Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29606353

ABSTRACT

Fetal hemoglobin (HbF, α2γ2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2ß2) disorders, sickle cell disease, and ß-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from γ- to ß- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in γ-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. Our findings reveal that direct γ-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Subject(s)
Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Nuclear Proteins/metabolism , Base Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Editing , Humans , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins , Zinc Fingers/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/pathology , gamma-Globins/genetics
2.
Genes Dev ; 32(23-24): 1537-1549, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30463901

ABSTRACT

Human globin gene production transcriptionally "switches" from fetal to adult synthesis shortly after birth and is controlled by macromolecular complexes that enhance or suppress transcription by cis elements scattered throughout the locus. The DRED (direct repeat erythroid-definitive) repressor is recruited to the ε-globin and γ-globin promoters by the orphan nuclear receptors TR2 (NR2C1) and TR4 (NR2C2) to engender their silencing in adult erythroid cells. Here we found that nuclear receptor corepressor-1 (NCoR1) is a critical component of DRED that acts as a scaffold to unite the DNA-binding and epigenetic enzyme components (e.g., DNA methyltransferase 1 [DNMT1] and lysine-specific demethylase 1 [LSD1]) that elicit DRED function. We also describe a potent new regulator of γ-globin repression: The deubiquitinase BRCA1-associated protein-1 (BAP1) is a component of the repressor complex whose activity maintains NCoR1 at sites in the ß-globin locus, and BAP1 inhibition in erythroid cells massively induces γ-globin synthesis. These data provide new mechanistic insights through the discovery of novel epigenetic enzymes that mediate γ-globin gene repression.


Subject(s)
Gene Expression Regulation/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , gamma-Globins/genetics , Binding Sites , Cell Line , Enzyme Activation/genetics , Epigenesis, Genetic/genetics , Erythroid Cells/metabolism , Gene Silencing , HEK293 Cells , Humans , K562 Cells , Nuclear Receptor Subfamily 2, Group C, Member 1/metabolism , Protein Domains , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism
3.
Genes Dev ; 31(16): 1704-1713, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28916711

ABSTRACT

Chromatin structure is tightly intertwined with transcription regulation. Here we compared the chromosomal architectures of fetal and adult human erythroblasts and found that, globally, chromatin structures and compartments A/B are highly similar at both developmental stages. At a finer scale, we detected distinct folding patterns at the developmentally controlled ß-globin locus. Specifically, new fetal stage-specific contacts were uncovered between a region separating the fetal (γ) and adult (δ and ß) globin genes (encompassing the HBBP1 and BGLT3 noncoding genes) and two distal chromosomal sites (HS5 and 3'HS1) that flank the locus. In contrast, in adult cells, the HBBP1-BGLT3 region contacts the embryonic ε-globin gene, physically separating the fetal globin genes from the enhancer (locus control region [LCR]). Deletion of the HBBP1 region in adult cells alters contact landscapes in ways more closely resembling those of fetal cells, including increased LCR-γ-globin contacts. These changes are accompanied by strong increases in γ-globin transcription. Notably, the effects of HBBP1 removal on chromatin architecture and gene expression closely mimic those of deleting the fetal globin repressor BCL11A, implicating BCL11A in the function of the HBBP1 region. Our results uncover a new critical regulatory region as a potential target for therapeutic genome editing for hemoglobinopathies and highlight the power of chromosome conformation analysis in discovering new cis control elements.


Subject(s)
Chromatin/chemistry , Erythroblasts/metabolism , Gene Expression Regulation, Developmental , Regulatory Elements, Transcriptional , beta-Globins/genetics , Adult , Carrier Proteins/genetics , Fetus , Gene Silencing , Humans , Locus Control Region , Nuclear Proteins/genetics , Pseudogenes , Repressor Proteins , Transcriptome , gamma-Globins/genetics
4.
Am J Hum Genet ; 108(4): 709-721, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33735615

ABSTRACT

The fetal-to-adult hemoglobin switch is regulated in a developmental stage-specific manner and reactivation of fetal hemoglobin (HbF) has therapeutic implications for treatment of ß-thalassemia and sickle cell anemia, two major global health problems. Although significant progress has been made in our understanding of the molecular mechanism of the fetal-to-adult hemoglobin switch, the mechanism of epigenetic regulation of HbF silencing remains to be fully defined. Here, we performed whole-genome bisulfite sequencing and RNA sequencing analysis of the bone marrow-derived GYPA+ erythroid cells from ß-thalassemia-affected individuals with widely varying levels of HbF groups (HbF ≥ 95th percentile or HbF ≤ 5th percentile) to screen epigenetic modulators of HbF and phenotypic diversity of ß-thalassemia. We identified an ETS2 repressor factor encoded by ERF, whose promoter hypermethylation and mRNA downregulation are associated with high HbF levels in ß-thalassemia. We further observed that hypermethylation of the ERF promoter mediated by enrichment of DNMT3A leads to demethylation of γ-globin genes and attenuation of binding of ERF on the HBG promoter and eventually re-activation of HbF in ß-thalassemia. We demonstrated that ERF depletion markedly increased HbF production in human CD34+ erythroid progenitor cells, HUDEP-2 cell lines, and transplanted NCG-Kit-V831M mice. ERF represses γ-globin expression by directly binding to two consensus motifs regulating γ-globin gene expression. Importantly, ERF depletion did not affect maturation of erythroid cells. Identification of alterations in DNA methylation of ERF as a modulator of HbF synthesis opens up therapeutic targets for ß-hemoglobinopathies.


Subject(s)
Epigenesis, Genetic , Gene Expression Profiling , Repressor Proteins/deficiency , Repressor Proteins/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics , Animals , Antigens, CD34/metabolism , Base Sequence , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Line , Child , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Female , Fetal Hemoglobin/genetics , Gene Editing , Humans , Male , Mice , Promoter Regions, Genetic/genetics , Reproducibility of Results , Sulfites , Whole Genome Sequencing , beta-Thalassemia/pathology
5.
Blood ; 139(21): 3181-3193, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35040907

ABSTRACT

Anemia of inflammation, also known as anemia of chronic disease, is refractory to erythropoietin (EPO) treatment, but the mechanisms underlying the EPO refractory state are unclear. Here, we demonstrate that high mobility group box-1 protein (HMGB1), a damage-associated molecular pattern molecule recently implicated in anemia development during sepsis, leads to reduced expansion and increased death of EPO-sensitive erythroid precursors in human models of erythropoiesis. HMGB1 significantly attenuates EPO-mediated phosphorylation of the Janus kinase 2/STAT5 and mTOR signaling pathways. Genetic ablation of receptor for advanced glycation end products, the only known HMGB1 receptor expressed by erythroid precursors, does not rescue the deleterious effects of HMGB1 on EPO signaling, either in human or murine precursors. Furthermore, surface plasmon resonance studies highlight the ability of HMGB1 to interfere with the binding between EPO and the EPOR. Administration of a monoclonal anti-HMGB1 antibody after sepsis onset in mice partially restores EPO signaling in vivo. Thus, HMGB1-mediated restriction of EPO signaling contributes to the chronic phase of anemia of inflammation.


Subject(s)
Anemia , Erythropoietin , HMGB1 Protein , Sepsis , Anemia/genetics , Animals , Erythropoiesis/genetics , Erythropoietin/metabolism , Inflammation , Mice , Receptors, Erythropoietin/metabolism , Sepsis/complications
6.
Transfusion ; 63(6): 1122-1128, 2023 06.
Article in English | MEDLINE | ID: mdl-37154531

ABSTRACT

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


Subject(s)
Erythrocytes , Erythroid Cells , Humans , Erythrocytes/metabolism , Erythroid Precursor Cells , K562 Cells , Flow Cytometry
7.
Protein Expr Purif ; 210: 106313, 2023 10.
Article in English | MEDLINE | ID: mdl-37276914

ABSTRACT

Many therapeutic proteins are expressed in Escherichia coli bacteria for the low cost and high yield obtained. However, these gram-negative bacteria also generate undesirable endotoxin byproducts such as lipopolysaccharides (LPS). These endotoxins can induce a human immune response and cause severe inflammation. To mitigate this problem, we have employed the ClearColi BL21 (DE3) endotoxin-free cells as an expression host for Cas9 protein production. Cas9 is an endonuclease enzyme that plays a key role in the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR associated protein 9 (CRISPR/Cas9) genome editing technique. This technology is very promising for use in diagnostics as well as treatment of diseases, especially for genetic diseases such as thalassemia. The potential uses for this technology thus generate a considerable interest for Cas9 utilization as a therapeutic protein in clinical treatment. Therefore, special care in protein production should be a major concern. Accordingly, we expressed the Cas9 protein in endotoxin-free bacterial cells achieving 99% purity with activity comparable to commercially available Cas9. Our protocol therefore yields a cost-effective product suitable for invitro experiments with stem cells.


Subject(s)
CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Humans , Endotoxins/genetics , Gene Editing/methods , Repressor Proteins
8.
Vox Sang ; 118(5): 392-397, 2023 May.
Article in English | MEDLINE | ID: mdl-36908196

ABSTRACT

BACKGROUND AND OBJECTIVES: The RHAG blood group system contains five antigens: Duclos (RHAG001), Ola (RHAG002), DSLK (RHAG003), Kg (RHAG005) and SHER (RHAG006). Individuals who are DSLK-negative and Kg-positive have the same allele RHAG*01.-3, with a single-nucleotide variation (rs144305805), c.490A>C (p.Lys164Gln), in exon 3 of the RHAG gene. We aimed to confirm whether DSLK and Kg are antithetical antigens. MATERIALS AND METHODS: Blood samples of the original DSLK-negative proband with anti-DSLK, her son and another DSLK-negative individual were examined. The RHAG gene was analysed by polymerase chain reaction and Sanger sequencing. Immunocomplex capture fluorescence assays (ICFAs) and monocyte phagocytosis assays were performed to characterize the anti-DSLK antibody. Cross-testing of alloanti-DSLK and monoclonal anti-Kg (OSK46) was performed using transduced HEK293 cells by inducing the construct of expression vectors encoding wild-type RHAG*01 or the variant RHAG*01.-3. RESULTS: ICFA using monoclonal anti-RHAG (LA18.18) revealed that the anti-DSLK and anti-Kg antibodies reacted with the wild-type and variant RhAG (Rh-associated glycoprotein), respectively. The proband and a DSLK-negative individual appeared to be homozygous for variant RHAG*01.-3, and the proband's son was typed as RHAG*01/RHAG*01.-3 heterozygote. HEK293 cells with wild-type RhAG reacted with the anti-DSLK but not anti-Kg antibody, whereas HEK293 cells expressing the variant RhAG reacted with the anti-Kg but not anti-DSLK antibody. Monocyte phagocytosis assays indicated that 64% of red cells sensitized with anti-DSLK were phagocytosed by monocytes. CONCLUSION: Our results demonstrate that DSLK and Kg are antithetical antigens in the RHAG blood group system. Anti-DSLK may be a clinically significant antibody.


Subject(s)
Blood Group Antigens , Rh-Hr Blood-Group System , Humans , Female , Rh-Hr Blood-Group System/genetics , HEK293 Cells , Membrane Glycoproteins/genetics , Blood Group Antigens/genetics , Erythrocytes/metabolism , Blood Proteins
9.
Nucleic Acids Res ; 49(17): 9711-9723, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34379783

ABSTRACT

Human fetal globin (γ-globin) genes are developmentally silenced after birth, and reactivation of γ-globin expression in adulthood ameliorates symptoms of hemoglobin disorders, such as sickle cell disease (SCD) and ß-thalassemia. However, the mechanisms by which γ-globin expression is precisely regulated are still incompletely understood. Here, we found that NonO (non-POU domain-containing octamer-binding protein) interacted directly with SOX6, and repressed the expression of γ-globin gene in human erythroid cells. We showed that NonO bound to the octamer binding motif, ATGCAAAT, of the γ-globin proximal promoter, resulting in inhibition of γ-globin transcription. Depletion of NonO resulted in significant activation of γ-globin expression in K562, HUDEP-2, and primary human erythroid progenitor cells. To confirm the role of NonO in vivo, we further generated a conditional knockout of NonO by using IFN-inducible Mx1-Cre transgenic mice. We found that induced NonO deletion reactivated murine embryonic globin and human γ-globin gene expression in adult ß-YAC mice, suggesting a conserved role for NonO during mammalian evolution. Thus, our data indicate that NonO acts as a novel transcriptional repressor of γ-globin gene expression through direct promoter binding, and is essential for γ-globin gene silencing.


Subject(s)
DNA-Binding Proteins/metabolism , Fetal Hemoglobin/genetics , Gene Silencing , RNA-Binding Proteins/metabolism , gamma-Globins/genetics , Animals , Cells, Cultured , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Humans , K562 Cells , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , SOXD Transcription Factors/metabolism , gamma-Globins/biosynthesis
10.
Rinsho Ketsueki ; 64(6): 482-488, 2023.
Article in Japanese | MEDLINE | ID: mdl-37407471

ABSTRACT

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.


Subject(s)
Transfusion Medicine , Humans , Erythrocytes , Erythroid Precursor Cells/metabolism , Erythropoiesis , Hematopoietic Stem Cells/metabolism , Cell Differentiation
11.
Mol Ther ; 29(4): 1625-1638, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33515514

ABSTRACT

Ongoing clinical trials for treatment of beta-globinopathies by gene therapy involve the transfer of the beta-globin gene, which requires integration of three to four copies per genome in most target cells. This high proviral load may increase genome toxicity, potentially limiting the safety of this therapy and relegating its use to total body myeloablation. We hypothesized that introducing an additional hypersensitive site from the locus control region, the complete sequence of the second intron of the beta-globin gene, and the ankyrin insulator may enhance beta-globin expression. We identified a construct, ALS20, that synthesized significantly higher adult hemoglobin levels than those of other constructs currently used in clinical trials. These findings were confirmed in erythroblastic cell lines and in primary cells isolated from sickle cell disease patients. Bone marrow transplantation studies in beta-thalassemia mice revealed that ALS20 was curative at less than one copy per genome. Injection of human CD34+ cells transduced with ALS20 led to safe, long-term, and high polyclonal engraftment in xenograft experiments. Successful treatment of beta-globinopathies with ALS20 could potentially be achieved at less than two copies per genome, minimizing the risk of cytotoxic events and lowering the intensity of myeloablation.


Subject(s)
Anemia, Sickle Cell/genetics , Bone Marrow Transplantation , Genetic Therapy , beta-Globins/genetics , beta-Thalassemia/genetics , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , Animals , Gene Expression/genetics , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Hemoglobins/genetics , Heterografts , Humans , Lentivirus/genetics , Locus Control Region/genetics , Mice , Transduction, Genetic , beta-Globins/therapeutic use , beta-Thalassemia/blood , beta-Thalassemia/pathology , beta-Thalassemia/therapy
12.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35163006

ABSTRACT

Molecular therapies and functional studies greatly benefit from spatial and temporal precision of genetic intervention. We therefore conceived and explored tag-activated microRNA (miRNA)-mediated endogene deactivation (TAMED) as a research tool and potential lineage-specific therapy. For proof of principle, we aimed to deactivate γ-globin repressor BCL11A in erythroid cells by tagging the 3' untranslated region (UTR) of BCL11A with miRNA recognition sites (MRSs) for the abundant erythromiR miR-451a. To this end, we employed nucleofection of CRISPR/Cas9 ribonucleoprotein (RNP) particles alongside double- or single-stranded oligodeoxynucleotides for, respectively, non-homologous-end-joining (NHEJ)- or homology-directed-repair (HDR)-mediated MRS insertion. NHEJ-based tagging was imprecise and inefficient (≤6%) and uniformly produced knock-in- and indel-containing MRS tags, whereas HDR-based tagging was more efficient (≤18%), but toxic for longer donors encoding concatenated and thus potentially more efficient MRS tags. Isolation of clones for robust HEK293T cells tagged with a homozygous quadruple MRS resulted in 25% spontaneous reduction in BCL11A and up to 36% reduction after transfection with an miR-451a mimic. Isolation of clones for human umbilical cord blood-derived erythroid progenitor-2 (HUDEP-2) cells tagged with single or double MRS allowed detection of albeit weak γ-globin induction. Our study demonstrates suitability of TAMED for physiologically relevant modulation of gene expression and its unsuitability for therapeutic application in its current form.


Subject(s)
Erythroid Cells/cytology , Gene Editing/methods , MicroRNAs/genetics , Repressor Proteins/genetics , 3' Untranslated Regions , CRISPR-Cas Systems , Cell Line , DNA End-Joining Repair , Erythroid Cells/metabolism , HEK293 Cells , Humans , Proof of Concept Study
13.
Br J Haematol ; 193(6): 1220-1227, 2021 06.
Article in English | MEDLINE | ID: mdl-33997955

ABSTRACT

Reactivation of fetal haemoglobin (HbF) expression is an effective way to treat ß-thalassaemia and sickle cell anaemia. In the present study, we identified a novel GATA zinc finger domain-containing protein 2A (GATAD2A) mutation, which contributed to the elevation of HbF and ameliorated clinical severity in a patient with ß-thalassaemia, by targeted next-generation sequencing. Knockout of GATAD2A led to a significant induction of HbF in both human umbilical cord blood-derived erythroid progenitor-2 (HUDEP-2) and human cluster of differentiation (CD)34+ cells with a detectable impact on erythroid differentiation. Furthermore, heterozygous knockout of GATAD2A impaired recruitment of chromodomain helicase DNA-binding protein 4 (CHD4) to the methyl-binding domain protein 2 (MBD2)-containing nucleosome remodelling and deacetylation (NuRD) complex. Our present data suggest that mutations causing the haploinsufficiency of GATAD2A might contribute to amelioration of clinical severity in patients with ß-thalassaemia.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nucleosomes/metabolism , Repressor Proteins/deficiency , beta-Thalassemia/metabolism , Acetylation , Adolescent , Cell Line , Child , Codon, Nonsense , DNA-Binding Proteins/genetics , Fetal Hemoglobin/genetics , Haploinsufficiency , Humans , Male , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Nucleosomes/genetics , Repressor Proteins/metabolism , beta-Thalassemia/genetics
14.
Br J Haematol ; 192(2): 395-404, 2021 01.
Article in English | MEDLINE | ID: mdl-33216968

ABSTRACT

Hereditary persistence of fetal haemoglobin (HPFH) is the major modifier of the clinical severity of ß-thalassaemia. The homozygous mutation c.-196 C>T in the Aγ-globin (HBG1) promoter, which causes Sardinian δß0 -thalassaemia, is able to completely rescue the ß-major thalassaemia phenotype caused by the ß0 39-thalassaemia mutation, ensuring high levels of fetal haemoglobin synthesis during adulthood. Here, we describe a CRISPR/Cas9 genome-editing approach, combined with the non-homologous end joining (NHEJ) pathway repair, aimed at reproducing the effects of this naturally occurring HPFH mutation in both HBG promoters. After selecting the most efficient guide RNA in K562 cells, we edited the HBG promoters in human umbilical cord blood-derived erythroid progenitor 2 cells (HUDEP-2) and in haematopoietic stem and progenitor cells (HSPCs) from ß0 -thalassaemia patients to assess the therapeutic potential of HbF induction. Our results indicate that small deletions targeting the -196-promoter region restore high levels of fetal haemoglobin (HbF) synthesis in all cell types tested. In pools of HSPCs derived from homozygous ß0 39-thalassaemia patients, a 20% editing determined a parallel 20% increase of HbF compared to unedited pools. These results suggest that editing the region of HBG promoters around the -196 position has the potential to induce therapeutic levels of HbF in patients with most types of ß-thalassaemia irrespective of the ß-globin gene (HBB) mutations.


Subject(s)
Fetal Hemoglobin/genetics , Gene Editing/methods , Hematopoietic Stem Cells/metabolism , beta-Thalassemia/genetics , CRISPR-Cas Systems , Cells, Cultured , HEK293 Cells , Humans , K562 Cells , Up-Regulation
15.
Biochem Biophys Res Commun ; 552: 157-163, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33744764

ABSTRACT

ß-Thalassemia is an autosomal recessive genetic disease caused by defects in the production of adult hemoglobin (HbA, α2ß2), which leads to an imbalance between α- and non-α-globin chains. Reactivation of γ-globin expression is an effective strategy to treat ß-thalassemia patients. Previously, it was demonstrated that hemoglobin subunit beta pseudogene 1 (HBBP1) is associated with elevated fetal hemoglobin (HbF, α2γ2) in ß-thalassemia patients. However, the mechanism underlying HBBP1-mediated HbF production is unknown. In this study, using bioinformatics analysis, we found that HBBP1 is involved in γ-globin production, and then preliminarily confirmed this finding in K562 cells. When HBBP1 was overexpressed, γ-globin expression was increased at the transcript and protein levels in HUDEP-2 cells. Next, we found that ETS transcription factor ELK1 (ELK1) binds to the HBBP1 proximal promoter and significantly promotes its activity. Moreover, the synthesis of γ-globin was enhanced when ELK1 was overexpressed in HUDEP-2 cells. Surprisingly, ELK1 also directly bound to and activated the γ-globin proximal promoter. Furthermore, we found that HBBP1 and ELK1 can interact with each other in HUDEP-2 cells. Collectively, these findings suggest that HBBP1 can induce γ-globin by enhancing ELK1 expression, providing some clues for γ-globin reactivation in ß-thalassemia.


Subject(s)
Gene Expression Regulation , RNA, Long Noncoding/genetics , beta-Thalassemia/genetics , ets-Domain Protein Elk-1/genetics , gamma-Globins/genetics , Cell Differentiation/genetics , Cell Line , Erythroid Precursor Cells/metabolism , Gene Expression Profiling/methods , Humans , K562 Cells , RNA Interference , beta-Thalassemia/metabolism , ets-Domain Protein Elk-1/metabolism , gamma-Globins/metabolism
16.
Blood ; 133(8): 852-856, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30617196

ABSTRACT

ß-hemoglobinopathies, such as sickle cell disease and ß-thalassemia, result from mutations in the adult ß-globin gene. Reactivating the developmentally silenced fetal γ-globin gene elevates fetal hemoglobin levels and ameliorates symptoms of ß-hemoglobinopathies. The continued expression of fetal γ-globin into adulthood occurs naturally in a genetic condition termed hereditary persistence of fetal hemoglobin (HPFH). Point mutations in the fetal γ-globin proximal promoter can cause HPFH. The -113A>G HPFH mutation falls within the -115 cluster of HPFH mutations, a binding site for the fetal globin repressor BCL11A. We demonstrate that the -113A>G HPFH mutation, unlike other mutations in the cluster, does not disrupt BCL11A binding but rather creates a de novo binding site for the transcriptional activator GATA1. Introduction of the -113A>G HPFH mutation into erythroid cells using the clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) system increases GATA1 binding and elevates fetal globin levels. These results reveal the mechanism by which the -113A>G HPFH mutation elevates fetal globin and demonstrate the sensitivity of the fetal globin promoter to point mutations that often disrupt repressor binding sites but here create a de novo site for an erythroid activator.


Subject(s)
Anemia, Sickle Cell , Fetal Hemoglobin , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , Point Mutation , Response Elements , beta-Thalassemia , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Cell Line , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , GATA1 Transcription Factor/genetics , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/genetics , beta-Thalassemia/metabolism
17.
Nature ; 527(7577): 192-7, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26375006

ABSTRACT

Enhancers, critical determinants of cellular identity, are commonly recognized by correlative chromatin marks and gain-of-function potential, although only loss-of-function studies can demonstrate their requirement in the native genomic context. Previously, we identified an erythroid enhancer of human BCL11A, subject to common genetic variation associated with the fetal haemoglobin level, the mouse orthologue of which is necessary for erythroid BCL11A expression. Here we develop pooled clustered regularly interspaced palindromic repeat (CRISPR)-Cas9 guide RNA libraries to perform in situ saturating mutagenesis of the human and mouse enhancers. This approach reveals critical minimal features and discrete vulnerabilities of these enhancers. Despite conserved function of the composite enhancers, their architecture diverges. The crucial human sequences appear to be primate-specific. Through editing of primary human progenitors and mouse transgenesis, we validate the BCL11A erythroid enhancer as a target for fetal haemoglobin reinduction. The detailed enhancer map will inform therapeutic genome editing, and the screening approach described here is generally applicable to functional interrogation of non-coding genomic elements.


Subject(s)
CRISPR-Associated Proteins/metabolism , Carrier Proteins/genetics , Enhancer Elements, Genetic/genetics , Genetic Engineering , Mutagenesis/genetics , Nuclear Proteins/genetics , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins , Erythroblasts/metabolism , Fetal Hemoglobin/genetics , Genome/genetics , Humans , Mice , Molecular Sequence Data , Organ Specificity , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , Reproducibility of Results , Species Specificity
18.
Mol Ther ; 28(1): 129-141, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31677955

ABSTRACT

Recent advances in gene therapy technologies have enabled the treatment of congenital disorders and cancers and facilitated the development of innovative methods, including induced pluripotent stem cell (iPSC) production and genome editing. We recently developed a novel non-transmissible and non-integrating measles virus (MV) vector capable of transferring multiple genes simultaneously into a wide range of cells through the CD46 and CD150 receptors. The MV vector expresses four genes for iPSC generation and the GFP gene for a period of time sufficient to establish iPSCs from human fibroblasts as well as peripheral blood T cells. The transgenes were expressed differentially depending on their gene order in the vector. Human hematopoietic stem/progenitor cells were directly and efficiently reprogrammed to naive-like cells that could proliferate and differentiate into primed iPSCs by the same method used to establish primed iPSCs from other cell types. The novel MV vector has several advantages for establishing iPSCs and potential future applications in gene therapy.


Subject(s)
Cellular Reprogramming/genetics , Genetic Vectors , Genome, Viral/genetics , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Measles virus/genetics , RNA, Viral/genetics , Animals , Blood Donors , Cell Differentiation/genetics , Fibroblasts/metabolism , Genetic Therapy/methods , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Sendai virus/genetics , T-Lymphocytes/metabolism , Transduction, Genetic , Transgenes
19.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807258

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs crucial for post-transcriptional and translational regulation of cellular and developmental pathways. The study of miRNAs in erythropoiesis elucidates underlying regulatory mechanisms and facilitates related diagnostic and therapy development. Here, we used DNA Nanoball (DNB) small RNA sequencing to comprehensively characterize miRNAs in human erythroid cell cultures. Based on primary human peripheral-blood-derived CD34+ (hCD34+) cells and two influential erythroid cell lines with adult and fetal hemoglobin expression patterns, HUDEP-2 and HUDEP-1, respectively, our study links differential miRNA expression to erythroid differentiation, cell type, and hemoglobin expression profile. Sequencing results validated by reverse-transcription quantitative PCR (RT-qPCR) of selected miRNAs indicate shared differentiation signatures in primary and immortalized cells, characterized by reduced overall miRNA expression and reciprocal expression increases for individual lineage-specific miRNAs in late-stage erythropoiesis. Despite the high similarity of same-stage hCD34+ and HUDEP-2 cells, differential expression of several miRNAs highlighted informative discrepancies between both cell types. Moreover, a comparison between HUDEP-2 and HUDEP-1 cells displayed changes in miRNAs, transcription factors (TFs), target genes, and pathways associated with globin switching. In resulting TF-miRNA co-regulatory networks, major therapeutically relevant regulators of globin expression were targeted by many co-expressed miRNAs, outlining intricate combinatorial miRNA regulation of globin expression in erythroid cells.


Subject(s)
Erythroid Cells/classification , Erythroid Cells/metabolism , MicroRNAs/genetics , Adult , Age Factors , Cell Differentiation/genetics , Cell Line , Erythropoiesis/genetics , Fetal Blood/cytology , Fetal Hemoglobin/genetics , Fetus/metabolism , Humans , RNA, Messenger/genetics , Transcription Factors , gamma-Globins/genetics
20.
Blood ; 131(17): 1960-1973, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29519807

ABSTRACT

Naturally occurring, large deletions in the ß-globin locus result in hereditary persistence of fetal hemoglobin, a condition that mitigates the clinical severity of sickle cell disease (SCD) and ß-thalassemia. We designed a clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9) strategy to disrupt a 13.6-kb genomic region encompassing the δ- and ß-globin genes and a putative γ-δ intergenic fetal hemoglobin (HbF) silencer. Disruption of just the putative HbF silencer results in a mild increase in γ-globin expression, whereas deletion or inversion of a 13.6-kb region causes a robust reactivation of HbF synthesis in adult erythroblasts that is associated with epigenetic modifications and changes in chromatin contacts within the ß-globin locus. In primary SCD patient-derived hematopoietic stem/progenitor cells, targeting the 13.6-kb region results in a high proportion of γ-globin expression in erythroblasts, increased HbF synthesis, and amelioration of the sickling cell phenotype. Overall, this study provides clues for a potential CRISPR/Cas9 genome editing approach to the therapy of ß-hemoglobinopathies.


Subject(s)
Anemia, Sickle Cell , CRISPR-Cas Systems , Fetal Hemoglobin , Gene Editing , Genetic Loci , Hematopoietic Stem Cells/metabolism , beta-Globins/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/therapy , Cell Line , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Hematopoietic Stem Cells/pathology , Humans , beta-Globins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL