applied translational research for precision medicine in personalized therapy of ß-thalassemia. Accordingly, our data suggest that the rs368698783 polymorphism might be considered among the parameters useful to recruit patients with the highest probability of responding to in vivo hydroxyurea (HU) treatment.


Assuntos
Células Precursoras Eritroides , Talassemia beta , Humanos , Talassemia beta/tratamento farmacológico , Talassemia beta/genética , Talassemia beta/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/análise , gama-Globinas/genética , gama-Globinas/metabolismo , Proteínas Nucleares/genética , Polimorfismo Genético
10.
Cell Signal ; 104: 110583, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36596353

RESUMO

Chronic kidney disease (CKD) is a global health condition characterized by a progressive deterioration of kidney function. It is associated with high serum levels of uremic toxins (UT), such as Indoxyl Sulfate (IS), which may participate in the genesis of several uremic complications. Anemia is one of the major complications in CKD patients that contribute to cardiovascular disease, increase morbi-mortality, and is associated with a deterioration of kidney failure in these patients. Our study aimed to characterize the impact of IS on CKD-related erythropoiesis. Using cellular and pre-clinical models, we studied cellular and molecular effects of IS on the growth and differentiation of erythroid cells. First, we examined the effect of clinically relevant concentrations of IS (up to 250 µM) in the UT7/EPO cell line. IS at 250 µM increased apoptosis of UT7/EPO cells at 48 h compared to the control condition. We confirmed this apoptotic effect of IS in erythropoiesis in human primary CD34+ cells during the later stages of erythropoiesis. Then, in IS-treated human primary CD34+ cells and in a (5/6 Nx) mice model, a blockage at the burst-forming unit-erythroid (BFU-E) stage of erythropoiesis was also observed. Finally, IS deregulates a number of erythropoietic related genes such as GATA-1, Erythropoietin-Receptor (EPO-R), and ß-globin. Our findings suggest that IS could affect cell viability and differentiation of erythroid progenitors by altering erythropoiesis and contributing to the development of anemia in CKD.


Assuntos
Anemia , Eritropoetina , Insuficiência Renal Crônica , Camundongos , Animais , Humanos , Eritropoese/fisiologia , Indicã/metabolismo , Indicã/farmacologia , Células Precursoras Eritroides/metabolismo , Anemia/metabolismo , Insuficiência Renal Crônica/metabolismo
11.
Hematology ; 27(1): 1152-1162, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36178486

RESUMO

OBJECTIVE: This study attempted to investigate miR-92a-3p expression in peripheral blood of patients with severe ß-thalassemia, and the effect and action mechanism of miR-92a-3p on γ-globin expression and oxidative stress in erythroid precursor cells. METHODS: CD34+ hematopoietic progenitor cells (HPCs) were isolated from peripheral blood of healthy volunteers and patients with severe ß-thalassemia. The levels of miR-92a-3p, BCL11A, and γ-globin were measured in erythroid precursor cells. High-performance liquid chromatography (HPLC) was used to analyze hemoglobin F (HbF) content. HPCs were induced with erythroid differentiation and erythroid precursor cells were then obtained. The relevance between miR-92a-3p and BCL11A was studied using dual luciferase reporter gene assay, and the correlation between miR-92a-3p and HbF was assayed by Pearson correlation analysis. Reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) in erythroid precursor cells were tested to evaluate oxidative stress. Cell apoptosis was examined by flow cytometry. RESULTS: Remarkably higher expression of miR-92a-3p was observed in erythroid precursor cells. Increased expression of miR-92a-3p resulted in elevated levels of γ-globin, GSH, and SOD, reduced expression of ROS and MDA, and decreased cell apoptosis. BCL11A was identified as a target of miR-92a-3p and to be downregulated by miR-92a-3p. Moreover, BCL11A knockdown alone increased the expression of γ-globin, SOD and GSH, and repressed the levels of ROS and MDA and cell apoptosis, and the following inhibition of miR-92a-3p changed these patterns. CONCLUSIONS: Our data indicated that miR-92a-3p might increase γ-globin level and reduce oxidative stress and apoptosis in erythroid precursor cells by downregulating BCL11A.


Assuntos
MicroRNAs , Talassemia beta , Apoptose , Proteínas de Transporte/genética , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Glutationa , Humanos , Malondialdeído/metabolismo , Proteínas Nucleares/genética , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/metabolismo , Superóxido Dismutase , Fatores de Transcrição/metabolismo , Talassemia beta/genética , gama-Globinas/genética , gama-Globinas/metabolismo
12.
Blood ; 140(22): 2371-2384, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36054916

RESUMO

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.


Assuntos
Eritropoetina , Transtornos Mieloproliferativos , Neoplasias , Policitemia , Humanos , Eritropoese/fisiologia , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Policitemia/metabolismo , Eritropoetina/metabolismo , Transtornos Mieloproliferativos/metabolismo , Células Precursoras Eritroides/metabolismo , Neoplasias/metabolismo , Receptor IGF Tipo 1/metabolismo
13.
Saudi Med J ; 43(8): 899-906, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35964959

RESUMO

OBJECTIVES: To assess the effect of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection on erythropoiesis and red blood cells (RBC) surface markers by evaluating erythroid progenitor cells (CD [cluster of differentiation]71+/CD235a+) and RBC surface markers (CD235a and CD36), together with various hematological parameters. METHODS: This case-control study includes 47 participants recruited in the study: 30 patients with coronavirus disease 2019 (COVID-19) and 17 healthy individuals. The COVID-19 patients were recruited from the intensive care unit (ICU) of various hospitals in Makkah, Saudi Arabia. Blood samples were collected during July and September 2021. Red blood cells indices were measured using a CBC analyzer. The expression of CD235a, CD71, and CD36 was obtained using flow cytometry technique. The unpaired t-test was conducted to evaluate the differences in these markers in COVID-19 patients and healthy individuals. RESULTS: The data showed that more than half of the COVID-19 patients were anemic (64%). Expansion of erythroid progenitors (CD71+/CD235a+) was detected in the COVID-19 patients. Analysis of the expression of RBC surface markers, such as CD235a and CD36, showed that SARS-CoV-2 was associated with significantly higher expression of these markers in COVID-19 patients. CONCLUSION: Severe acute respiratory syndrome coronavirus-2 promoted the expansion of erythroid progenitors in the peripheral blood of COVID-19 patients. In addition, the expression of RBC surface markers was higher in COVID-19 patients. The expansion of erythroid progenitors and alteration of RBC surface markers can contribute to erythrocytopathies observed in severe COVID-19 patients and can therefore be used as prognostic factors.


Assuntos
COVID-19 , Biomarcadores/metabolismo , Estudos de Casos e Controles , Células Precursoras Eritroides/metabolismo , Eritropoese , Humanos , SARS-CoV-2
14.
Rinsho Ketsueki ; 63(6): 600-607, 2022.
Artigo em Japonês | MEDLINE | ID: mdl-35831194

RESUMO

Sideroblastic anemias (SAs) are a group of heterogeneous congenital and acquired disorders characterized by anemia and presence of ring sideroblasts in the bone marrow. Congenital SA is a rare condition caused by mutations of genes involved in heme biosynthesis, iron-sulfur cluster biosynthesis, and mitochondrial protein synthesis. SAs can also occur following exposure to certain drugs or alcohol or caused by copper deficiency (secondary SA). SAs have been found to be associated with myelodysplastic syndrome (idiopathic SA), which strongly correlates with specific somatic mutations in SF3B1 (splicing factor 3b subunit 1), involved in the RNA splicing machinery. The recent widespread use of genome-editing technology and next-generation sequencing has led to a better understanding of the molecular pathophysiology of SAs. This review discusses the current understanding of the pathophysiology of SAs.


Assuntos
Anemia Sideroblástica , Síndromes Mielodisplásicas , Anemia Sideroblástica/genética , Células Precursoras Eritroides/metabolismo , Humanos , Mutação , Síndromes Mielodisplásicas/complicações , Splicing de RNA
15.
Exp Hematol ; 112-113: 9-14.e7, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35839944

RESUMO

The accumulation of unbound α-globin chains in red blood cells is a crucial pathophysiology of ß-thalassemia. IOX1 (5-carboxy-8-hydroxyquinoline) is a broad-spectrum 2-oxoglutarate (2OG)-dependent oxygenase inhibitor that can reduce α-globin mRNA expression in human cord blood erythroid progenitor cells. Therefore, IOX1 has been proposed as a potential compound for ß-thalassemia treatment through the decrease in α-globin chain synthesis. However, there is no empirical evidence regarding the consequences of IOX1 in ß-thalassemia. In this study, the therapeutic effects of IOX1 were investigated in ß0-thalassemia/hemoglobin E (HbE) erythroid progenitor cells during in vitro erythropoiesis. The results indicated that IOX1 had no impact on α-globin gene expression, but it led instead to significant decreases in γ-globin and fetal hemoglobin (HbF, α2γ2) production without affecting well-known globin regulators: KLF1, BCL11A, LRF, and GATA1. In addition, differential mRNA expression of several genes in the hypoxia response pathway revealed the induction of EGLN1, the PHD2-encoding gene, as a result of IOX1 treatment. These findings suggested that IOX1 fails to lower α-globin gene expression; on the contrary, it mediates γ-globin and HbF silencing in ß0-thalassemia/HbE erythroid progenitor cells. Because of the negative correlation of EGLN1 and γ-globin gene expression after IOX1 treatment, repurposing IOX1 to study the hypoxia response pathway and γ-globin regulation may provide beneficial information for ß-thalassemia.


Assuntos
Hemoglobina E , Talassemia , Talassemia beta , Adulto , Proteínas de Transporte/metabolismo , Células Eritroides/metabolismo , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal , Hemoglobina E/genética , Hemoglobina E/metabolismo , Humanos , Hipóxia/metabolismo , RNA Mensageiro/genética , Talassemia/metabolismo , alfa-Globinas/metabolismo , Talassemia beta/terapia , gama-Globinas/genética
16.
J Biotechnol ; 355: 10-20, 2022 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-35777457

RESUMO

Human erythropoietin (EPO) is a key cytokine in erythropoiesis by regulating differentiation of erythroid progenitor cells into red blood cells (RBCs). Plant cell cultures are considered as promising alternative bioproduction platforms for EPO. To overcome the bottlenecks of low protein productivity and secretion, EPO was expressed in tobacco BY-2 cells with a designer peptide tag, termed (SP)20 that consists of 20 tandem repeats of a "Ser-Pro" motif. This de novo designed tag directed extensive O-glycosylation on each Pro residue in plant cells and acted as a molecular carrier to promote the extracellular secretion of EPO. To facilitate the establishment of stable and high-expression BY-2 cell lines, EPO molecules were co-expressed with a reporter protein GFP, which could be used as a visual marker to monitor the protein expression during the subculture. The engineered (SP)20 glycomodule substantially increased the secreted yields of EPO up to 4.31 µg/mL. The (SP)20-tagged EPOs exhibited the expected activity in promoting the proliferation of TF-1 cells, though their EC50 was 12-fold higher than that of EPO standard. The (SP)20-tagged EPOs could also stimulate the ex vivo expansion and differentiation of hematopoietic stem cell (CD34+ cells) towards RBCs.


Assuntos
Eritropoetina , Diferenciação Celular , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Eritropoetina/genética , Eritropoetina/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , /genética
17.
Elife ; 112022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35713400

RESUMO

Acute anemia induces rapid expansion of erythroid precursors and accelerated differentiation to replenish erythrocytes. Paracrine signals-involving cooperation between stem cell factor (SCF)/Kit signaling and other signaling inputs-are required for the increased erythroid precursor activity in anemia. Our prior work revealed that the sterile alpha motif (SAM) domain 14 (Samd14) gene increases the regenerative capacity of the erythroid system in a mouse genetic model and promotes stress-dependent Kit signaling. However, the mechanism underlying Samd14's role in stress erythropoiesis is unknown. We identified a protein-protein interaction between Samd14 and the α- and ß-heterodimers of the F-actin capping protein (CP) complex. Knockdown of the CP ß subunit increased erythroid maturation in murine ex vivo cultures and decreased colony forming potential of stress erythroid precursors. In a genetic complementation assay for Samd14 activity, our results revealed that the Samd14-CP interaction is a determinant of erythroid precursor cell levels and function. Samd14-CP promotes SCF/Kit signaling in CD71med spleen erythroid precursors. Given the roles of Kit signaling in hematopoiesis and Samd14 in Kit pathway activation, this mechanism may have pathological implications in acute/chronic anemia.


Anemia is a condition in which the body has a shortage of healthy red blood cells to carry enough oxygen to support its organs. A range of factors are known to cause anemia, including traumatic blood loss, toxins or nutritional deficiency. An estimated one-third of all women of reproductive age are anemic, which can cause tiredness, weakness and shortness of breath. Severe anemia drives the release of hormones and growth factors, leading to a rapid regeneration of precursor red blood cells to replenish the supply in the blood. To understand how red blood cell regeneration is controlled, Ray et al. studied proteins involved in regenerating blood using mice in which anemia had been induced with chemicals. Previous research had shown that the protein Samd14 is produced at higher quantities in individuals with anemia, and is involved with the recovery of lost red blood cells. However, it is not known how the Samd14 protein plays a role in regenerating blood cells, or whether Samd14 interacts with other proteins required for red blood cell production. To shed light on these questions, mouse cells exposed to anemia conditions were used to see what proteins Samd14 binds to. Purifying Samd14 revealed that it interacts with the actin capping protein. This interaction relies on a specific region of Samd14 that is similar to regions in other proteins that bind capping proteins. Ray et al. found that the interaction between Samd14 and the actin capping protein increased the signals needed for the development and survival of new red blood cells. These results identify a signaling mechanism that, if disrupted, could cause anemia to develop. They lead to a better understanding of how our bodies recover from anemia, and potential avenues to treat this condition.


Assuntos
Anemia , Eritropoese , Animais , Diferenciação Celular , Eritrócitos , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Camundongos , Proteínas/metabolismo
18.
Blood Cells Mol Dis ; 97: 102688, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35717902

RESUMO

Erythropoiesis is a tightly regulated process. It is stimulated by decreased oxygen in circulation, which leads to the secretion of the hormone erythropoietin (Epo) by the kidneys. An additional layer of control involves the coordinated sensing and use of nutrients. Much cellular machinery contributes to sensing and responding to nutrient status in cells, and one key participant is the kinase LKB1. The current study examines the role of LKB1 in erythropoiesis using a murine in vivo and ex vivo conditional knockout system. In vivo analysis showed erythroid loss of LKB1 to be associated with a robust increase in serum Epo and mild reticulocytosis. Despite these abnormalities, no evidence of anemia or hemolysis was found. Further characterization using an ex vivo progenitor culture assay demonstrated accelerated erythroid maturation in the LKB1-deficient cells. Based on pharmacologic evidence, this phenotype appeared to result from impaired AMP-activated protein kinase (AMPK) signaling downstream of LKB1. These findings reveal a role for LKB1 in fine-tuning Epo-driven erythropoiesis in association with maturational control.


Assuntos
Proteínas Quinases Ativadas por AMP , Células Precursoras Eritroides , Eritropoese , Eritropoetina , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Precursoras Eritroides/metabolismo , Eritropoese/genética , Eritropoese/fisiologia , Eritropoetina/genética , Eritropoetina/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo
19.
Int J Mol Sci ; 23(9)2022 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-35563395

RESUMO

Recently developed Prime Editor 3 (PE3) has been implemented to induce genome editing in various cell types but has not been proven in human hematopoietic stem and progenitor cells. Using PE3, we successfully installed the beta-thalassemia (beta-thal) mutations in the HBB gene in the erythroid progenitor cell line HUDEP-2. We inserted the mCherry reporter gene cassette into editing plasmids, each including the prime editing guide RNA (pegRNA) and nick sgRNA. The plasmids were electroporated into HUDEP-2 cells, and the PE3 modified cells were identified by mCherry expression and collected using fluorescence-activated cell sorting (FACS). Sanger sequencing of the positive cells confirmed that PE3 induced precise beta-thal mutations with editing ratios from 4.55 to 100%. Furthermore, an off-target analysis showed no unintentional edits occurred in the cells. The editing ratios and parameters of pegRNA and nick sgRNA were also analyzed and summarized and will contribute to enhanced PE3 design in future studies. The characterization of the HUDEP-2 beta-thal cells showed typical thalassemia phenotypes, involving ineffective erythropoiesis, abnormal erythroid differentiation, high apoptosis rate, defective alpha-globin colocalization, cell viability deterioration, and ROS resisting deficiency. These HUDEP-2 beta-thal cells could provide ideal models for future beta-thal gene therapy studies.


Assuntos
Células Precursoras Eritroides , Edição de Genes , Hemoglobinas , Talassemia beta , Células Precursoras Eritroides/metabolismo , Edição de Genes/métodos , Hemoglobinas/genética , Hemoglobinas/uso terapêutico , Humanos , Mutação , alfa-Globinas/genética , Globinas beta/genética , Talassemia beta/genética , Talassemia beta/terapia
20.
Int J Mol Sci ; 23(9)2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35563527

RESUMO

Erythropoietin (EPO) signaling plays a vital role in erythropoiesis by regulating proliferation and lineage-specific differentiation of murine hematopoietic progenitor cells (HPCs). An important downstream response of EPO signaling is calcium (Ca2+) influx, which is regulated by transient receptor potential channel (TRPC) proteins, particularly TRPC2 and TRPC6. While EPO induces Ca2+ influx through TRPC2, TRPC6 inhibits the function of TRPC2. Thus, interactions between TRPC2 and TRPC6 regulate the rate of Ca2+ influx in EPO-induced erythropoiesis. In this study, we observed that the expression of TRPC6 in KIT-positive erythroid progenitor cells was regulated by DOT1L. DOT1L is a methyltransferase that plays an important role in many biological processes during embryonic development including early erythropoiesis. We previously reported that Dot1l knockout (Dot1lKO) HPCs in the yolk sac failed to develop properly, which resulted in lethal anemia. In this study, we detected a marked downregulation of Trpc6 gene expression in Dot1lKO progenitor cells in the yolk sac compared to the wild type (WT). The promoter and the proximal regions of the Trpc6 gene locus exhibited an enrichment of H3K79 methylation, which is mediated solely by DOT1L. However, the expression of Trpc2, the positive regulator of Ca2+ influx, remained unchanged, resulting in an increased TRPC2/TRPC6 ratio. As the loss of DOT1L decreased TRPC6, which inhibited Ca2+ influx by TRPC2, Dot1lKO HPCs in the yolk sac exhibited accelerated and sustained elevated levels of Ca2+ influx. Such heightened Ca2+ levels might have detrimental effects on the growth and proliferation of HPCs in response to EPO.


Assuntos
Cálcio , Eritropoetina , Histona-Lisina N-Metiltransferase , Animais , Cálcio/metabolismo , Cálcio da Dieta , Epoetina alfa , Células Precursoras Eritroides/metabolismo , Eritropoese , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Camundongos , Receptores da Eritropoetina/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...