Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 137
1.
Hemoglobin ; 48(1): 4-14, 2024 Jan.
Article En | MEDLINE | ID: mdl-38419555

Long noncoding RNAs (lncRNAs) are important because they are involved in a variety of life activities and have many downstream targets. Moreover, there is also increasing evidence that some lncRNAs play important roles in the expression and regulation of γ-globin genes. In our previous study, we analyzed genetic material from nucleated red blood cells (NRBCs) extracted from premature and full-term umbilical cord blood samples. Through RNA sequencing (RNA-Seq) analysis, lncRNA H19 emerged as a differentially expressed transcript between the two blood types. While this discovery provided insight into H19, previous studies had not investigated its effect on the γ-globin gene. Therefore, the focus of our study was to explore the impact of H19 on the γ-globin gene. In this study, we discovered that overexpressing H19 led to a decrease in HBG mRNA levels during erythroid differentiation in K562 cells. Conversely, in CD34+ hematopoietic stem cells and human umbilical cord blood-derived erythroid progenitor (HUDEP-2) cells, HBG expression increased. Additionally, we observed that H19 was primarily located in the nucleus of K562 cells, while in HUDEP-2 cells, H19 was present predominantly in the cytoplasm. These findings suggest a significant upregulation of HBG due to H19 overexpression. Notably, cytoplasmic localization in HUDEP-2 cells hints at its potential role as a competing endogenous RNA (ceRNA), regulating γ-globin expression by targeting microRNA/mRNA interactions.


RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Up-Regulation , RNA, Messenger/genetics , Gene Expression
2.
Biochem Biophys Res Commun ; 701: 149555, 2024 Mar 15.
Article En | MEDLINE | ID: mdl-38325179

Fetal-to-adult hemoglobin switching is controlled by programmed silencing of γ-globin while the re-activation of fetal hemoglobin (HbF) is an effective strategy for ameliorating the clinical severity of ß-thalassemia and sickle cell disease. The identification of enhancer RNAs (eRNAs) related to the fetal (α2γ2) to adult hemoglobin (α2ß2) switching remains incomplete. In this study, the transcriptomes of GYPA+ cells from six ß-thalassemia patients with extreme HbF levels were sequenced to identify differences in patterns of noncoding RNA expression. It is interesting that an enhancer upstream of CHD4, an HbF-related core subunit of the NuRD complex, was differentially transcribed. We found a significantly positive correlation of eRNA-CHD4 enhancer-gene interaction using the public database of FANTOM5. Specifically, the eRNA-CHD4 expression was found to be significantly higher in both CD34+ HSPCs and HUDEP-2 than those in K562 cells which commonly expressed high level of HbF, suggesting a correlation between eRNA and HbF expression. Furthermore, prediction of transcription binding sites of cis-eQTLs and the CHD4 genomic region revealed a putative interaction site between rs73264846 and ZNF410, a known transcription factor regulating HbF expression. Moreover, in-vitro validation showed that the inhibition of eRNA could reduce the expression of HBG expression in HUDEP-2 cells. Taken together, the findings of this study demonstrate that a distal enhancer contributes to stage-specific silencing of γ-globin genes through direct modulation of CHD4 expression and provide insights into the epigenetic mechanisms of NuRD-mediated hemoglobin switching.


Anemia, Sickle Cell , beta-Thalassemia , Adult , Humans , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , beta-Thalassemia/genetics , Gene Expression Regulation , Anemia, Sickle Cell/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism
3.
Blood ; 143(19): 1980-1991, 2024 May 09.
Article En | MEDLINE | ID: mdl-38364109

ABSTRACT: The switch from fetal hemoglobin (γ-globin, HBG) to adult hemoglobin (ß-globin, HBB) gene transcription in erythroid cells serves as a paradigm for a complex and clinically relevant developmental gene regulatory program. We previously identified HIC2 as a regulator of the switch by inhibiting the transcription of BCL11A, a key repressor of HBG production. HIC2 is highly expressed in fetal cells, but the mechanism of its regulation is unclear. Here we report that HIC2 developmental expression is controlled by microRNAs (miRNAs), as loss of global miRNA biogenesis through DICER1 depletion leads to upregulation of HIC2 and HBG messenger RNA. We identified the adult-expressed let-7 miRNA family as a direct posttranscriptional regulator of HIC2. Ectopic expression of let-7 in fetal cells lowered HIC2 levels, whereas inhibition of let-7 in adult erythroblasts increased HIC2 production, culminating in decommissioning of a BCL11A erythroid enhancer and reduced BCL11A transcription. HIC2 depletion in let-7-inhibited cells restored BCL11A-mediated repression of HBG. Together, these data establish that fetal hemoglobin silencing in adult erythroid cells is under the control of a miRNA-mediated inhibitory pathway (let-7 ⊣ HIC2 ⊣ BCL11A ⊣ HBG).


Fetal Hemoglobin , Kruppel-Like Transcription Factors , MicroRNAs , Repressor Proteins , Humans , beta-Globins/genetics , beta-Globins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Erythroblasts/metabolism , Erythroblasts/cytology , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Transcription, Genetic
4.
Clin Epigenetics ; 16(1): 12, 2024 01 13.
Article En | MEDLINE | ID: mdl-38218889

The mechanism that drives the switch from fetal to adult hemoglobin (Hb) provides a therapeutic target for ß-thalassemia. We have previously identified that hypermethylation of transcription factor ERF promoter reactivated γ-globin expression. To uncover the mechanism underlying the hypermethylation of ERF promoter, we performed RNA sequencing in ß0/ß0-thalassemia patients and identified an upregulated long noncoding RNA (RP11-196G18.23) associated with HbF production. RP11-196G18.23 bound to the ERF promoter and recruited DNA methyltransferase 3A to promote DNA hypermethylation-mediated ERF downregulation, thereby ameliorating ERF-induced γ-globin inactivation. The identification of RP11-196G18.23 provides an epigenetic mechanism for the reactivation of fetal γ-globin expression for ß-hemoglobinopathies.


RNA, Long Noncoding , beta-Thalassemia , Adult , Humans , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/genetics , gamma-Globins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Fetal Hemoglobin/genetics , DNA Methylation , Repressor Proteins/genetics
5.
J Mol Biol ; 436(7): 168343, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-37924864

In humans, specific aberrations in ß-globin results in sickle cell disease and ß-thalassemia, symptoms of which can be ameliorated by increased expression of fetal globin (HbF). Two recent CRISPR-Cas9 screens, centered on ∼1500 annotated sequence-specific DNA binding proteins and performed in a human erythroid cell line that expresses adult hemoglobin, uncovered four groups of candidate regulators of HbF gene expression. They are (1) members of the nucleosome remodeling and deacetylase (NuRD) complex proteins that are already known for HbF control; (2) seven C2H2 zinc finger (ZF) proteins, including some (ZBTB7A and BCL11A) already known for directly silencing the fetal γ-globin genes in adult human erythroid cells; (3) a few other transcription factors of different structural classes that might indirectly influence HbF gene expression; and (4) DNA methyltransferase 1 (DNMT1) that maintains the DNA methylation marks that attract the MBD2-associated NuRD complex to DNA as well as associated histone H3 lysine 9 methylation. Here we briefly discuss the effects of these regulators, particularly C2H2 ZFs, in inducing HbF expression for treating ß-hemoglobin disorders, together with recent advances in developing safe and effective small-molecule therapeutics for the regulation of this well-conserved hemoglobin switch.


CYS2-HIS2 Zinc Fingers , Hemoglobinopathies , Humans , Cell Line, Tumor , DNA , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Hemoglobinopathies/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Sci Rep ; 13(1): 21997, 2023 12 11.
Article En | MEDLINE | ID: mdl-38081985

Adults with sickle cell disease bear a mutation in the ß-globin gene, leading to the expression of sickle hemoglobin (HbS; α2ßS2). Adults also possess the gene for γ-globin, which is a component of fetal hemoglobin (HbF, α2γ2); however, γ-chain expression normally ceases after birth. As HbF does not form the fibers that cause the disease, pharmacological and gene-modifying interventions have attempted to either reactivate expression of the γ chain or introduce a gene encoding a modified ß chain having γ-like character. Here, we show that a single-site modification on the α chain, αPro114Arg, retards fiber formation as effectively as HbF. Because this addition to the repertoire of anti-sickling approaches acts independently of other modifications, it could be coupled with other therapies to significantly enhance their effectiveness.


Anemia, Sickle Cell , Fetal Hemoglobin , Adult , Humans , Fetal Hemoglobin/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/drug therapy , gamma-Globins/genetics , gamma-Globins/metabolism , Hemoglobin, Sickle/genetics
7.
Cell Stem Cell ; 30(12): 1624-1639.e8, 2023 12 07.
Article En | MEDLINE | ID: mdl-37989316

Reactivating silenced γ-globin expression through the disruption of repressive regulatory domains offers a therapeutic strategy for treating ß-hemoglobinopathies. Here, we used transformer base editor (tBE), a recently developed cytosine base editor with no detectable off-target mutations, to disrupt transcription-factor-binding motifs in hematopoietic stem cells. By performing functional screening of six motifs with tBE, we found that directly disrupting the BCL11A-binding motif in HBG1/2 promoters triggered the highest γ-globin expression. Via a side-by-side comparison with other clinical and preclinical strategies using Cas9 nuclease or conventional BEs (ABE8e and hA3A-BE3), we found that tBE-mediated disruption of the BCL11A-binding motif at the HBG1/2 promoters triggered the highest fetal hemoglobin in healthy and ß-thalassemia patient hematopoietic stem/progenitor cells while exhibiting no detectable DNA or RNA off-target mutations. Durable therapeutic editing by tBE persisted in repopulating hematopoietic stem cells, demonstrating that tBE-mediated editing in HBG1/2 promoters is a safe and effective strategy for treating ß-hemoglobinopathies.


Gene Editing , Hemoglobinopathies , Humans , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , CRISPR-Cas Systems , Mutation/genetics , Hemoglobinopathies/genetics , Hemoglobinopathies/metabolism , Hematopoietic Stem Cells/metabolism , Transcription Factors/metabolism
8.
Genes (Basel) ; 14(10)2023 10 11.
Article En | MEDLINE | ID: mdl-37895276

The anticancer drug mithramycin (MTH), has been proposed for drug repurposing after the finding that it is a potent inducer of fetal hemoglobin (HbF) production in erythroid precursor cells (ErPCs) from ß-thalassemia patients. In this respect, previously published studies indicate that MTH is very active in inducing increased expression of γ-globin genes in erythroid cells. This is clinically relevant, as it is firmly established that HbF induction is a valuable approach for the therapy of ß-thalassemia and for ameliorating the clinical parameters of sickle-cell disease (SCD). Therefore, the identification of MTH biochemical/molecular targets is of great interest. This study is inspired by recent robust evidence indicating that the expression of γ-globin genes is controlled in adult erythroid cells by different transcriptional repressors, including Oct4, MYB, BCL11A, Sp1, KLF3 and others. Among these, BCL11A is very important. In the present paper we report evidence indicating that alterations of BCL11A gene expression and biological functions occur during MTH-mediated erythroid differentiation. Our study demonstrates that one of the mechanisms of action of MTH is a down-regulation of the transcription of the BCL11A gene, while a second mechanism of action is the inhibition of the molecular interactions between the BCL11A complex and specific sequences of the γ-globin gene promoter.


beta-Thalassemia , gamma-Globins , Humans , gamma-Globins/genetics , gamma-Globins/metabolism , beta-Thalassemia/genetics , Plicamycin/pharmacology , Repressor Proteins/genetics , Transcription Factors/genetics , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Expression , Kruppel-Like Transcription Factors/genetics
9.
Bioorg Chem ; 140: 106768, 2023 11.
Article En | MEDLINE | ID: mdl-37586133

Pharmacological induction of fetal hemoglobin has proven to be a promising therapeutic intervention in ß-hemoglobinopathies by reducing the globin chain imbalance and inhibiting sickle cell polymerization. Fagonia indica has shown therapeutic relevance to ß-thalassemia. Therefore, we study the ethnopharmacological potential of Fagonia indica and its biomarker compounds for their HbF induction ability for the treatment of ß-thalassemia. Here, we identify, compound 8 (triterpenoid glycosides) of F. indica. as a prominent HbF inducer in-vitro and in-vivo. Compound 8 showed potent erythroid differentiation, enhanced cellular proliferation, ample accumulation of total hemoglobin, and a strong notion of γ-globin gene expression in K562 cultures. Compound 8 treatment also revealed strong induction of erythroid differentiation and fetal hemoglobin mRNA and protein in adult erythroid precursor cells. This induction was associated with simultaneous downregulation of BCL11A and SOX6, and overexpression of the GATA-1 gene, suggesting a compound 8-mediated partial mechanism involved in the reactivation of fetal-like globin genes. The in vivo study with compound 8 (10 mg/kg) in ß-YAC mice resulted in significant HbF synthesis demonstrated by the enhanced level of F-cells (84.14 %) and an 8.85-fold increase in the γ-globin gene. Overall, the study identifies compound 8 as a new HbF-inducing entity and provides an early "proof-of-concept" to enable the initiation of preclinical and clinical studies in the development of this HbF-inducing agent for ß-thalassemia.


Hemoglobinopathies , Triterpenes , beta-Thalassemia , Humans , Animals , Mice , gamma-Globins/genetics , gamma-Globins/metabolism , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , Glycosides/pharmacology , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , K562 Cells , Transcription Factors , Gene Expression , Repressor Proteins
10.
Nat Genet ; 55(7): 1210-1220, 2023 07.
Article En | MEDLINE | ID: mdl-37400614

Inducing fetal hemoglobin (HbF) in red blood cells can alleviate ß-thalassemia and sickle cell disease. We compared five strategies in CD34+ hematopoietic stem and progenitor cells, using either Cas9 nuclease or adenine base editors. The most potent modification was adenine base editor generation of γ-globin -175A>G. Homozygous -175A>G edited erythroid colonies expressed 81 ± 7% HbF versus 17 ± 11% in unedited controls, whereas HbF levels were lower and more variable for two Cas9 strategies targeting a BCL11A binding motif in the γ-globin promoter or a BCL11A erythroid enhancer. The -175A>G base edit also induced HbF more potently than a Cas9 approach in red blood cells generated after transplantation of CD34+ hematopoietic stem and progenitor cells into mice. Our data suggest a strategy for potent, uniform induction of HbF and provide insights into γ-globin gene regulation. More generally, we demonstrate that diverse indels generated by Cas9 can cause unexpected phenotypic variation that can be circumvented by base editing.


Anemia, Sickle Cell , beta-Thalassemia , Mice , Animals , gamma-Globins/genetics , gamma-Globins/metabolism , Gene Editing , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Anemia, Sickle Cell/genetics , Antigens, CD34/metabolism , beta-Thalassemia/genetics
11.
Int J Mol Sci ; 24(11)2023 May 31.
Article En | MEDLINE | ID: mdl-37298481

Beta-hemoglobinopathies are the most common genetic disorders worldwide, caused by a wide spectrum of mutations in the ß-globin locus, and associated with morbidity and early mortality in case of patient non-adherence to supportive treatment. Allogeneic transplantation of hematopoietic stem cells (allo-HSCT) used to be the only curative option, although the indispensable need for an HLA-matched donor markedly restricted its universal application. The evolution of gene therapy approaches made possible the ex vivo delivery of a therapeutic ß- or γ- globin gene into patient-derived hematopoietic stem cells followed by the transplantation of corrected cells into myeloablated patients, having led to high rates of transfusion independence (thalassemia) or complete resolution of painful crises (sickle cell disease-SCD). Hereditary persistence of fetal hemoglobin (HPFH), a syndrome characterized by increased γ-globin levels, when co-inherited with ß-thalassemia or SCD, converts hemoglobinopathies to a benign condition with mild clinical phenotype. The rapid development of precise genome editing tools (ZFN, TALENs, CRISPR/Cas9) over the last decade has allowed the targeted introduction of mutations, resulting in disease-modifying outcomes. In this context, genome editing tools have successfully been used for the introduction of HPFH-like mutations both in HBG1/HBG2 promoters or/and in the erythroid enhancer of BCL11A to increase HbF expression as an alternative curative approach for ß-hemoglobinopathies. The current investigation of new HbF modulators, such as ZBTB7A, KLF-1, SOX6, and ZNF410, further expands the range of possible genome editing targets. Importantly, genome editing approaches have recently reached clinical translation in trials investigating HbF reactivation in both SCD and thalassemic patients. Showing promising outcomes, these approaches are yet to be confirmed in long-term follow-up studies.


Anemia, Sickle Cell , Hemoglobinopathies , beta-Thalassemia , Humans , CRISPR-Cas Systems , Cell Line, Tumor , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Gene Editing/methods , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/therapy , beta-Thalassemia/metabolism
12.
Int J Mol Sci ; 24(1)2023 Jan 01.
Article En | MEDLINE | ID: mdl-36614221

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.


Erythroid Precursor Cells , beta-Thalassemia , Humans , beta-Thalassemia/drug therapy , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/analysis , gamma-Globins/genetics , gamma-Globins/metabolism , Nuclear Proteins/genetics , Polymorphism, Genetic
13.
Nat Prod Res ; 37(6): 985-989, 2023 Mar.
Article En | MEDLINE | ID: mdl-35776113

Hydroxyurea (HU) is an anti-cancer drug that is used for the treatment of hemoglobinopathies as a γ-globin inducer. However, its dose-dependent effects have hampered its clinical reliability. Resveratrol (RSV) is an antioxidant and γ-globin inducer. The present study aimed to assess their combined effects on the γ-globin gene expression and reactive oxygen species (ROS) level of K562 cells. The results indicated that the γ-globin gene expression was approximately two folds higher in the group treated with RSV 50 µM + HU 25 µM in comparison to HU 100 µM alone (***p < 0.001). However, there was an inverse relationship between the expression of γ-globin gene and HU concentration in the combined groups. Furthermore, the combinations of RSV and HU significantly reduced ROS levels compared to single drugs. Overall, the combination of these compounds was an appropriate strategy for increasing γ-globin expression, reducing oxidant levels, and alleviating the adverse effects of HU.


Hydroxyurea , gamma-Globins , Humans , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , gamma-Globins/genetics , gamma-Globins/metabolism , K562 Cells , Resveratrol/pharmacology , Reactive Oxygen Species , Reproducibility of Results , Gene Expression
14.
Hematology ; 27(1): 1152-1162, 2022 Dec.
Article En | MEDLINE | ID: mdl-36178486

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.


MicroRNAs , beta-Thalassemia , Apoptosis , Carrier Proteins/genetics , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Glutathione , Humans , Malondialdehyde/metabolism , Nuclear Proteins/genetics , Oxidative Stress , Reactive Oxygen Species/metabolism , Repressor Proteins/metabolism , Superoxide Dismutase , Transcription Factors/metabolism , beta-Thalassemia/genetics , gamma-Globins/genetics , gamma-Globins/metabolism
15.
J Immunol Res ; 2022: 8440422, 2022.
Article En | MEDLINE | ID: mdl-35942209

Reactivation of fetal hemoglobin (HbF, α2γ2) alleviates clinical symptoms in patients with ß-thalassemia and sickle cell disease, although the regulatory mechanisms of γ-globin expression have not yet been fully elucidated. Recent studies found that interfering with the expression of the membrane protein ANTXR1 gene upregulated γ-globin levels. However, the exact mechanism by which ANTXR1 regulates γ-globin levels remains unclear. Our study showed that overexpression and knockdown of ANTXR1 in K562, cord blood CD34+, and HUDEP-2 cells decreased and increased γ-globin expression, respectively. ANTXR1 regulates the reactivation of fetal hemoglobin (HbF, α2γ2) in K562, cord blood CD34+, and adult peripheral blood CD34+ cells through interaction with LRP6 to promote the nuclear entry of ß-catenin and activate the Wnt/ß-catenin signaling pathway. The overexpression or knockdown of ANTXR1 on γ-globin and Wnt/ß-catenin signaling in K562 cells was reversed by the inhibitor XAV939 and the activator LiCl, respectively, where XAV939 inhibits the transcription of ß-catenin in the Wnt pathway, but LiCl inhibits GSK3-ß. We also showed that the binding ability of the rank4 site in the transcriptional regulatory region of the SOX6 gene to c-Jun was significantly increased after overexpression of ANTXR1 in K562 cells. SOX6 protein expression was increased significantly after overexpression of the c-Jun gene, indicating that the transcription factor c-Jun initiated the transcription of SOX6, thereby silencing γ-globin. Our findings may provide a new intervention target for the treatment of ß-hemoglobinopathies.


Fetal Hemoglobin , gamma-Globins , Adult , Antigens, CD34 , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Membrane Proteins/genetics , Microfilament Proteins/metabolism , Receptors, Cell Surface , Wnt Signaling Pathway , beta Catenin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
16.
Int J Mol Sci ; 23(13)2022 Jun 24.
Article En | MEDLINE | ID: mdl-35806029

The hemoglobin switch from fetal (HbF) to adult (HbA) has been studied intensively as an essential model for gene expression regulation, but also as a beneficial therapeutic approach for ß-hemoglobinopathies, towards the objective of reactivating HbF. The transcription factor LRF (Leukemia/lymphoma-related), encoded from the ZBTB7A gene has been implicated in fetal hemoglobin silencing, though has a wide range of functions that have not been fully clarified. We thus established the LRF/ZBTB7A-overexpressing and ZBTB7A-knockdown K562 (human erythroleukemia cell line) clones to assess fetal vs. adult hemoglobin production pre- and post-induction. Transgenic K562 clones were further developed and studied under the influence of epigenetic chromatin regulators, such as DNA methyl transferase 3 (DNMT3) and Histone Deacetylase 1 (HDAC1), to evaluate LRF's potential disturbance upon the aberrant epigenetic background and provide valuable information of the preferable epigenetic frame, in which LRF unfolds its action on the ß-type globin's expression. The ChIP-seq analysis demonstrated that LRF binds to γ-globin genes (HBG2/1) and apparently associates BCL11A for their silencing, but also during erythropoiesis induction, LRF binds the BGLT3 gene, promoting BGLT3-lncRNA production through the γ-δ intergenic region of ß-type globin's locus, triggering the transcriptional events from γ- to ß-globin switch. Our findings are supported by an up-to-date looping model, which highlights chromatin alterations during erythropoiesis at late stages of gestation, to establish an "open" chromatin conformation across the γ-δ intergenic region and accomplish ß-globin expression and hemoglobin switch.


RNA, Long Noncoding , Transcription Factors , Adult , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , DNA, Intergenic/genetics , DNA, Intergenic/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Hemoglobin A/genetics , Hemoglobin A/metabolism , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
17.
Blood Adv ; 6(23): 6016-6022, 2022 12 13.
Article En | MEDLINE | ID: mdl-35667093

The fetal-to-adult hemoglobin switching at about the time of birth involves a shift in expression from γ-globin to ß-globin in erythroid cells. Effective re-expression of fetal γ-globin can ameliorate sickle cell anemia and ß-thalassemia. Despite the physiological and clinical relevance of this switch, its posttranscriptional regulation is poorly understood. Here, we identify Pumilo 1 (PUM1), an RNA-binding protein with no previously reported functions in erythropoiesis, as a direct posttranscriptional regulator of ß-globin switching. PUM1, whose expression is regulated by the erythroid master transcription factor erythroid Krüppel-like factor (EKLF/KLF1), peaks during erythroid differentiation, binds γ-globin messenger RNA (mRNA), and reduces γ-globin (HBG1) mRNA stability and translational efficiency, which culminates in reduced γ-globin protein levels. Knockdown of PUM1 leads to a robust increase in fetal hemoglobin (∼22% HbF) without affecting ß-globin levels in human erythroid cells. Importantly, targeting PUM1 does not limit the progression of erythropoiesis, which provides a potentially safe and effective treatment strategy for sickle cell anemia and ß-thalassemia. In support of this idea, we report elevated levels of HbF in the absence of anemia in an individual with a novel heterozygous PUM1 mutation in the RNA-binding domain (p.(His1090Profs∗16); c.3267_3270delTCAC), which suggests that PUM1-mediated posttranscriptional regulation is a critical player during human hemoglobin switching.


Anemia, Sickle Cell , beta-Thalassemia , Adult , Humans , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , beta-Thalassemia/genetics , beta-Globins/genetics , Carrier Proteins , Anemia, Sickle Cell/genetics , RNA-Binding Proteins/genetics
18.
Nat Genet ; 54(6): 874-884, 2022 06.
Article En | MEDLINE | ID: mdl-35618846

The mechanisms by which the fetal-type ß-globin-like genes HBG1 and HBG2 are silenced in adult erythroid precursor cells remain a fundamental question in human biology and have therapeutic relevance to sickle cell disease and ß-thalassemia. Here, we identify via a CRISPR-Cas9 genetic screen two members of the NFI transcription factor family-NFIA and NFIX-as HBG1/2 repressors. NFIA and NFIX are expressed at elevated levels in adult erythroid cells compared with fetal cells, and function cooperatively to repress HBG1/2 in cultured cells and in human-to-mouse xenotransplants. Genomic profiling, genome editing and DNA binding assays demonstrate that the potent concerted activity of NFIA and NFIX is explained in part by their ability to stimulate the expression of BCL11A, a known silencer of the HBG1/2 genes, and in part by directly repressing the HBG1/2 genes. Thus, NFI factors emerge as versatile regulators of the fetal-to-adult switch in ß-globin production.


Fetal Hemoglobin , gamma-Globins , Animals , Carrier Proteins/genetics , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Editing , Mice , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism , Transcription Factors/genetics , beta-Globins/genetics , beta-Globins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism
19.
Hemoglobin ; 46(3): 153-159, 2022 May.
Article En | MEDLINE | ID: mdl-35506261

Hydroxyurea (HU) is an effective drug to increase fetal γ-globin gene (Hb F) expression, replacing the missing adult ß-globin gene. The mechanism of Hb F induction by HU and improvement in clinical symptoms are still poorly understood. The current study aimed to improve the molecular understanding of drug-induced alterations and reveals genes related to HU treatment responsiveness in ß-thalassemia (ß-thal). We analyzed the GSE109186 dataset using system biology and weighted gene coexpression network analysis (WGCNA) to identify and quantify gene expression changes reflected in the HU-treated human erythroblastic leukemia cells. The K562 cell line was treated in 50, 100, and 150 µM concentrations of HU for 24, 48, and 72 hours with three replications. The alteration of CA1, LIN28B and Hb F gene expression in HU-treated cells was evaluated using the real-time polymerase chain (real-time PCR) technique. The results showed that LIN28B has an increase of 4.27-fold on the first day of HU-treatment in 50 µM (p < 0.01). The CA1 expression showed a decrease at all times and doses of treatment, and the most decrease happened in 48 hours and 50 µM (p < 0.04). Hb F also showed the highest increase in 100 µM after 24 hours of treatment (5.18-fold). In summary, the data suggest that alteration of LIN28B and CA1 gene expression is associated with γ-globin increasing in HU-treated cells.


Fetal Hemoglobin , beta-Thalassemia , Adult , Fetal Hemoglobin/analysis , Humans , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , RNA-Binding Proteins/therapeutic use , beta-Globins/genetics , beta-Thalassemia/genetics , gamma-Globins/metabolism
20.
Blood Adv ; 6(11): 3280-3285, 2022 06 14.
Article En | MEDLINE | ID: mdl-35240686

Human γ-globin is predominantly expressed in fetal liver erythroid cells during gestation from 2 nearly identical genes, HBG1 and HBG2, that are both perinatally silenced. Reactivation of these fetal genes in adult red blood cells can ameliorate many symptoms associated with the inherited ß-globinopathies, sickle cell disease, and Cooley anemia. Although promising genetic strategies to reactivate the γ-globin genes to treat these diseases have been explored, there are significant barriers to their effective implementation worldwide; alternatively, pharmacological induction of γ-globin synthesis could readily reach the majority of affected individuals. In this study, we generated a CRISPR knockout library that targeted all erythroid genes for which prospective or actual therapeutic compounds already exist. By probing this library for genes that repress fetal hemoglobin (HbF), we identified several novel, potentially druggable, γ-globin repressors, including VHL and PTEN. We demonstrate that deletion of VHL induces HbF through activation of the HIF1α pathway and that deletion of PTEN induces HbF through AKT pathway stimulation. Finally, we show that small-molecule inhibitors of PTEN and EZH induce HbF in both healthy and ß-thalassemic human primary erythroid cells.


beta-Thalassemia , gamma-Globins , Adult , Erythroid Cells/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Humans , Prospective Studies , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/genetics , gamma-Globins/metabolism
...