Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.558
Filter
1.
PLoS One ; 19(7): e0305816, 2024.
Article in English | MEDLINE | ID: mdl-39038020

ABSTRACT

Erythroid cells, serving as progenitors and precursors to erythrocytes responsible for oxygen transport, were shown to exhibit an immunosuppressive and immunoregulatory phenotype. Previous investigations from our research group have revealed an antimicrobial gene expression profile within murine bone marrow erythroid cells which suggested a role for erythroid cells in innate immunity. In the present study, we focused on elucidating the characteristics of human bone marrow erythroid cells through comprehensive analyses, including NanoString gene expression profiling utilizing the Immune Response V2 panel, a BioPlex examination of chemokine and TGF-beta family proteins secretion, and analysis of publicly available single-cell RNA-seq data. Our findings demonstrate that an erythroid cell subpopulation manifests a myeloid-like gene expression signature comprised of antibacterial immunity and neutrophil chemotaxis genes which suggests an involvement of human erythroid cells in the innate immunity. Furthermore, we found that human erythroid cells secreted CCL22, CCL24, CXCL5, CXCL8, and MIF chemokines. The ability of human erythroid cells to express these chemokines might facilitate the restriction of immune cells in the bone marrow under normal conditions or contribute to the ability of erythroid cells to induce local immunosuppression by recruiting immune cells in their immediate vicinity in case of extramedullary hematopoiesis.


Subject(s)
Erythroid Cells , Monocytes , Humans , Monocytes/metabolism , Monocytes/cytology , Monocytes/immunology , Erythroid Cells/metabolism , Erythroid Cells/cytology , Immunity, Innate , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Transcriptome , Gene Expression Profiling , Chemokine CXCL5/metabolism , Chemokine CXCL5/genetics , Myeloid Cells/metabolism , Chemokines/metabolism , Chemokines/genetics , Interleukin-8 , Intramolecular Oxidoreductases
2.
Int J Mol Sci ; 25(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39000360

ABSTRACT

Mitochondrial dysfunction has been increasingly recognized as a trigger for systemic lupus erythematosus (SLE). Recent bioinformatics studies have suggested Fam210b as a significant candidate for the classification and therapeutic targeting of SLE. To experimentally prove the role of Fam210b in SLE, we constructed Fam210b knockout (Fam210b-/-) mice using the CRISPR-Cas9 method. We found that approximately 15.68% of Fam210b-/- mice spontaneously developed lupus-like autoimmunity, which was characterized by skin ulcerations, splenomegaly, and an increase in anti-double-stranded DNA (anti-dsDNA) IgG antibodies and anti-nuclear antibodies(ANA). Single-cell sequencing showed that Fam210b was mainly expressed in erythroid cells. Critically, the knockout of Fam210b resulted in abnormal erythrocyte differentiation and development in the spleens of mice. Concurrently, the spleens exhibited an increased number of CD71+ erythroid cells, along with elevated levels of reactive oxygen species (ROS) in the erythrocytes. The co-culture of CD71+ erythroid cells and lymphocytes resulted in lymphocyte activation and promoted dsDNA and IgG production. In summary, Fam210b knockout leads to a low probability of lupus-like symptoms in mice through the overproduction of ROS in CD71+ erythroid cells. Thus, Fam210b reduction may serve as a novel key marker that triggers the development of SLE.


Subject(s)
Lupus Erythematosus, Systemic , Mice, Knockout , Animals , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Antibodies, Antinuclear , Mitochondrial Membranes/metabolism , Erythroid Cells/metabolism , Erythroid Cells/pathology , Disease Models, Animal , Immunoglobulin G/metabolism , Mice, Inbred C57BL , Spleen/metabolism , Spleen/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Female
3.
Adv Exp Med Biol ; 1459: 199-215, 2024.
Article in English | MEDLINE | ID: mdl-39017845

ABSTRACT

BCL11A, a zinc finger repressor, is a stage-specific transcription factor that controls the switch from fetal (HbF, α2γ2) to adult (HbA, α2ß2) hemoglobin in erythroid cells. While BCL11A was known as a factor critical for B-lymphoid cell development, its relationship to erythroid cells and HbF arose through genome-wide association studies (GWAS). Subsequent work validated its role as a silencer of γ-globin gene expression in cultured cells and mice. Erythroid-specific loss of BCL11A rescues the phenotype of engineered sickle cell disease (SCD) mice, thereby suggesting that downregulation of BCL11A expression might be beneficial in patients with SCD and ß-thalassemia. Common genetic variation in GWAS resides in an erythroid-specific enhancer within the BCL11A gene that is required for its own expression. CRISPR/Cas9 gene editing of the enhancer revealed a GATA-binding site that confers a large portion of its regulatory function. Disruption of the GATA site leads to robust HbF reactivation. Advancement of a guide RNA targeting the GATA-binding site in clinical trials has recently led to approval of first-in-man use of ex vivo CRISPR editing of hematopoietic stem/progenitor cells (HSPCs) as therapy of SCD and ß-thalassemia. Future challenges include expanding access and infrastructure for delivery of genetic therapy to eligible patients, reducing potential toxicity and costs, exploring prospects for in vivo targeting of hematopoietic stem cells (HSCs), and developing small molecule drugs that impair function of BCL11A protein as an alternative option.


Subject(s)
Erythroid Cells , Repressor Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Humans , Animals , Erythroid Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Mice , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , CRISPR-Cas Systems , Gene Editing/methods , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Gene Expression Regulation , Genome-Wide Association Study
4.
Adv Exp Med Biol ; 1459: 217-242, 2024.
Article in English | MEDLINE | ID: mdl-39017846

ABSTRACT

Erythroid Krüppel-like factor (KLF1), first discovered in 1992, is an erythroid-restricted transcription factor (TF) that is essential for terminal differentiation of erythroid progenitors. At face value, KLF1 is a rather inconspicuous member of the 26-strong SP/KLF TF family. However, 30 years of research have revealed that KLF1 is a jack of all trades in the molecular control of erythropoiesis. Initially described as a one-trick pony required for high-level transcription of the adult HBB gene, we now know that it orchestrates the entire erythroid differentiation program. It does so not only as an activator but also as a repressor. In addition, KLF1 was the first TF shown to be directly involved in enhancer/promoter loop formation. KLF1 variants underlie a wide range of erythroid phenotypes in the human population, varying from very mild conditions such as hereditary persistence of fetal hemoglobin and the In(Lu) blood type in the case of haploinsufficiency, to much more serious non-spherocytic hemolytic anemias in the case of compound heterozygosity, to dominant congenital dyserythropoietic anemia type IV invariably caused by a de novo variant in a highly conserved amino acid in the KLF1 DNA-binding domain. In this chapter, we present an overview of the past and present of KLF1 research and discuss the significance of human KLF1 variants.


Subject(s)
Erythropoiesis , Kruppel-Like Transcription Factors , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Humans , Erythropoiesis/genetics , Animals , Cell Differentiation/genetics , Erythroid Cells/metabolism , Erythroid Cells/cytology , Mutation
5.
Nat Commun ; 15(1): 5678, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971858

ABSTRACT

Inherited non-hemolytic anemia is a group of rare bone marrow disorders characterized by erythroid defects. Although concerted efforts have been made to explore the underlying pathogenetic mechanisms of these diseases, the understanding of the causative mutations are still incomplete. Here we identify in a diseased pedigree that a gain-of-function mutation in toll-like receptor 8 (TLR8) is implicated in inherited non-hemolytic anemia. TLR8 is expressed in erythroid lineage and erythropoiesis is impaired by TLR8 activation whereas enhanced by TLR8 inhibition from erythroid progenitor stage. Mechanistically, TLR8 activation blocks annexin A2 (ANXA2)-mediated plasma membrane localization of STAT5 and disrupts EPO signaling in HuDEP2 cells. TLR8 inhibition improves erythropoiesis in RPS19+/- HuDEP2 cells and CD34+ cells from healthy donors and inherited non-hemolytic anemic patients. Collectively, we identify a gene implicated in inherited anemia and a previously undescribed role for TLR8 in erythropoiesis, which could potentially be explored for therapeutic benefit in inherited anemia.


Subject(s)
Anemia , Erythropoiesis , Toll-Like Receptor 8 , Humans , Erythropoiesis/genetics , Toll-Like Receptor 8/metabolism , Toll-Like Receptor 8/genetics , Female , Anemia/genetics , Male , Pedigree , Erythropoietin/metabolism , Erythropoietin/genetics , Adult , Signal Transduction , Mutation , Erythroid Cells/metabolism , Animals , Erythroid Precursor Cells/metabolism
6.
Microbiome ; 12(1): 142, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080725

ABSTRACT

BACKGROUND: The establishment of microbial communities in neonatal mammals plays a pivotal role in shaping their immune responses to infections and other immune-related conditions. This process is influenced by a combination of endogenous and exogenous factors. Previously, we reported that depletion of CD71 + erythroid cells (CECs) results in an inflammatory response to microbial communities in newborn mice. RESULTS: Here, we systemically tested this hypothesis and observed that the small intestinal lamina propria of neonatal mice had the highest frequency of CECs during the early days of life. This high abundance of CECs was attributed to erythropoiesis niches within the small intestinal tissues. Notably, the removal of CECs from the intestinal tissues by the anti-CD71 antibody disrupted immune homeostasis. This disruption was evident by alteration in the expression of antimicrobial peptides (AMPs), toll-like receptors (TLRs), inflammatory cytokines/chemokines, and resulting in microbial dysbiosis. Intriguingly, these alterations in microbial communities persisted when tested 5 weeks post-treatment, with a more notable effect observed in female mice. This illustrates a sex-dependent association between CECs and neonatal microbiome modulation. Moreover, we extended our studies on pregnant mice, observing that modulating CECs substantially alters the frequency and diversity of their microbial communities. Finally, we found a significantly lower proportion of CECs in the cord blood of pre-term human newborns, suggesting a potential role in dysregulated immune responses to microbial communities in the gut. CONCLUSIONS: Our findings provide novel insights into pivotal role of CECs in immune homeostasis and swift adaptation of microbial communities in newborns. Despite the complexity of the cellular biology of the gut, our findings shed light on the previously unappreciated role of CECs in the dialogue between the microbiota and immune system. These findings have significant implications for human health. Video Abstract.


Subject(s)
Animals, Newborn , Antigens, CD , Erythroid Cells , Gastrointestinal Microbiome , Receptors, Transferrin , Animals , Female , Mice , Pregnancy , Antigens, CD/metabolism , Erythroid Cells/immunology , Receptors, Transferrin/metabolism , Male , Symbiosis , Dysbiosis/microbiology , Humans , Mice, Inbred C57BL , Intestine, Small/microbiology , Intestine, Small/immunology
7.
J Clin Lab Anal ; 38(11-12): e25084, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924171

ABSTRACT

BACKGROUND/OBJECTIVES: CD71+ erythroid cells (CECs) are immature red blood cells (proerythroblasts, erythroblasts, and reticulocytes). CECs play an important role in the development of sepsis and cancer by causing immunosuppression. We examined the CEC levels in the peripheral blood of beta thalassemia (ßThal) patients and investigated the relationship between CECs and the clinical status of the patients, especially splenectomy. METHODS: Sixty-eight patients with ßThal (46 splenectomized and 22 nonsplenectomized) and 15 healthy controls were included in this study. The hemogram parameters, ferritin, and CECs (flow cytometry method) were measured. RESULTS: It was observed that the CEC level in the patient group was significantly higher than the control group (p < 0.05). CEC levels were found to be significantly higher in patients with splenectomy than in patients with nonsplenectomy (p < 0.05). CEC levels were higher in patients with nontransfusion-dependent ßT (NTD-ßThal) than in patients with transfusion-dependent ßT (TD-ßThal) (p < 0.05). CEC levels were found to be significantly higher in patients with splenectomy than in patients with nonsplenectomy in both TD-ßThal and NTD-ßThal groups (p < 0.05). There was a moderate-negative correlation was detected between CECs and Hb levels (r = -0.467; p < 0.05). CONCLUSIONS: High CEC levels in ßThal patients develop as a result of ineffective erythropoiesis. We think that keeping CEC levels under control is important for prognosis, especially in patients with splenectomy.


Subject(s)
Antigens, CD , Receptors, Transferrin , Splenectomy , beta-Thalassemia , Humans , beta-Thalassemia/blood , beta-Thalassemia/surgery , Female , Male , Receptors, Transferrin/blood , Prognosis , Antigens, CD/blood , Adult , Adolescent , Young Adult , Case-Control Studies , Erythrocytes/metabolism , Child , Erythroid Cells/metabolism , Erythroid Cells/pathology
8.
Toxicology ; 505: 153843, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38801936

ABSTRACT

Benzene, a widely used industrial chemical, has been clarified to cause hematotoxicity. Our previous study suggested that miR-451a may play a role in benzene-induced impairment of erythroid differentiation. However, the mechanism underlying remains unclear. In this study, we explored the role of miR-451a and its underlying mechanisms in hydroquinone (HQ)-induced suppression of erythroid differentiation in K562 cells. 0, 1.0, 2.5, 5.0, 10.0, and 50 µM HQ treatment of K562 cells resulted in a dose-dependent inhibition of erythroid differentiation, as well as the expression of miR-451a. Bioinformatics analysis was conducted to predict potential target genes of miR-451a and dual-luciferase reporter assays confirmed that miR-451a can directly bind to the 3'-UTR regions of BATF, SETD5, and ARHGEF3 mRNAs. We further demonstrated that over-expression or down-regulation of miR-451a altered the expression of BATF, SETD5, and ARHGEF3, and also modified erythroid differentiation. In addition, BATF, SETD5, and ARHGEF3 were verified to play a role in HQ-induced inhibition of erythroid differentiation in this study. Knockdown of SETD5 and ARHGEF3 reversed HQ-induced suppression of erythroid differentiation while knockdown of BATF had the opposite effect. On the other hand, we also identified c-Jun as a potential transcriptional regulator of miR-451a. Forced expression of c-Jun increased miR-451a expression and reversed the inhibition of erythroid differentiation induced by HQ, whereas knockdown of c-Jun had the opposite effect. And the binding site of c-Jun and miR-451a was verified by dual-luciferase reporter assay. Collectively, our findings indicate that miR-451a and its downstream targets BATF, SETD5, and ARHGEF3 are involved in HQ-induced erythroid differentiation disorder, and c-Jun regulates miR-451a as a transcriptional regulator in this process.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cell Differentiation , MicroRNAs , Rho Guanine Nucleotide Exchange Factors , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Differentiation/drug effects , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , K562 Cells , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Erythroid Cells/drug effects , Erythroid Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-jun/genetics , Methyltransferases/genetics , Methyltransferases/metabolism
9.
Int J Hematol ; 120(2): 157-166, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38814500

ABSTRACT

G protein pathway suppressor 2 (GPS2) has been shown to play a pivotal role in human and mouse definitive erythropoiesis in an EKLF-dependent manner. However, whether GPS2 affects human primitive erythropoiesis is still unknown. This study demonstrated that GPS2 positively regulates erythroid differentiation in K562 cells, which have a primitive erythroid phenotype. Overexpression of GPS2 promoted hemin-induced hemoglobin synthesis in K562 cells as assessed by the increased percentage of benzidine-positive cells and the deeper red coloration of the cell pellets. In contrast, knockdown of GPS2 inhibited hemin-induced erythroid differentiation of K562 cells. GPS2 overexpression also enhanced erythroid differentiation of K562 cells induced by cytosine arabinoside (Ara-C). GPS2 induced hemoglobin synthesis by increasing the expression of globin and ALAS2 genes, either under steady state or upon hemin treatment. Promotion of erythroid differentiation of K562 cells by GPS2 mainly relies on NCOR1, as knockdown of NCOR1 or lack of the NCOR1-binding domain of GPS2 potently diminished the promotive effect. Thus, our study revealed a previously unknown role of GPS2 in regulating human primitive erythropoiesis in K562 cells.


Subject(s)
Cell Differentiation , Erythropoiesis , Hemin , Leukemia, Erythroblastic, Acute , Nuclear Receptor Co-Repressor 1 , Humans , K562 Cells , Erythropoiesis/genetics , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Hemin/pharmacology , Erythroid Cells/metabolism , Erythroid Cells/cytology , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Hemoglobins/metabolism , Gene Knockdown Techniques , Intracellular Signaling Peptides and Proteins
10.
Sci Rep ; 14(1): 10287, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38704454

ABSTRACT

The identification of regulatory networks contributing to fetal/adult gene expression switches is a major challenge in developmental biology and key to understand the aberrant proliferation of cancer cells, which often reactivate fetal oncogenes. One key example is represented by the developmental gene LIN28B, whose aberrant reactivation in adult tissues promotes tumor initiation and progression. Despite the prominent role of LIN28B in development and cancer, the mechanisms of its transcriptional regulation are largely unknown. Here, by using quantitative RT-PCR and single cell RNA sequencing data, we show that in erythropoiesis the expression of the transcription factor SOX6 matched a sharp decline of LIN28B mRNA during human embryo/fetal to adult globin switching. SOX6 overexpression repressed LIN28B not only in a panel of fetal-like erythroid cells (K562, HEL and HUDEP1; ≈92% p < 0.0001, 54% p = 0.0009 and ≈60% p < 0.0001 reduction, respectively), but also in hepatoblastoma HepG2 and neuroblastoma SH-SY5H cells (≈99% p < 0.0001 and ≈59% p < 0.0001 reduction, respectively). SOX6-mediated repression caused downregulation of the LIN28B/Let-7 targets, including MYC and IGF2BP1, and rapidly blocks cell proliferation. Mechanistically, Lin28B repression is accompanied by SOX6 physical binding within its locus, suggesting a direct mechanism of LIN28B downregulation that might contribute to the fetal/adult erythropoietic transition and restrict cancer proliferation.


Subject(s)
RNA-Binding Proteins , SOXD Transcription Factors , Humans , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation, Developmental , Erythropoiesis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Hep G2 Cells , K562 Cells , Gene Expression Regulation, Neoplastic , Erythroid Cells/metabolism
11.
Mol Med Rep ; 29(6)2024 06.
Article in English | MEDLINE | ID: mdl-38695236

ABSTRACT

During hematopoiesis, megakaryocytic erythroid progenitors (MEPs) differentiate into megakaryocytic or erythroid lineages in response to specific transcriptional factors, yet the regulatory mechanism remains to be elucidated. Using the MEP­like cell line HEL western blotting, RT­qPCR, lentivirus­mediated downregulation, flow cytometry as well as chromatin immunoprecipitation (ChIp) assay demonstrated that the E26 transformation­specific (ETS) transcription factor friend leukemia integration factor 1 (Fli­1) inhibits erythroid differentiation. The present study using these methods showed that while FLI1­mediated downregulation of GATA binding protein 1 (GATA1) suppresses erythropoiesis, its direct transcriptional induction of GATA2 promotes megakaryocytic differentiation. GATA1 is also involved in megakaryocytic differentiation through regulation of GATA2. By contrast to FLI1, the ETS member erythroblast transformation­specific­related gene (ERG) negatively controls GATA2 and its overexpression through exogenous transfection blocks megakaryocytic differentiation. In addition, FLI1 regulates expression of LIM Domain Binding 1 (LDB1) during erythroid and megakaryocytic commitment, whereas shRNA­mediated depletion of LDB1 downregulates FLI1 and GATA2 but increases GATA1 expression. In agreement, LDB1 ablation using shRNA lentivirus expression blocks megakaryocytic differentiation and modestly suppresses erythroid maturation. These results suggested that a certain threshold level of LDB1 expression enables FLI1 to block erythroid differentiation. Overall, FLI1 controlled the commitment of MEP to either erythroid or megakaryocytic lineage through an intricate regulation of GATA1/GATA2, LDB1 and ERG, exposing multiple targets for cell fate commitment and therapeutic intervention.


Subject(s)
Cell Differentiation , Erythroid Cells , Megakaryocytes , Humans , Cell Differentiation/genetics , Cell Line , Erythroid Cells/metabolism , Erythroid Cells/cytology , GATA1 Transcription Factor/metabolism , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , Gene Expression Regulation , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Megakaryocytes/metabolism , Megakaryocytes/cytology , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Transcriptional Regulator ERG/metabolism , Transcriptional Regulator ERG/genetics
12.
STAR Protoc ; 5(2): 103016, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38640065

ABSTRACT

Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.


Subject(s)
CRISPR-Cas Systems , Erythroid Cells , Gene Editing , Gene Knock-In Techniques , CRISPR-Cas Systems/genetics , Animals , Mice , Erythroid Cells/metabolism , Erythroid Cells/cytology , Gene Knock-In Techniques/methods , Gene Editing/methods , Transfection/methods , Cell Line , Homologous Recombination/genetics , Electroporation/methods
13.
PLoS One ; 19(4): e0300958, 2024.
Article in English | MEDLINE | ID: mdl-38625890

ABSTRACT

Oxidative damage to erythroid cells plays a key role in the pathogenesis of thalassemia. The oxidative stress in thalassemia is potentiated by heme, nonheme iron, and free iron produced by the Fenton reaction, due to degradation of the unstable hemoglobin and iron overload. In addition, the levels of antioxidant enzymes and molecules are significantly decreased in erythrocytes in α- and ß-thalassemia. The control of oxidative stress in red blood cells (RBCs) is known to be mediated by microRNAs (miRNAs). In erythroid cells, microR-214 (miR-214) has been reported to respond to external oxidative stress. However, the molecular mechanisms underlying this phenomenon remain unclear, especially during thalassemic erythropoiesis. In the present study, to further understand how miR-214 aggravates oxidative stress in thalassemia erythroid cells, we investigated the molecular mechanism of miR-214 and its regulation of the oxidative status in thalassemia erythrocytes. We have reported a biphasic expression of miR-214 in ß- and α-thalassemia. In the present study the effect of miR-214 expression was investigated by using miR -inhibitor and -mimic transfection in erythroid cell lines induced by hemin. Our study showed a biphasic expression of miR-214 in ß- and α-thalassemia. Subsequently, we examined the effect of miR-214 on erythroid differentiation in thalassemia. Our study reveals the loss-of-function of miR-214 during translational activation of activating transcription factor 4 mRNA, leading to decreased reactive oxygen species levels and increased glutathione levels in thalassemia erythroid cell. Our results suggest that the expression of activating transcription factor 4 regulated by miR-214 is important for oxidative stress modulation in thalassemic erythroid cells. Our findings can help to better understand the molecular mechanism of miRNA and transcription factors in regulation of oxidative status in erythroid cells, particularly in thalassemia, and could be useful for managing and relieving severe anemia symptoms in patients in the future.


Subject(s)
MicroRNAs , alpha-Thalassemia , beta-Thalassemia , Humans , Activating Transcription Factor 4/metabolism , Oxidative Stress/genetics , Erythroid Cells/metabolism , beta-Thalassemia/pathology , MicroRNAs/metabolism , Iron
14.
J Cell Mol Med ; 28(9): e18308, 2024 May.
Article in English | MEDLINE | ID: mdl-38683131

ABSTRACT

Destruction of erythropoiesis process leads to various diseases, including thrombocytopenia, anaemia, and leukaemia. miR-429-CT10 regulation of kinase-like (CRKL) axis involved in development, progression and metastasis of cancers. However, the exact role of miR-429-CRKL axis in leukaemic cell differentiation are still unknown. The current work aimed to uncover the effect of miR-429-CRKL axis on erythropoiesis. In the present study, CRKL upregulation was negatively correlated with miR-429 downregulation in both chronic myeloid leukaemia (CML) patient and CR patient samples. Moreover, CRKL expression level was significantly decreased while miR-429 expression level was increased during the erythroid differentiation of K562 cells following hemin treatment. Functional investigations revealed that overexpression and knockdown of CRKL was remarkably effective in suppressing and promoting hemin-induced erythroid differentiation of K562 cells, whereas, miR-429 exhibited opposite effects to CRKL. Mechanistically, miR-429 regulates erythroid differentiation of K562 cells by downregulating CRKL via selectively targeting CRKL-3'-untranslated region (UTR) through Raf/MEK/ERK pathway. Conversely, CRKII had no effect on erythroid differentiation of K562 cells. Taken together, our data demonstrated that CRKL (but not CRKII) and miR-429 contribute to development, progression and erythropoiesis of CML, miR-429-CRKL axis regulates erythropoiesis of K562 cells via Raf/MEK/ERK pathway, providing novel insights into effective diagnosis and therapy for CML patients.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Erythroid Cells , Hemin , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , MicroRNAs , Proto-Oncogene Proteins c-crk , Humans , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation/drug effects , Erythroid Cells/metabolism , Erythroid Cells/drug effects , Erythroid Cells/pathology , Erythroid Cells/cytology , Erythropoiesis/genetics , Erythropoiesis/drug effects , Gene Expression Regulation, Leukemic/drug effects , Hemin/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , MAP Kinase Signaling System/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-crk/metabolism , Proto-Oncogene Proteins c-crk/genetics
15.
Exp Hematol ; 135: 104191, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38493949

ABSTRACT

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


Subject(s)
ATP-Binding Cassette Transporters , Cell Differentiation , Erythropoiesis , Hematopoietic Stem Cells , Mice, Knockout , Mitochondria , Animals , Mice , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Mitochondria/metabolism , Erythropoiesis/genetics , Iron/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Oxidative Stress , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/cytology , Mice, Inbred C57BL
16.
Curr Opin Hematol ; 31(3): 71-81, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38415349

ABSTRACT

PURPOSE OF REVIEW: In this review, we present an overview of recent studies of primitive erythropoiesis, focusing on advances in deciphering its embryonic origin, defining species-specific differences in its developmental regulation, and better understanding the molecular and metabolic pathways involved in terminal differentiation. RECENT FINDINGS: Single-cell transcriptomics combined with state-of-the-art lineage tracing approaches in unperturbed murine embryos have yielded new insights concerning the origin of the first (primitive) erythroid cells that arise from mesoderm-derived progenitors. Moreover, studies examining primitive erythropoiesis in rare early human embryo samples reveal an overall conservation of primitive erythroid ontogeny in mammals, albeit with some interesting differences such as localization of erythropoietin (EPO) production in the early embryo. Mechanistically, the repertoire of transcription factors that critically regulate primitive erythropoiesis has been expanded to include regulators of transcription elongation, as well as epigenetic modifiers such as the histone methyltransferase DOT1L. For the latter, noncanonical roles aside from enzymatic activity are being uncovered. Lastly, detailed surveys of the metabolic and proteomic landscape of primitive erythroid precursors reveal the activation of key metabolic pathways such as pentose phosphate pathway that are paralleled by a striking loss of mRNA translation machinery. SUMMARY: The ability to interrogate single cells in vivo continues to yield new insights into the birth of the first essential organ system of the developing embryo. A comparison of the regulation of primitive and definitive erythropoiesis, as well as the interplay of the different layers of regulation - transcriptional, epigenetic, and metabolic - will be critical in achieving the goal of faithfully generating erythroid cells in vitro for therapeutic purposes.


Subject(s)
Erythropoiesis , Proteomics , Mice , Humans , Animals , Erythropoiesis/genetics , Erythroid Cells , Transcription Factors/genetics , Gene Expression Regulation, Developmental , Mammals/genetics
17.
Br J Haematol ; 204(5): 2086-2096, 2024 May.
Article in English | MEDLINE | ID: mdl-38296352

ABSTRACT

Morphological dysplasia in haematopoietic cells, defined by a 10% threshold in each lineage, is one of the diagnostic criteria for myelodysplastic neoplasms. Dysplasia limited to the erythroid lineage has also been reported in some cases of aplastic anaemia (AA); however, its significance remains unclear. We herein examined the impact of erythroid dysplasia on immunosuppressive therapy responses and survival in AA patients. The present study included 100 eligible AA patients without ring sideroblasts. Among them, 32 had dysplasia in the erythroid lineage (AA with minimal dysplasia [mini-D]). No significant sex or age differences were observed between AA groups with and without erythroid dysplasia. In severe/very severe AA and non-severe AA patients, a response to anti-thymocyte globulin + ciclosporin within 12 months was observed in 80.0% and 60.0% of AA with mini-D and 42.9% and 90.0% of those without dysplasia, with no significant difference (p = 0.29 and p = 0.24 respectively). Overall survival and leukaemia-free survival did not significantly differ between the groups. Collectively, the present results indicate that the presence of erythroid dysplasia did not significantly affect clinical characteristics or outcomes in AA patients, suggesting that its presence in AA is acceptable. Therefore, erythroid dysplasia should not exclude an AA diagnosis.


Subject(s)
Anemia, Aplastic , Registries , Humans , Anemia, Aplastic/mortality , Anemia, Aplastic/pathology , Anemia, Aplastic/drug therapy , Female , Male , Middle Aged , Adult , Aged , Young Adult , Erythroid Cells/pathology , Adolescent , Aged, 80 and over
18.
Nat Commun ; 15(1): 538, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38225226

ABSTRACT

Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.


Subject(s)
Anemia , Dioxygenases , Humans , Spleen , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Iron/metabolism , Anemia/metabolism , Erythroid Cells , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism
19.
Blood Cells Mol Dis ; 104: 102792, 2024 01.
Article in English | MEDLINE | ID: mdl-37633023

ABSTRACT

Sickle cell disease (SCD) is the most common ß-hemoglobinopathy caused by various mutations in the adult ß-globin gene resulting in sickle hemoglobin production, chronic hemolytic anemia, pain, and progressive organ damage. The best therapeutic strategies to manage the clinical symptoms of SCD is the induction of fetal hemoglobin (HbF) using chemical agents. At present, among the Food and Drug Administration-approved drugs to treat SCD, hydroxyurea is the only one proven to induce HbF protein synthesis, however, it is not effective in all people. Therefore, we evaluated the ability of the novel Bach1 inhibitor, HPP-D to induce HbF in KU812 cells and primary sickle erythroid progenitors. HPP-D increased HbF and decreased Bach1 protein levels in both cell types. Furthermore, chromatin immunoprecipitation assay showed reduced Bach1 and increased NRF2 binding to the γ-globin promoter antioxidant response elements. We also observed increased levels of the active histone marks H3K4Me1 and H3K4Me3 supporting an open chromatin configuration. In primary sickle erythroid progenitors, HPP-D increased γ-globin transcription and HbF positive cells and reduced sickled erythroid progenitors under hypoxia conditions. Collectively, our data demonstrate that HPP-D induces γ-globin gene transcription through Bach1 inhibition and enhanced NRF2 binding in the γ-globin promoter antioxidant response elements.


Subject(s)
Anemia, Sickle Cell , gamma-Globins , Humans , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , Hemoglobin, Sickle/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/therapeutic use , Transcriptional Activation/drug effects , Erythroid Cells/drug effects , Erythroid Cells/metabolism
20.
Iberoam. j. med ; 6(1): 3-9, 2024. tab
Article in English | IBECS | ID: ibc-229284

ABSTRACT

Introduction: Studies on nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) levels in COVID-19 patients are limited. This study aimed to investigate the relationship between some biomarkers of oxidant-antioxidant status with COVID-19 disease. Material and methods: The patients older than 18 years of age who tested positive for SARS CoV-2 PCR (polymerase chain reaction) with clinical symptoms and signs were included in this study. Total antioxidant status (TAS), total antioxidant status (TOS), oxidative stress index (OSI) and HO-1 and Nrf2 levels were analyzed from serum samples taken before and after treatment. Results: In this study, 16 patients followed up with the diagnosis of COVID-19 were included. 9 (56.3%) of the patients were female and 7 (43.8%) were male. The mean age was 33.75 ± 17.03 years. All patients were symptomatic and were hospitalized to be followed up. It was determined that Nrf2 and HO-1 values increased significantly after treatment. Moreover, there was a significant positive correlation between Nrf2 and TAS values and TAS increases significantly in parallel to an increase in Nrf2, and there was a significant but negative correlation between Nrf2 and TOS and OSI values, and thus an increase in Nrf2 led to a decrease in TOS and OSI values. There was a significant positive correlation between HO-1 and TAS, and TAS increased significantly, as HO-1 increased. Conclusions: The decrease in TOS and OSI and the increase in Nrf2 and HO-1 during the follow-up period in COVID-19 patients suggest that the body tries to prevent ROS-related oxidative stress via Nrf2 and HO-1 and that oxidative stress may have a key role in the pathophysiology of COVID-19. (AU)


Introducción: Los estudios sobre los niveles del factor 2 relacionado con el factor nuclear eritroide 2 (Nrf2) y la hemo oxigenasa-1 (HO-1) en pacientes con COVID-19 son limitados. Este estudio tuvo como objetivo investigar la relación entre algunos biomarcadores del estado oxidante-antioxidante con la enfermedad COVID-19. Material y métodos: Se incluyeron en este estudio los pacientes mayores de 18 años que dieron positivo a PCR (reacción en cadena de la polimerasa) de SARS CoV-2 con síntomas y signos clínicos. Se analizaron el estado antioxidante total (TAS), el estado antioxidante total (TOS), el índice de estrés oxidativo (OSI) y los niveles de HO-1 y Nrf2 a partir de muestras de suero tomadas antes y después del tratamiento. Resultados: En este estudio se incluyeron 16 pacientes seguidos con diagnóstico de COVID-19. 9 (56,3%) de los pacientes eran mujeres y 7 (43,8%) eran hombres. La edad media fue 33,75 ± 17,03 años. Todos los pacientes presentaban síntomas y fueron hospitalizados para seguimiento. Se determinó que los valores de Nrf2 y HO-1 aumentaron significativamente después del tratamiento. Además, hubo una correlación positiva significativa entre los valores de Nrf2 y TAS y TAS aumenta significativamente en paralelo a un aumento en Nrf2, y también hubo una correlación significativa pero negativa entre Nrf2 y los valores de TOS y OSI y, por lo tanto, un aumento en Nrf2 condujo a una disminución en los valores TOS y OSI. Hubo una correlación positiva significativa entre HO-1 y TAS, y TAS aumentó significativamente a medida que aumentaba HO-1. Conclusiones: La disminución de TOS y OSI y el aumento de Nrf2 y HO-1 durante el período de seguimiento en pacientes con COVID-19 sugieren que el cuerpo intenta prevenir el estrés oxidativo relacionado con ROS a través de Nrf2 y HO-1 y que el estrés oxidativo puede tener un papel clave en la fisiopatología de COVID-19. (AU)


Subject(s)
Humans , Male , Female , Adolescent , Young Adult , Adult , Biomarkers/analysis , Oxidants , Antioxidants , Heme Oxygenase-1 , Erythroid Cells
SELECTION OF CITATIONS
SEARCH DETAIL