Next Article in Journal
Estimates of Genomic Heritability and the Marker-Derived Gene for Re(Production) Traits in Xinggao Sheep
Previous Article in Journal
DNMT3B rs2424913 as a Risk Factor for Congenital Heart Defects in Down Syndrome
Previous Article in Special Issue
In Vivo Hematopoietic Stem Cell Genome Editing: Perspectives and Limitations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epigenetic Regulation of β-Globin Genes and the Potential to Treat Hemoglobinopathies through Epigenome Editing

1
Laboratory of Chromatin and Gene Regulation during Development, INSERM UMR 1163, Imagine Institute, Université Paris Cité, F-75015 Paris, France
2
Genome Engineering, Discovery Sciences, BioPharmaceuticals R&D Unit, AstraZeneca, 431 50 Gothenburg, Sweden
*
Authors to whom correspondence should be addressed.
Genes 2023, 14(3), 577; https://doi.org/10.3390/genes14030577
Submission received: 9 December 2022 / Revised: 21 February 2023 / Accepted: 23 February 2023 / Published: 25 February 2023
(This article belongs to the Special Issue Gene Editing for Therapy and Reverse Genetics of Blood Diseases)

Abstract

:
Beta-like globin gene expression is developmentally regulated during life by transcription factors, chromatin looping and epigenome modifications of the β-globin locus. Epigenome modifications, such as histone methylation/demethylation and acetylation/deacetylation and DNA methylation, are associated with up- or down-regulation of gene expression. The understanding of these mechanisms and their outcome in gene expression has paved the way to the development of new therapeutic strategies for treating various diseases, such as β-hemoglobinopathies. Histone deacetylase and DNA methyl-transferase inhibitors are currently being tested in clinical trials for hemoglobinopathies patients. However, these approaches are often uncertain, non-specific and their global effect poses serious safety concerns. Epigenome editing is a recently developed and promising tool that consists of a DNA recognition domain (zinc finger, transcription activator-like effector or dead clustered regularly interspaced short palindromic repeats Cas9) fused to the catalytic domain of a chromatin-modifying enzyme. It offers a more specific targeting of disease-related genes (e.g., the ability to reactivate the fetal γ-globin genes and improve the hemoglobinopathy phenotype) and it facilitates the development of scarless gene therapy approaches. Here, we summarize the mechanisms of epigenome regulation of the β-globin locus, and we discuss the application of epigenome editing for the treatment of hemoglobinopathies.

1. Introduction

1.1. Hemoglobins

Hemoglobin (Hb), the main component of mature red blood cells (RBCs), is a tetrameric metalloprotein composed of four polypeptide globin helicoid chains (two α-like and two β-like globins), a heme molecule and a ferrous ion (Fe). Different variants of Hb, with a different composition of α- and β-like chains, are produced throughout life in a stage-specific manner [1] with two Hb switches taking place during life, a primary switch from embryonic to fetal and a second switch from fetal to adult that occurs shortly after birth. Fetal Hb (HbF) is replaced by adult Hb (HbA), which is the predominant Hb during adult life (>95% of total Hb), while HbF and HbA2 are normally expressed at low levels (~1%) [2,3,4] (Figure 1). Even though the different Hb types present a very similar structure with only a few amino acid differences in the globin chains, they have a different O2 affinity. Embryonic Hb (Hb Gower 1, ζ2ε2; Hb Gower 2, α2ε2; and Hb Portland 1, ζ2γ2) and fetal Hb (HbF, α2γ2) exhibit a higher O2 affinity than the adult one (HbA, α2β2; and HbA2, α2δ2) [5,6].

1.2. Globin Gene Regulation

Human globin genes are organized in two different clusters, the α- and the β-globin loci, located in chromosomes 16 and 11, respectively. The human α- and β-globin locus genes are arranged in a 5′ to 3′ direction according to their sequential expression during development (Figure 1). Human globin genes are expressed in an erythroid- and developmental stage-specific manner during life. The regulation of gene expression is accomplished at three levels: by proximal regulation through transcription factors (TFs) that bind to globin gene promoters and allow their expression [7,8,9]; by distal regulation through the locus control regions (LCR) of α- and β-globin loci that loop with globin promoters and allow developmental stage-specific expression via their DNase I hypersensitive sites (HS) that act as enhancers or insulators [4,10,11,12,13,14]; and lastly, by epigenetic modifications associated with high or low gene expression levels (see “Epigenetic regulation of the β-globin genes”; Figure 2).

1.3. The Fetal-to-Adult Hb Switching

The fetal-to-adult Hb switch results in the silencing of γ-globin genes and the activation of the β-globin gene. The specific binding of TFs to the β-like globin genes and the chromatin remodeling through LCR-mediated looping formation is required for the regulated expression of β-like globin genes. The chromatin looping drives the γ- or β-globin gene expression in fetal and adult erythroblasts, respectively. The looping is mediated by a protein complex and facilitates the juxtaposition of the LCR to the γ- or β-globin gene promoter, thus promoting their expression [32,33,34,35,36].
Numerous TFs and co-factors are involved in the repression or activation of the HBG genes (Figure 2). Genome-wide association studies have linked single-nucleotide polymorphisms (SNPs) with abnormally elevated HbF levels [37,38]. The location of these SNPs facilitated the identification of one of the major transcriptional repressors of γ-globin genes, B-cell lymphoma/leukemia 11A (BCL11A) [39] that suppresses gene expression by binding to the γ-globin promoters [40].
Different isoforms of BCL11A are expressed in adult and fetal erythroblasts, thanks to alternative mRNA splicing. Larger forms (XL and L) are expressed in adult cells, whereas shorter forms (S and XS) are expressed in fetal cells, indicating the stage-specific repressive action of larger BCL11A isoforms [41]. BCL11A knockdown experiments showed an increase in γ-globin mRNA and HbF levels in erythroid cells derived from human HSPCs, with no impairment of the erythroid differentiation. BCL11A interacts and recruits other factors/complexes that act as co-repressors, such as the nucleosome remodeling deacetylase (NuRD) complex and polycomb repressive complex 2 (PRC2 containing Enhancer of zeste homolog 2 (EZH2), Embryonic ectoderm development (EED), and SUZ12) [26,42,43]. More recently, the zinc finger and BTB domain containing 7A (ZBTB7A), also known as lymphoma/leukemia-related factor (LRF), was identified as another major γ-globin repressor that binds to the γ-globin promoters, recruits NuRD and downregulates gene expression in a similar yet independent way from BCL11A [44,45]. The NuRD polymeric protein complex, upon recruitment at the γ-globin genes by BCL11A or LRF, is thought to act through its components, such as histone deacetylases (HDACs) [43] to silence gene expression.

1.4. Beta-Hemoglobinopathies

Beta-hemoglobinopathies are monogenic diseases caused by mutations in the β-globin locus that affect the synthesis, structure or properties of HbA. β-thalassemia is caused by mutations in the β-globin locus that reduce (β+) or abolish (β0) the production of adult β-globin chains composing the HbA tetramer. This leads to the precipitation of uncoupled α-globin chains, ineffective erythropoiesis, erythroid cell death by apoptosis and anemia [46]. In SCD, an A>T mutation in the β-globin gene (HBB) causes the substitution of valine for glutamic acid at position 6 of the β-globin chain (βS) that is responsible for deoxygenation-induced polymerization of the sickle hemoglobin (HbS). This primary event drives ineffective erythropoiesis, apoptosis of erythroid precursors, red blood cell (RBC) sickling, hemolysis, vaso-occlusive crises and multi-organ damage, often associated with severely reduced life expectancy [47].
Allogeneic HSC transplantation is the only curative therapy for β-hemoglobinopathies. However, the absence of sibling donors and the risk of immunological complications prevent its use in a large fraction of patients. The clinical severity of β-hemoglobinopathies is alleviated by the persistent expression of fetal γ-globin chains in adulthood, e.g., in hereditary persistence of fetal hemoglobin (HPFH). In β-thalassemia, elevated fetal γ-globin levels lower the imbalance between α- and non-α globin chains, reduce the formation of α-globin precipitates and thus improve the clinical phenotype. In SCD, fetal γ-globin exerts a potent anti-sickling effect by replacing the sickle β-globin in the Hb tetramers [48]. Therefore, the γ-to-β-globin switch is the subject of extensive study, as understanding the underlying mechanisms is crucial to develop novel therapeutic strategies to combat this multifaceted disease group.

2. Epigenetic Regulation of the β-Globin Genes

Epigenetic modifications, which are defined as chromatin alterations that result in a determined expression pattern without changing the genetic sequence, have been shown to occur during the fetal-to-adult hemoglobin switching (Figure 3). These potentially heritable modifications, which include DNA methylation and histone modifications (acetylation and methylation), can be cell-, tissue- and developmental-stage specific. Epigenetic modifications change the chromatin structure and DNA accessibility, and consequently affect gene expression, as epigenetic modifiers co-operate with TFs to repress or activate gene expression (Figure 2).

2.1. DNA Methylation

The promoters of many human genes (including globin genes) contain Cytosine-phosphate Guanine (CpG) sites, which occur at a high frequency in regions known as “CpG islands”, and are amenable to epigenetic modifications. DNA methyltransferases (DNMT) deposit methyl groups in the fifth carbon atom of the cytosine of a CpG site. DNMT3A and DNMT3B mediate the de novo deposition of methyl groups in a non-methylated 5′-CpG-3′ and symmetrically to the non-methylated 3′-GpC-5′ of the complementary strand [49]. DNMT1 is responsible for the maintenance of the methylation of asymmetrically semi-methylated CpG sites upon DNA replication [50]. The ten-eleven translocation methyl-cytosine dioxygenase (TET) proteins TET1, TET2 and TET3 are responsible for DNA demethylation [51,52]. TET is responsible for the removal of the methyl group from a 5-methyl-Cytosine (5-mC) through the generation of a 5-hydroxy-methyl-cytosine (5-hmC) intermediate [51,52]. High DNA methylation levels normally characterize transcriptional inactive promoter regions, while hypomethylated promoters and promoters bearing the 5-hmC intermediate are generally highly expressed [53]. However, CpG methylation is not always associated with gene repression, such as when it occurs at enhancer regions. For this reason, some studies suggest that CpG methylation denotes active enhancer regions and might be required for depositing the active H3K27 histone acetylation mark (see below) [54].
In the γ-globin promoters, CpG sites span a 500 nt region centered on the transcription start site (−162, −53, −50, +6, +17, +50, CpG sites) [55,56,57] (Figure 3). Studies in erythroid cells obtained from human hematopoietic stem and progenitor cells (HSPCs) demonstrated that the γ-globin promoters were significantly hypomethylated in cord blood and fetal liver samples compared to adult bone marrow samples [55,56,57]. Conversely, the DNA methylation levels of the adult β-globin promoter were higher in fetal cells than in adult samples [55]. Moreover, an abnormal DNA hypomethylation pattern in the γ-globin promoters was associated with an increased HbF expression in β-thalassemia patient cells as compared to healthy donors [56] and in individuals with HPFH mutations [58]. By contrast, in a non-human primate baboon model, 5hmC levels positively correlated with γ-globin expression, although the 5hmC levels were substantially lower than 5Mc [53]. Interestingly, a missense mutation in the DNMT1 gene found in patients with HbE/β-thalassemia, leads to lower stability and enzymatic activity loss of DNMT1. This was associated with the reactivation of HbF due to the diminished recruitment of DNMT1 to the γ-globin promoters and the reduced methylation levels in CpG sites in the γ-globin promoters (−53, −50, +6, +17, and +50) of erythroid cells differentiated from patient HSPCs [59].

2.2. Histone Modifications

The DNA of eukaryotes is wrapped around histone octamers, i.e., nucleosomes, forming the chromatin. A 147 bp-long DNA fragment is wrapped around a histone octamer, composed of two copies of histone (H) 2A, H2B, H3 and H4, while histone H1 binds to the linker DNA between two nucleosomes. A variety of post-translational modifications (e.g., methylation, acetylation and phosphorylation) in different amino acid residues of the histones have been identified and associated with distinct regulatory functions [60,61,62,63].

2.2.1. Histone Methylations

The transfer of methyl groups to histone amino acid residues by methyl-transferase proteins (lysine methyltransferase (KMT) and protein arginine methyltransferase (PRMT)) is associated either with an open chromatin state and gene activation, or to a compact chromatin state and gene repression. For instance, the H3 lysine 4 monomethylation, dimethylation or trimethylation (H3K4me1, H3K4me2 and H3K4me3, respectively) and the asymmetric H4 arginine 3 dimethylation (H4R3me2a) mark transcriptionally active genomic regions (with H3K4me1 typically marking enhancers, H3K4me2 marking promoters and enhancers, and H3K4me3 marking promoters). On the other hand, the deposition of methyl, dimethyl or trimethyl groups on H3 lysine 9 or lysine 27 (H3K9me1, H3K9me2, H3K9me3, H3K27me2 and H3K27me3, respectively) or symmetric H4 arginine 3 (H4R3me2s) or H4 lysine 20 (H4K20me3) is associated with transcription inactivation [64]. Interestingly, the simultaneous presence of activating H3K4me3 and repressive H3K27me3 characterizes a bivalent chromatin state typical of genes poised for rapid activation [61]. These modifications can be removed by demethylases (e.g., lysine-specific histone demethylase 1 (KDM1A), also known as lysine-specific demethylase (LSD1)).
The role of histone methylation in the regulation of the β-globin genes has been studied for decades (Figure 3). The developmental-stage specific pattern of histone modifications along the β-globin locus was analyzed by comparing human embryonic cell-, fetal liver- and bone marrow-derived erythroblasts. The three sources have a distinct globin expression profile corresponding to an embryonic/fetal, fetal and adult state, respectively. Accordingly, the presence of the active H3K4me3 mark is correlated with gene activity in the different cell types [65]. Studies have revealed a correlation between the type of histone methylation and the activation or repression of globin genes [66,67,68]. For instance, in human adult erythroid cells, the activating H3K4me2/me3 and the repressive H3K9me1 marks are present at the β- and γ-globin promoters, respectively [43,66]. Interestingly, upon administration of tranylcypromine (TCP), a KDM1A inhibitor [69], the activating H3K4me2 mark increased in the γ-globin but not in the β-globin promoter along differentiation, suggesting that KDM1A is responsible for the repression of the fetal genes by removing the activating H3K4me2 mark.
Additional proteins are involved in the determination of the histone methylation status of the β-globin locus. In particular, the expression of EHMT1/2 as well as the global H3K9me2 levels decrease along the erythroid differentiation in cells derived from adult HSPCs [70]. Pharmacological inhibition of EHMT1/2 (using UNC0638) or shRNA-mediated knockdown of EHMT1/2 led to decreased levels of H3K9me2 and, consequently, HbF reactivation [70,71]. However, the reactivation of HbF observed upon EHMT1/2 downregulation was associated with an additional activating event, the acetylation of H3 lysine 9 (H3K9Ac) at the HBG1/2 promoters, which is typically associated with gene activation (see paragraph “Histone acetylations”) [70]. Promisingly, UNC0638 also led to high levels of HbF expression in erythroid cells from β-thalassemia patients [72]. FTX-6058, and inhibitor of EDD, was also tested in healthy volunteers and induced high HbF levels without major side effects (NCT04586985).
Methylation of arginine 3 on H4 also plays an important role in globin gene regulation. In human erythroid cell lines, PRMT5 induces symmetric H4R3me2 in the γ-globin promoters [73]. The developmental-stage specific pattern of H4R3me2s marks along the β-globin locus was observed in primary erythroid cells. H4R3me2s levels in the γ-globin promoters were higher in human bone marrow-derived erythroid cells compared to cord blood-derived erythroid cells [73]. Conversely, H4R3me2s marks and PRMT5 were absent from the β-globin promoters of human bone marrow-derived erythroid cells. Small-molecule inhibition of PRMT5 (using adenosine-2′,3′-dialdehyde (Adox)) in erythroid cells derived from adult bone marrow HSPCs reduce H4R3me2s levels at the γ-globin promoters with a concomitant HbF reactivation. PRMT5 is thought to be recruited at the γ-globin promoters by LYAR. In fact, upon LYAR knockdown, both PRMT5 occupancy and H4R3me2 levels decrease at the γ- globin promoters [74].
An interplay between the different histone methylations and between histone methylations and DNA methylation has been reported at the β-like globin promoters. In human erythroid cell lines, PRMT5-induced symmetric H4R3me2 in the γ-globin promoters is associated with the recruitment of DNMT3A that methylates close CpG sites, highlighting the interplay between histone and DNA methylation [73]. Furthermore, in human erythroid cell lines, PRMT5 also induces the deposition of additional repressive histone methylation marks in the γ-globin promoters, such as H4K20me3, H3K9me3 and H3K27me3, likely by recruiting other repressor complexes such as KMT5B (also known as SUV4-20h1), a lysine methyltransferase that deposits the repressive H4K20me3 mark. The H4K20me3 mark was higher at the embryonic/fetal ε- and γ-globin promoters in human bone marrow-derived erythroblasts as compared to cord blood-derived erythroid cells. Accordingly, shRNA-mediated downregulation of SUV4-20h1 led to γ-globin gene reactivation in adult cells [64].

2.2.2. Histone Acetylations

The deposition of acetyl groups on histone lysine residues loosens the interactions between histones and DNA due to a neutralization of the positive histone charge [75,76]. As a result, chromatin is in an open state, and TFs can bind to the DNA resulting in transcription activation. The proteins that are responsible for histone acetylation are histone acetyl-transferases (HAT; for example, p300), which are lysine acetyl-transferases (KAT) that deposit acetyl groups in lysine residues of the histone tail. Conversely, lysine deacetylases (KDAC), including histone deacetylases (HDAC), can remove the acetyl groups from histones, thus compacting the chromatin. There are four classes of HDACs, Rpd3-like proteins (class I; HDAC1, HDAC2, HDAC3 and HDAC8), Hda1-like proteins (class II; HDAC4, HDAC5, HDAC6, HDAC7, HDAC9 and HDAC10), HDAC11 (class IV) and Sir2-like proteins (class III; SIRT1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6 and SIRT7).
Histone acetylation plays a dynamic role in the developmental-stage specific regulation of β-like globin gene expression (Figure 3). A positive correlation has been established between the β-like globin gene expression levels and H3 and H4 acetylation in the LCR and in the β-like globin genes in murine fetal liver cells and erythroid cell lines harboring the human β-globin locus [77,78,79].
Studies using human fetal and adult erythroid cells have shown that the levels of H3 acetylation in the LCR are stable during development, while the same marker in the β-globin coding regions positively correlates with HBB expression levels [65,80]. In general, in human primary cells, when a β-like globin gene is activated (e.g., the γ-globin gene in fetal erythroblasts), there is an H3 loss from the promoter and a strong acetylation of the remaining histones leading to a more open chromatin state [80]. In fact, human primitive-like, fetal and adult erythroid cells display H3 lysine 9 or 27 hyperacetylation in the ε-, γ- and β-globin gene promoters, respectively [55,66,67,80]
Interestingly, histone deacetylase activity is not always correlated with γ-globin repression. In fact, SIRT1, an histone deacetylase has been positively associated with increased γ-globin expression, and SIRT1 activators enhance the expression of γ-globin in primary human erythroid cells [81]. In fetal cord blood erythroid cells, the levels of SIRT1 are higher as compared to adult bone marrow cells [81]. SIRT1 was found to enhance the looping between the LCR and the γ-globin promoters, and this was also associated with an indirect increase in the levels of the activating H4K16ac mark in the γ-globin promoters [81]. Therefore, SIRT1 activators could potentially be used for reactivating HbF in hemoglobinopathy patients.
The interplay between different types of histone modifications also characterizes the β-globin locus regulation and has also been investigated at the level of enhancers (typically marked by H3K4me1) in human erythroid cell lines. In the β-globin LCR, H3K4me1 was correlated with histone acetylation, namely, H3K27ac. Interestingly, the loss of H3K4me1 led to a concomitant reduction in H3K27ac, but not vice versa, indicating a hierarchy that places methylation upstream of acetylation in the β-globin locus regulation [82].
Histone acetylations also cooperate with DNA methylation in regulating globin gene expression. In human cells, H3 and H4 acetylation regulates the β-globin genes in concert with other histone modifications (e.g., histone methylations and phosphorylation) and DNA methylation. Along the human erythroid differentiation, the activating H3K9ac marks in the β-globin locus overlap with histone methylation marks (H3K4me1 and H3K4me3), and 5-hmC [83], highlighting the interplay between different epigenetic marks to control gene expression. Furthermore, PRMT5 [64] or LYAR [74] downregulation in human erythroid cell lines led not only to the eradication of the H4R3me2s repressive mark, but also led to a reduction in the repressive H4 serine 1 phosphorylation (H4S1ph) mark and an increase in the activating H4K12ac, H4K8ac and H3K9ac marks. Inhibition of PRMT5 by Adox in adult bone marrow erythroid cells also increased the H4 acetylation levels at the γ-globin promoters [84].

2.3. The Interplay between Epigenetic Modifiers, Transcription Factors and Chromatin Looping

The fine-tuning of globin gene expression is mediated by the direct or indirect repressive or activating effects of TFs that bind to the β-globin locus, and by an interplay between these TFs and DNA or histone modifiers that alter the epigenetic profile of the locus and, thus, its chromatin accessibility. By way of example, the GATA1 TF interacts with the CBP/p300 acetyltransferase during erythropoiesis [85]. With regard to the fetal-to-adult Hb switching, the major players of this interplay are the BCL11A and LRF TFs that interact with co-factors including epigenome modifiers. BCL11A and LRF interact with the NuRD complex, which contains HDACs and MBD2. MBD2 binds to methylated CpG regions and recruits other NuRD components to repress gene expression [86]. BCL11A also interacts with histone demethylases, such as KDM1A, which removes activating histone methylation marks in the γ-globin promoters [43]. Another repressive complex that regulates the expression of γ-globin genes is the direct repeat erythroid-definitive (DRED) complex that contains KDM1A, DNMT1 and testicular receptor 2 and 4 (TR2/TR4)TF [43,87] (Figure 2).
Finally, the specific binding of TFs to the β-like globin genes and the chromatin remodeling through LCR-mediated looping formation is required for the regulated expression of β-like globin genes. The chromatin looping drives the γ-globin or β-globin gene expression in fetal and adult erythroblasts, respectively. The looping is mediated by a protein complex containing LDB1, LMO2, GATA1, TAL1 and E2A and facilitates the juxtaposition of the LCR to the γ- or the β-globin gene promoter, thus promoting their expression [32,33,34,35,36]. The recruitment of this protein complex inducing chromatin looping is also associated with epigenetic changes in the β-globin locus. In fact, in a murine erythroid cell line, GATA1 induces LDB1-mediated chromatin looping and H3K4me3 deposition at the β-globin gene to regulate its expression [88]. Furthermore, in a human fetal erythroid cell line, the ETO2 transcriptional co-repressor impairs the binding of this protein complex to the β-globin locus and, as a consequence, the looping between the LCR to the γ-globin gene. This is paralleled by the recruitment of the NuRD complex, the decrease in H3K27ac and H3K9ac at the β-globin locus, and the repression of the γ-globin genes [89]. Taken together, these results indicate a tight interplay between chromatin looping and epigenetic modifications at the β-globin locus to promote proper β-like globin gene regulation.

3. Epigenome Editing

Growing evidence of the crucial role of the epigenome in gene regulation, and the reversible nature of epigenetic modifications has led to the development of “epidrugs” as a clinical treatment for different diseases [90,91]. However, as they usually target epigenome modifiers, their effect is not sequence specific and can lead to a broad alteration of gene expression, sometimes leading to cell death [92].
In order to mitigate this effect, engineered epigenetic effectors that are capable of performing targeted epigenome editing have been developed. Targeted epigenome modifications can be generated by fusing an effector domain (that either has intrinsic epigenetic enzymatic activity or recruits epigenome modifiers in the case of TFs) to a sequence-specific DNA-binding domain (DBD) to ensure that the effector domain acts only on a specific target (Figure 4).

3.1. Zinc Finger-Based Epigenome Editors

Zinc-finger proteins (ZFs) have been the DBD of choice for the first epigenetic applications. Those proteins are composed of repetitive Cys2His2 domains, consisting of 28–30 amino acids, which allow binding to double-stranded DNA through a zinc ion, coordinated by four conserved residues [93]. Multiple fingers are joined together to form multi-ZF proteins, generating a versatile binding domain that binds DNA with very high affinity [94]. Indeed, each finger recognizes a specific 3 or 4 base pair sequence, allowing the binding to a specific target region. Those characteristics have been exploited to generate artificial polydactyl ZF proteins in vitro to target regions of interest [95].
Initially, ZFs were fused to TFs or epigenetic modifiers. They were coupled with TFs such as p65, viral activation domains such as VP16, or transcriptional activators composed of several tandem copies of VP16 to achieve activation of specific endogenous genes [96,97,98,99]. However, the use of these artificial TFs only leads to a modulation of the gene expression, since these tools might not efficiently modify the local epigenetic landscape, and their outcome can be influenced by the accessibility of the target site for the DBDs [100].
To overcome this limitation, a wide range of effector domains with intrinsic epigenetic activity have been employed in conjunction with ZF DBDs to either increase or repress gene expression [100,101,102,103]. Hence, ZFs have been fused to TET1 or TET2 domains to induce promoter demethylation, thus increasing gene expression [100,101], or to DNMT3A and its co-factor DNMT3L to de novo methylate promoters and decrease gene expression [102]. Moreover, the HAT domain of p300 has been fused to a ZF DBD to increase gene expression by depositing histone acetylation at promoter regions [103]. All these fusion proteins are characterized by a small size, which allows easy delivery by plasmid transfection [103] and retroviral vectors [104,105].
Although different platforms using ZFs as DBDs have been developed and have reached clinical application, the low specificity of the tools remains an important barrier for their use in the epigenome editing field. ChIP-seq analysis has showed that although ZFs preferentially bind to the target genomic region, there is also widespread binding at potentially off-target sites [99,101], most frequently mapping to promoter regions that show a significant sequence similarity with the intended target site [100]. Likewise, the addition of effector domains can alter the ZF binding distribution, increasing the off-target rate [106]. To overcome this issue, the effector domains have been divided into two separate components engineered with ZFs targeting adjacent regions. For instance, a monomeric methyltransferase was split into two fragments that cannot function unless assembled into an active heterodimeric enzyme. Each fragment was subsequently fused to a different zinc finger. This tool was used to drive CpG methylation at different target sites, resulting in a limited methyltransferase activity in the absence of the flanking zinc finger binding site [107]. However, it should be noted that protein complementation strategies remain challenging since they require complex protein engineering.
Lessons learned from ZF-based epigenome editors encouraged a faster development of other epigenome editing platforms with higher DNA-recognition capacities, namely, transcription activator-like effectors (TALE) and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9.

3.2. TALE-Based Epigenome Editors

An alternative DBD option for epigenome editing applications arose from the study of TALEs in bacterial plant pathogens [108]. These proteins present a modular DBD which can target any DNA region [109]. The TALE DBD is composed of 7 to 34 tandem repeats, each containing 33–35 amino acids [110]. These repeats are highly conserved, except at the hypervariable positions 12 and 13, the so-called repeat-variable diresidue (RVD) which determines the base preference of the repeat [111]). Thus, there is a 1:1 interaction between each TALE repeat and a single nucleotide. The specific binding properties of a RVD vary depending on the structure of the TALE. It is influenced by the total number of repeats, the position of the repeats within the TALE array and the surrounding RVD [112,113]. All these characteristics are deeply evaluated during the design of TALE DBDs for in vitro production to increase the specificity of the DBDs.
TALEs have been fused to various effector domains to either up- or down-regulate gene expression. TFs such as VP16 and VP64 were fused to TALEs with the aim of reactivating the pluripotency in somatic stem cells, by promoting the expression of SOX2, KLF4 and OCT4 [103,114,115,116].
Other artificial transcriptional repressors have been developed by combining TALEs with the catalytic domain of KRAB to silence endogenous gene expression. The KRAB domain has a transcriptional repressive activity and operates through interactions with different corepressors and chromatin modifiers that remove histone H3 acetylation and increase H3K9me3 [117]. TALE:KRAB fusions have been tested in HEK293FT cells to repress the SOX2 gene [118], as well as in mice HSPCs to knock down the c-kit and Pu.1 genes [119]. Moreover, TALEs fused with the KRAB domain have been described as an important tool to maintain pluripotency and self-renewal capacity in human HSPCs. In particular, TALE:KRAB has been successfully used to transiently silence the expression of TCF3, involved in the suppression of pluripotency and self-renewal pathways, thus providing a promising option for obtaining a sufficient number of HSCs for transplantation [120].
With the same intent of gene silencing, TALEs have also been tested in combination with other repressive domains. For instance, TALEs have been tethered with the mSin interaction domain (SID), which acts by reducing H3K9 acetylation, thus inducing transcriptional repression in cell lines (to repress the calcium channel related CACNA1C gene) [118] and primary murine neurons (to downregulate the expression of Grm2 gene, encoding one of the major excitatory neurotransmitters) [121].
Several TALE-based tools have been developed to directly modulate DNA methylation or deposit or remove histone modifications. TALE-based epigenome editors have been used to remove aberrant DNA methylation, which is associated with several diseases. By way of example, TALE DBDs have been fused with TET1 to remove critical CpG methylations at the endogenous promoter of KLF4 in the K562 cell line and reactivate its expression [101]. However, the role of CpG methylation in gene regulation is not always associated with gene repression especially when it occurs at enhancers or imprinting control regions [54]. Therefore, more recently, the TALE:TET1 epigenome editor has been used to target the imprinting control region 2 (ICR2) of p57 (a negative regulator of cell proliferation) to reduce its expression and stimulate the proliferation of β-cells, which can be used as replacement therapy in the context of type I and type II diabetes [122].
Other strategies with a focus on altering disease-associated epigenetic marks include TALE DBDs tethered with DNMTs to promote DNA methylation at specific loci. TALE:DNMT fusions have been used to silence the expression of CDKN2A, encoding the p16 protein that participates in cell senescence, with the aim of increasing the replication of primary human fibroblasts [123].
TALEs have also been fused with histone modifiers, such as KDM1A, which demethylates H3K4me2 and promotes the interaction with HDAC. When tested in a leukemia cell line, TALE:KDM1A was able to target an active H3K4me2+ and H3K27ac+ enhancer of the SCL gene (encoding for a transcription factor with critical function in hematopoiesis), and to significantly reduce histone methylation and acetylation, and as a consequence, SCL expression [124]. Furthermore, TALEs fused with HDAC8 were able to reduce acetylation of the Grm2 promoter in primary neurons and consequently downregulate Grm2. However, when tested on other genes in different cell types, TALE:HDAC8 showed variable effects on gene repression, suggesting that the chromatin state influences the outcome [121].
Of note, several studies reported how the interaction between different epigenetic modifications play a crucial role in stable regulation of gene expression over time. In this regard, several studies combined different tools simultaneously introducing multiple epigenetic modifications. TALEs fused with KRAB, DNMT3A and DNMT3L have been individually tested to silence B2M expression in a leukemia cell line, obtaining no or transient effects. However, co-delivery of these three components significantly reduced B2M expression, and the effect was stable after numerous cell divisions. Moreover, the same platform was tested for silencing other endogenous genes, namely, interferon alpha/beta/omega receptor 1 (IFNAR1) and vascular endothelial growth factor A (VEGFA), confirming the crucial role of the interaction between epigenetic modifications in gene regulation [125].
Following these observations, other tools have been developed to perform combinatorial epigenetic modifications. Designer epigenome modifiers combining a single TALE with KRAB, DNMT3A and DNMT3L have been tested in primary human CD4+ T cells to efficiently silence CCR5 and CXCR4. Interestingly, when splitting the KRAB and the DNMT activity into two distinct molecules, they failed to promote gene silencing [126].
Similarly to ZF DBDs, the off-target activity is one of the major concerns regarding TALE-based technology [101]. A number of studies have reported low but detectable off-target activity both in vitro and in vivo [113,127,128]. However, this depends on the specific TALEs, which can have differential off-target binding [113,129]. Moreover, a study showed that no changes in gene expression at off-target sites have been reported with TALE DBDs [130]. In addition, several improvements have been implemented to increase the binding specificity of TALE DBDs, such as the presence of divalent cations [131].
Another significant limitation of TALEs is their large number of tandem repeats, which makes TALE-expressing plasmid cloning and delivery difficult. However, after optimization and minimization of the number of tandem repeats, delivery as plasmid DNA or in vitro transcribed mRNA has been successful [126], as well as delivery with lentiviral [123], adenoviral [122] and adeno-associated viral (AAV) vectors. The latter are the preferred vectors for gene transfer in vivo, but have a limited packaging size, and thus, they can only express TALEs combined with functional domains of small size [121].

3.3. CRISPR/Cas9-Based Epigenome Editors

The CRISPR/Cas9 technology is commonly used for epigenome editing applications, thanks to its easier design and flexibility. The CRISPR/Cas9 system is based on a guide RNA (gRNA) which drives the Cas9 nuclease to the protospacer or target site, which has to be located upstream of a protospacer adjacent motif (PAM). The recognition of the target site is directed by the complementarity between the gRNA spacer and the DNA protospacer and allows the Cas9 nuclease to create a double-strand break (DSB) at the target site. Adaptations of Cas9 have been developed to create nuclease-deficient variants. For instance, the catalytically inactive dead Cas9 nuclease (dCas9 that does not generate DSBs but retains binding activity) has been fused to TFs or to epigenetic modifiers to modulate gene expression.
dCas9 was initially fused with VP64 and p65 (creating dCas9:VP64 and dCas9:p65) which were first used to target and activate GFP+ reporter gene in HEK293 cells [132]. The same tools have subsequently been used to target the promoter of endogenous genes, such as VEGFA and NFT3, increasing their expression in HEK293 cells [133]. KRAB domain has also been tethered to dCas9, and the first attempt was performed in HEK293 cells, whereby this tool efficiently reduced the expression of a GFP+ reporter gene [132]. More recently, dCas9:KRAB was used to develop a gene therapy strategy for retinitis pigmentosa. dCas9:KRAB was successfully delivered by AAV with the aim to repress the expression of Nrl gene, a master regulator of rod photoreceptor determination, which mediates the in situ reprogramming of rod cells into cone-like cells that are resistant to retinitis pigmentosa-specific mutations [134]. Moreover, dCas9:KRAB was shown to be able to cause potent and specific repression of PTEN (encoding a phosphatase that limits cell growth), in human cell lines and neural cells derived from human-induced pluripotent stem cells (iPSCs). Thus, suppression of PTEN resulted in a robust central nervous system (CNS) axon regrowth, providing a new option for promoting axon regeneration and functional recovery after CNS trauma [135].
Of note, experiments targeting both gene enhancers and promoters revealed that the design of the tools is critical for their activity. By way of example, the position of the fusion site between dCas9 and the TF, and the orientation of dCas9 and the effector domain can strongly impact the editing efficiency [132,136]. Moreover, dCas9:VP64 and dCas9:p300 have a lower efficiency compared to TALE:VP64 and TALE:p300, respectively, when targeting gene promoters, probably because the dCas9 fusions interfere with the binding of TFs [133,137].
The first studies aimed at optimizing the tools by increasing the number of transcriptional domains fused to the dCas9, leading to the generation of activators such as dCas9:VP160, which contains ten repeats of VP16 domain [138], or dCas9:VPR, where dCas9 was fused with a tripartite activator containing VP64, p65 and Rta [139]. More recently, dCas9 tethered with the VPR domain (MiniCAFE) was used to efficiently activate gene expression in Caenorhabditis elegans, and in murine and human cells [140].
However, a more stable epigenetic modification has been obtained by fusing dCas9 with domains of endogenous epigenome modifiers. dCas9 was initially fused to DNMT3A alone or in tandem with its cofactor DNMT3L, to obtain efficient site-specific DNA methylation and consequently transcriptional repression at several human genes. dCas9:DNMT3A, for instance, led to a therapeutic downregulation of SNCA in iPSCs from Parkinson’s disease patients, who typically present high SNCA levels [141]. Similarly, dCas9:DNMT3A-3L was used to target CXCR4 and TFRC leading to gene repression in SKOV-3 and HEK293 cell lines [142]. To increase the stability of the epigenetic repression, a combination of this dCas9:DNMT3A and dCas9 fused with the KRAB domain was used to stably silence endogenous genes, namely, VEGFA and IFNAR1 [125]. More recently, an optimized enzyme containing dCas9 fused with DNMT3A, DNMT3L and KRAB domain was successfully used to repress gene expression in iPSCs and other cell types and maintain a stable repression in iPSC-derived neurons [143].
To reactivate gene expression, CRISPR/Cas9-based epigenome editors capable of demethylating DNA have been developed. To this end, TET1 has been tethered to dCas9 targeting the promoter of BDNF in post-mitotic neurons, a gene that encodes a protein involved in neuronal survival and differentiation, leading to reduced DNA methylation and BDNF re-expression [144]. The same tool has been used in the context of fragile X syndrome (FXS), a disease characterized by a silencing of the FMR1 gene associated with hypermethylation of the CGG expanded repeats in the 5′UTR of FMR1. dCas9:TET1 led to efficient demethylation of the CGG repeats and to a consequent FMR1 reactivation in vivo in mice [145]. dCas9:TET1 has also been used to demethylate the promoter of the onco-suppressor BRCA1, which is often hypermethylated in breast cancer cells. In this case, dCas9:TET1 increased BRCA1 expression and inhibited the proliferation of a cancer cell line, highlighting the therapeutic potential of epigenome editing to reduce hypermethylation in promoters of tumor suppressor genes, which is one of the hallmarks of cancer [146]. dCas9:TET1 has also been used in Jurkat T cells to drive the demethylation of a regulatory region (FOXP3-TSDR; Treg specific demethylated region) of FOXP3, a master transcription factor of regulatory T (Treg) cells. Consequently, this approach might represent a tool for programming of primary T cells toward the Treg phenotype [147]. However, another study in primary murine T-lymphocytes proved that demethylation of the Foxp3 enhancer alone was insufficient to stabilize the Foxp3 expression and modulation of histone modifications was also necessary to activate the gene [148]. Finally, dCas9:TET1 has been used to study the role of TET1 in cardiac progenitor differentiation from murine embryonic stem cells (ESCs) [149]. Targeting the promoter of Hand1 (encoding a factor that plays a key role in cardiac development) using dCas9:TET1 up-regulated Hand1 expression, it was shown that TET1 and DNA methylation play a crucial role in regulating gene expression during cardiac differentiation [149].
CRISPR/Cas9-based epigenome editors able to modulate histone modifications have been generated by fusing dCas9 with different histone modifiers. For instance, the catalytic core domain of the human HAT p300 protein was fused to dCas9 to deposit H3K27 acetylation at both promoters and enhancers (IL1RN, MYOD and OCT4 gene promoters and HS2 enhancer of the β-globin locus) and activate gene expression in HEK293T cells [103,130,140]. More recently, dCas9:p300 has been used in the context of cystic fibrosis leading to elevated expression of the CFTR gene (affected in this disease) with a consequent restoration of the normal phenotype in patient-derived human bronchial epithelial cells when combined with a pharmaceutical treatment [150].
Conversely, targeted removal of histone acetylation can be achieved using HDAC fused with dCas9 resulting in gene repression, although one study has highlighted the importance of cellular context-dependent effects when using HDAC-containing epigenome editors. This work showed that histone deacetylation by some CRISPR/Cas9 epigenome editors can drive either gene repression or activation depending on the chromatin context and cell type. In particular, dCas9:HDAC3 was used to remove H3K27ac from the Mecp2 promoter in two murine cell lines expressing different Mecp2 levels; this led to activation or repression of gene expression according to the cell-specific chromatin state of the promoter [151].
Deactivation of enhancers and gene repression can likewise be achieved by using dCas9 fused to the histone demethylase KDM1A, which eliminates H3K4me2, a histone mark typically found at active promoters and enhancers. A first attempt to use dCas9:KDM1A was performed in murine ESCs, targeting the enhancers of genes that encode factors critical for maintaining the stem cell state (Oct4 and Sox2); this led to the loss of expression of these genes and consequent phenotypic changes [152]. Recently, dCas9:KDM1A has been used to target cis-regulatory elements in chicken embryos and alter the gene expression [153]. In this study, a comparison of dCas9:KDM1A and dCas9:KRAB suggests a more efficient silencing by KDM1A when used at early stages of gene activation, whereas KRAB-mediated repression might be more potent with genes already engaged in transcription [153].
H3K9me3 and H3K27me3 histone modifications can also be deposited at target loci using dCas9 fused with Suppressor of variegation 3–9 homolog 1(SUV39H1) and EZH2, respectively. However, these studies showed that a repressive histone modification alone is not sufficient for gene silencing [154]. Indeed, recent approaches combined different epigenetic modifications to achieve a more predictable and persistent silencing; e.g., strategies combining Cas9:EZH2d and DNMT3 to achieve gene silencing [155].
Overall, CRISPR/Cas9-based epigenome editors present several advantages compared to ZFs and TALEs. The main advantage of the CRISPR/Cas9 system is the ease of designing a single guide RNA (sgRNA) targeting a specific region compared to the protein design required for TALE- and ZF-based epigenome editors. Off-target binding can also occur with the CRISPR/Cas9 system [156]. However, when off-target binding was observed, changes in gene expression or even in epigenetic marks and chromatin accessibility were not detected outside of the intended target site [103,136,155]. Furthermore, CRISPR/Cas9 off-target effects can be reduced to undetectable levels with high-fidelity Cas9 variants. In particular, different versions of Cas9 have been developed, in which residues that normally make nonspecific contacts with the DNA are mutated [157]. These Cas9 variants have low or no off-target activity, while maintaining a high on-target efficiency [157,158]. Finally, the use of the CRISPR/Cas9 system facilitates simultaneous epigenome editing of multiple regions with the delivery of only one Cas9 enzyme [138,159].
CRISPR/Cas9-based epigenome editors have been successfully delivered into cells using plasmid [125], RNA [126], ribonucleoprotein [160] and lentiviral vectors [161]. However, the use of Cas9 DBDs would require vectors of large capacity for in vivo applications. Because of their limited packaging size, AAVs cannot accommodate dCas9 fused with the effector domain. To overcome this limitation, Cas9-based epigenome editors have been split into two subdomains and delivered using two different AAVs [134]. Alternatively, recently engineered smaller Cas orthologs can be accommodated in AAVs. Finding an appropriate delivery method remains crucial in developing Cas9-based epigenome editing in vivo.

3.4. Epigenetic Approaches to Modulate β-like Globin Expression

The importance of epigenetic regulations on globin gene expression has fueled the identification of novel medications capable of directly altering the epigenome for therapeutic purposes. Epigenetic regulation can act through multiple mechanisms that eventually lead to chromatin alterations that either permit or inhibit transcription [162]. Both DNA methylation and histone modifications, such as acetylation and methylation, play a crucial role in regulating the expression of β-globin chains [55,163].
For example, DNA methylation of the CpG dinucleotides at the HBG promoters negatively correlates with γ-globin gene expression in primary erythroblasts [55,57]. Furthermore, HBG promoters are hypomethylated in individuals harboring HPFH mutations [58]. As a consequence, DNA methyltransferase (DNMT) inhibitors, such as decitabine, have been used to treat patients with β-thalassemia and SCD. Clinical trials showed that decitabine is well tolerated and is efficacious in increasing HbF levels in both β-thalassemia [164] and SCD [165] patients. However, decitabine also had the effect of increasing platelet counts and has therefore yet to be approved as a treatment for the diseases due to concerns regarding its adverse effects. New compounds, such as GSK3482364, are currently being tested in pre-clinical studies [166].
Activating histone modifications, such as H3K27ac or H3K4me3, mark active HBG promoters in fetal but not in adult erythroblasts. HDAC inhibitors, such as vorinostat, have been used in clinical trials with the aim of reactivating HbF [167]. Vorinostat is a pan-HDAC inhibitor approved for the treatment of T-cell lymphoma and significantly upregulates γ-globin expression in erythroid cell lines and primary human HSPC-derived erythroblasts. However, the clinical trial conducted to evaluate its efficacy in SCF patients was terminated early due to poor patient recruitment. By contrast, a specific class I HDAC inhibitor leads to HbF reactivation and increased acetylation of H3 and H4 in the γ-globin promoters in erythroid cells derived from treated SCD HSPCs [168], and it could be further tested in clinical studies.
In addition, other drugs interfering with the activity of epigenetic modifiers modulating histone methylations have been used to reactivate HbF and will likely be further developed, such as RN-1, an inhibitor of KDM1A [169,170,171], UNC0638, an inhibitor of EHMT1/2 [70,71], and FTX-6058, an inhibitor of EED [172].
However, the mode of action of these pharmacological inhibitors are often uncertain as they are non-specific, and their global effect poses serious safety concerns. For instance, down-regulation of components of chromatin-remodeling and modifier complexes (e.g., HDACs, DNMT1) via shRNA or CRISPR/Cas9 is associated with HbF upregulation [43,173]. However, in many cases, this led to impaired cell fitness [174] as these factors play a pleiotropic role on the establishment and maintenance of the epigenetic and transcriptional cellular profiles. For the same reason, the mechanisms underlying HbF reactivation upon down-regulation of these factors are still not clear.
Several gene therapy strategies for reactivating the expression of the endogenous fetal γ-globin have been successfully developed. However, although they offer the opportunity to achieve permanent therapeutic changes in the genome, these approaches still rely on the intermediate formation of DSBs. This in turn poses serious safety concerns as the introduction of DSBs might result in unpredictable genotoxic effects [175]. Thus, a technology capable of silencing the expression of a target gene without altering the underlying genomic sequence is desirable.
Artificial transcription factors and epigenome editors have been used to directly reactivate human γ-globin or β-globin expression and to study the mechanisms of β-like globin gene regulation. The use of an artificial ZF transcriptional activator ZF:VP64 targeting the HBG promoters successfully increased HbF expression in primary human erythroid cells [98] and an in vivo study also demonstrated increased γ-globin gene expression in the peripheral blood from β-YAC transgenic mice stably expressing the artificial ZF transcriptional activator [176]. Nonetheless, the use of this tool was associated with some cell toxicity [98]. Fusion of the TET1 demethylase to a TALE DBD targeted to the β-globin promoter efficiently reactivated the HBB gene through targeted DNA demethylation in an HEK293 cell line [133].
Both dCas9:p300 [102] and dCas9:KRAB were used to target the HS2 enhancer, which regulates the expression of β-like globin genes. While dCas9:p300 caused H3K27 acetylation and activation of globin gene expression in HEK293T cells [103], dCas9:KRAB-induced H3K9 trimethylation at the HS2 enhancer and a consequent reduction in chromatin accessibility at the enhancer and globin gene promoters, demonstrating that repression mediated by dCas9:KRAB is able to reduce enhancer activity via local modification of the epigenome [177].
However, since epigenetic regulation at the globin loci is complex, it is reasonable to believe that the fine-tuned expression of the different genes requires the interplay of different epigenetic modifications in parallel to the recruitment of TFs. The first attempt to both deposit histone modifications and recruit TFs was performed by taking advantage of the natural interaction that occurs between the phage MS2 hairpins and MS2 bacteriophage coat protein (MCP). In particular, dCas9:p300 was fused to MS2-sgRNA sequence which recruits the VP64 activator domains fused with MCP and targets the HS2 enhancer. This allowed the concomitant introduction of H3K27ac, the recruitment of a transcriptional activator and, as a consequence, the potent activation of β-like globin genes in HEK293T cells [178].

4. Conclusions and Perspectives

The regulation of β-like globin gene expression has been extensively studied, which has led the identification of different layers of regulation, involving transcription factors, chromatin looping and epigenome modifications of the β-globin locus. The interplay between TFs, DNA methylation and histone modifications play a crucial role in determining gene activation or repression.
The availability of genome editing tools has allowed the development of therapeutic approaches for β-hemoglobinopathies through the direct genetic modification of HSCs that either correct the disease-causing mutation or reactivate fetal hemoglobin expression by altering transcription factor occupancy. However, genome editing tools can cause a DNA damage response and unwanted genomic alterations. By contrast, epigenome editing would allow the modulation of gene expression (e.g., re-activation of HBG genes) in a scarless manner, i.e., without affecting genome integrity. Moreover, epigenome editing is more targeted as compared to drugs able to change the epigenetic profile of the β-globin locus (such as HDAC and DNMT inhibitors).
Insights for the usage of epigenome editing as a therapeutic approach for hemoglobinopathies have been provided in cell lines and primary erythroid cells, through the therapeutic reactivation of HbF. Nevertheless, there are several steps that need to be taken before such an approach moves to a clinical phase. For instance, further studies are required in order to achieve persistent gene activation after transient exposure to the epigenome modifier, and the persistence of epigenome modifications (preferably without the constitutive expression of the epigenetic modulators) needs to be evaluated in long-term repopulating of patient HSCs. Furthermore, an extensive assessment of the off-target activity of epigenome modifiers is crucial, and last but not least, it is vital that clinically relevant protocols are developed for the delivery of this system.

Author Contributions

Visualization, L.F. and P.A.; Writing—original draft, L.F., A.M. and P.A.; Writing—review & editing, L.F., Z.A., A.M. and P.A.; Funding acquisition, A.M.; Supervision, A.M. and P.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by State funding from the Agence Nationale de la Recherche under “Investissements d’avenir” program [ANR-10-IAHU-01 and ANR-21-CE18-0066-01], by the European Research Council [865797 DITSB] and by Fondation pour la Recherche Médicale [FRM PLP202110014595].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Manning, L.R.; Russell, J.E.; Padovan, J.C.; Chait, B.T.; Popowicz, A.; Manning, R.S.; Manning, J.M. Human Embryonic, Fetal, and Adult Hemoglobins Have Different Subunit Interface Strengths. Correlation with Lifespan in the Red Cell. Protein Sci. 2007, 16, 1641–1658. [Google Scholar] [CrossRef] [Green Version]
  2. Jonxis, J.H.P. The Development of Hemoglobin. Pediatr. Clin. N. Am. 1965, 12, 535–550. [Google Scholar] [CrossRef]
  3. Lorkin, P.A. Fetal and Embryonic Haemoglobins. J. Med. Genet. 1973, 10, 50–64. [Google Scholar] [CrossRef] [Green Version]
  4. Maston, G.A.; Evans, S.K.; Green, M.R. Transcriptional Regulatory Elements in the Human Genome. Annu. Rev. Genom. Hum. Genet. 2006, 7, 29–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Perutz, M.F. Mechanisms of Cooperativity and Allosteric Regulation in Proteins. Quart. Rev. Biophys. 1989, 22, 139–237. [Google Scholar] [CrossRef]
  6. Manning, L.R.; Russell, J.E.; Popowicz, A.M.; Manning, R.S.; Padovan, J.C.; Manning, J.M. Energetic Differences at the Subunit Interfaces of Normal Human Hemoglobins Correlate with Their Developmental Profile. Biochemistry 2009, 48, 7568–7574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Efstratiadis, A. The Structure and Evolution of the Human β-Globin Gene Family. Cell 1980, 21, 653–668. [Google Scholar] [CrossRef] [PubMed]
  8. Mettananda, S.; Gibbons, R.J.; Higgs, D.R. Understanding α-Globin Gene Regulation and Implications for the Treatment of β-Thalassemia: α-Globin Regulation and β-Thalassemia. Ann. N. Y. Acad. Sci. 2016, 1368, 16–24. [Google Scholar] [CrossRef] [PubMed]
  9. Martyn, G.E.; Quinlan, K.G.R.; Crossley, M. The Regulation of Human Globin Promoters by CCAAT Box Elements and the Recruitment of NF-Y. Biochim. Et Biophys. Acta (BBA) Gene Regul. Mech. 2017, 1860, 525–536. [Google Scholar] [CrossRef]
  10. Philipsen, S.; Talbot, D.; Fraser, P.; Grosveld, F. The Beta-Globin Dominant Control Region: Hypersensitive Site 2. EMBO J. 1990, 9, 2159–2167. [Google Scholar] [CrossRef]
  11. Liu, X.; Zhang, Y.; Chen, Y.; Li, M.; Zhou, F.; Li, K.; Cao, H.; Ni, M.; Liu, Y.; Gu, Z.; et al. In Situ Capture of Chromatin Interactions by Biotinylated DCas9. Cell 2017, 170, 1028–1043.e19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Li, Q.; Stamatoyannopoulos, G. Hypersensitive Site 5 of the Human Beta Locus Control Region Functions as a Chromatin Insulator. Blood 1994, 84, 1399–1401. [Google Scholar] [CrossRef] [Green Version]
  13. Himadewi, P.; Wang, X.Q.D.; Feng, F.; Gore, H.; Liu, Y.; Yu, L.; Kurita, R.; Nakamura, Y.; Pfeifer, G.P.; Liu, J.; et al. 3’HS1 CTCF Binding Site in Human β-Globin Locus Regulates Fetal Hemoglobin Expression. Elife 2021, 10, e70557. [Google Scholar] [CrossRef] [PubMed]
  14. Talbot, D.; Philipsen, S.; Fraser, P.; Grosveld, F. Detailed Analysis of the Site 3 Region of the Human Beta-Globin Dominant Control Region. EMBO J. 1990, 9, 2169–2177. [Google Scholar] [CrossRef] [PubMed]
  15. Qin, K.; Huang, P.; Feng, R.; Keller, C.A.; Peslak, S.A.; Khandros, E.; Saari, M.S.; Lan, X.; Mayuranathan, T.; Doerfler, P.A.; et al. Dual Function NFI Factors Control Fetal Hemoglobin Silencing in Adult Erythroid Cells. Nat. Genet. 2022, 54, 874–884. [Google Scholar] [CrossRef]
  16. Bao, X.; Zhang, X.; Wang, L.; Wang, Z.; Huang, J.; Zhang, Q.; Ye, Y.; Liu, Y.; Chen, D.; Zuo, Y.; et al. Epigenetic Inactivation of ERF Reactivates γ-Globin Expression in β-Thalassemia. Am. J. Hum. Genet. 2021, 108, 709–721. [Google Scholar] [CrossRef]
  17. Fugazza, C.; Barbarani, G.; Elangovan, S.; Marini, M.G.; Giolitto, S.; Font-Monclus, I.; Marongiu, M.F.; Manunza, L.; Strouboulis, J.; Cantù, C.; et al. The Coup-TFII Orphan Nuclear Receptor Is an Activator of the γ-Globin Gene. Haematologica 2020, 106, 474–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Chambers, C.B.; Gross, J.; Pratt, K.; Guo, X.; Byrnes, C.; Lee, Y.T.; Lavelle, D.; Dean, A.; Miller, J.L.; Wilber, A. The MRNA-Binding Protein IGF2BP1 Restores Fetal Hemoglobin in Cultured Erythroid Cells from Patients with β-Hemoglobin Disorders. Mol. Ther. Methods Clin. Dev. 2020, 17, 429–440. [Google Scholar] [CrossRef]
  19. Feng, R.; Mayuranathan, T.; Huang, P.; Doerfler, P.A.; Li, Y.; Yao, Y.; Zhang, J.; Palmer, L.E.; Mayberry, K.; Christakopoulos, G.E.; et al. Activation of γ-Globin Expression by Hypoxia-Inducible Factor 1α. Nature 2022, 610, 783–790. [Google Scholar] [CrossRef]
  20. Doerfler, P.A.; Feng, R.; Li, Y.; Palmer, L.E.; Porter, S.N.; Bell, H.W.; Crossley, M.; Pruett-Miller, S.M.; Cheng, Y.; Weiss, M.J. Activation of γ-Globin Gene Expression by GATA1 and NF-Y in Hereditary Persistence of Fetal Hemoglobin. Nat. Genet. 2021, 53, 1177–1186. [Google Scholar] [CrossRef]
  21. Liu, N.; Xu, S.; Yao, Q.; Zhu, Q.; Kai, Y.; Hsu, J.Y.; Sakon, P.; Pinello, L.; Yuan, G.-C.; Bauer, D.E.; et al. Transcription Factor Competition at the γ-Globin Promoters Controls Hemoglobin Switching. Nat. Genet. 2021, 53, 511–520. [Google Scholar] [CrossRef] [PubMed]
  22. Wienert, B.; Martyn, G.E.; Kurita, R.; Nakamura, Y.; Quinlan, K.G.R.; Crossley, M. KLF1 Drives the Expression of Fetal Hemoglobin in British HPFH. Blood 2017, 130, 803–807. [Google Scholar] [CrossRef] [PubMed]
  23. Zhou, D.; Liu, K.; Sun, C.-W.; Pawlik, K.M.; Townes, T.M. KLF1 Regulates BCL11A Expression and γ- to β-Globin Gene Switching. Nat. Genet. 2010, 42, 742–744. [Google Scholar] [CrossRef]
  24. Grevet, J.D.; Lan, X.; Hamagami, N.; Edwards, C.R.; Sankaranarayanan, L.; Ji, X.; Bhardwaj, S.K.; Face, C.J.; Posocco, D.F.; Abdulmalik, O.; et al. Domain-Focused CRISPR Screen Identifies HRI as a Fetal Hemoglobin Regulator in Human Erythroid Cells. Science 2018, 361, 285–290. [Google Scholar] [CrossRef] [Green Version]
  25. Huang, P.; Peslak, S.A.; Lan, X.; Khandros, E.; Yano, J.A.; Sharma, M.; Keller, C.A.; Giardine, B.; Qin, K.; Abdulmalik, O.; et al. The HRI-Regulated Transcription Factor ATF4 Activates BCL11A Transcription to Silence Fetal Hemoglobin Expression. Blood 2020, 135, 2121–2132. [Google Scholar] [CrossRef] [PubMed]
  26. Xu, J.; Sankaran, V.G.; Ni, M.; Menne, T.F.; Puram, R.V.; Kim, W.; Orkin, S.H. Transcriptional Silencing of γ-Globin by BCL11A Involves Long-Range Interactions and Cooperation with SOX6. Genes Dev. 2010, 24, 783–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Norton, L.J.; Funnell, A.P.W.; Burdach, J.; Wienert, B.; Kurita, R.; Nakamura, Y.; Philipsen, S.; Pearson, R.C.M.; Quinlan, K.G.R.; Crossley, M. KLF1 Directly Activates Expression of the Novel Fetal Globin Repressor ZBTB7A/LRF in Erythroid Cells. Blood Adv. 2017, 1, 685–692. [Google Scholar] [CrossRef] [Green Version]
  28. Bianchi, E.; Zini, R.; Salati, S.; Tenedini, E.; Norfo, R.; Tagliafico, E.; Manfredini, R.; Ferrari, S. C-Myb Supports Erythropoiesis through the Transactivation of KLF1 and LMO2 Expression. Blood 2010, 116, e99–e110. [Google Scholar] [CrossRef] [Green Version]
  29. Boontanrart, M.Y.; Schröder, M.S.; Stehli, G.M.; Banović, M.; Wyman, S.K.; Lew, R.J.; Bordi, M.; Gowen, B.G.; DeWitt, M.A.; Corn, J.E. ATF4 Regulates MYB to Increase γ-Globin in Response to Loss of β-Globin. Cell Rep. 2020, 32, 107993. [Google Scholar] [CrossRef]
  30. Lan, X.; Ren, R.; Feng, R.; Ly, L.C.; Lan, Y.; Zhang, Z.; Aboreden, N.; Qin, K.; Horton, J.R.; Grevet, J.D.; et al. ZNF410 Uniquely Activates the NuRD Component CHD4 to Silence Fetal Hemoglobin Expression. Mol. Cell 2021, 81, 239–254.e8. [Google Scholar] [CrossRef]
  31. Vinjamur, D.S.; Yao, Q.; Cole, M.A.; McGuckin, C.; Ren, C.; Zeng, J.; Hossain, M.; Luk, K.; Wolfe, S.A.; Pinello, L.; et al. ZNF410 Represses Fetal Globin by Singular Control of CHD4. Nat. Genet. 2021, 53, 719–728. [Google Scholar] [CrossRef]
  32. Wadman, I.A. The LIM-Only Protein Lmo2 Is a Bridging Molecule Assembling an Erythroid, DNA-Binding Complex Which Includes the TAL1, E47, GATA-1 and Ldb1/NLI Proteins. EMBO J. 1997, 16, 3145–3157. [Google Scholar] [CrossRef]
  33. Song, S.-H.; Hou, C.; Dean, A. A Positive Role for NLI/Ldb1 in Long-Range β-Globin Locus Control Region Function. Mol. Cell 2007, 28, 810–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Deng, W.; Rupon, J.W.; Krivega, I.; Breda, L.; Motta, I.; Jahn, K.S.; Reik, A.; Gregory, P.D.; Rivella, S.; Dean, A.; et al. Reactivation of Developmentally Silenced Globin Genes by Forced Chromatin Looping. Cell 2014, 158, 849–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Krivega, I.; Dale, R.K.; Dean, A. Role of LDB1 in the Transition from Chromatin Looping to Transcription Activation. Genes Dev. 2014, 28, 1278–1290. [Google Scholar] [CrossRef] [Green Version]
  36. Yun, W.J.; Kim, Y.W.; Kang, Y.; Lee, J.; Dean, A.; Kim, A. The Hematopoietic Regulator TAL1 Is Required for Chromatin Looping between the β-Globin LCR and Human γ-Globin Genes to Activate Transcription. Nucleic Acids Res. 2014, 42, 4283–4293. [Google Scholar] [CrossRef]
  37. Lettre, G.; Sankaran, V.G.; Bezerra, M.A.C.; Araújo, A.S.; Uda, M.; Sanna, S.; Cao, A.; Schlessinger, D.; Costa, F.F.; Hirschhorn, J.N.; et al. DNA Polymorphisms at the BCL11A, HBS1L-MYB, and β- Globin Loci Associate with Fetal Hemoglobin Levels and Pain Crises in Sickle Cell Disease. Proc. Natl. Acad. Sci. USA 2008, 105, 11869–11874. [Google Scholar] [CrossRef] [Green Version]
  38. Uda, M.; Galanello, R.; Sanna, S.; Lettre, G.; Sankaran, V.G.; Chen, W.; Usala, G.; Busonero, F.; Maschio, A.; Albai, G.; et al. Genome-Wide Association Study Shows BCL11A Associated with Persistent Fetal Hemoglobin and Amelioration of the Phenotype of β-Thalassemia. Proc. Natl. Acad. Sci. USA 2008, 105, 1620–1625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Bauer, D.E.; Kamran, S.C.; Lessard, S.; Xu, J.; Fujiwara, Y.; Lin, C.; Shao, Z.; Canver, M.C.; Smith, E.C.; Pinello, L.; et al. An Erythroid Enhancer of BCL11A Subject to Genetic Variation Determines Fetal Hemoglobin Level. Science 2013, 342, 253–257. [Google Scholar] [CrossRef] [Green Version]
  40. Liu, N.; Hargreaves, V.V.; Zhu, Q.; Kurland, J.V.; Hong, J.; Kim, W.; Sher, F.; Macias-Trevino, C.; Rogers, J.M.; Kurita, R.; et al. Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch. Cell 2018, 173, 430–442.e17. [Google Scholar] [CrossRef] [Green Version]
  41. Basak, A.; Sankaran, V.G. Regulation of the Fetal Hemoglobin Silencing Factor BCL11A: BCL11A Regulation in Hemoglobin Expression. Ann. N. Y. Acad. Sci. 2016, 1368, 25–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Sankaran, V.G.; Menne, T.F.; Xu, J.; Akie, T.E.; Lettre, G.; Van Handel, B.; Mikkola, H.K.A.; Hirschhorn, J.N.; Cantor, A.B.; Orkin, S.H. Human Fetal Hemoglobin Expression Is Regulated by the Developmental Stage-Specific Repressor BCL11A. Science 2008, 322, 1839–1842. [Google Scholar] [CrossRef] [Green Version]
  43. Xu, J.; Bauer, D.E.; Kerenyi, M.A.; Vo, T.D.; Hou, S.; Hsu, Y.-J.; Yao, H.; Trowbridge, J.J.; Mandel, G.; Orkin, S.H. Corepressor-Dependent Silencing of Fetal Hemoglobin Expression by BCL11A. Proc. Natl. Acad. Sci. USA 2013, 110, 6518–6523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Martyn, G.E.; Wienert, B.; Yang, L.; Shah, M.; Norton, L.J.; Burdach, J.; Kurita, R.; Nakamura, Y.; Pearson, R.C.M.; Funnell, A.P.W.; et al. Natural Regulatory Mutations Elevate the Fetal Globin Gene via Disruption of BCL11A or ZBTB7A Binding. Nat. Genet. 2018, 50, 498–503. [Google Scholar] [CrossRef]
  45. Masuda, T.; Wang, X.; Maeda, M.; Canver, M.C.; Sher, F.; Funnell, A.P.W.; Fisher, C.; Suciu, M.; Martyn, G.E.; Norton, L.J.; et al. Transcription Factors LRF and BCL11A Independently Repress Expression of Fetal Hemoglobin. Science 2016, 351, 285–289. [Google Scholar] [CrossRef] [Green Version]
  46. Weatherall, D.J.; Clegg, J.B.; Knox-Macaulay, H.H.M.; Bunch, C.; Hopkins, C.R.; Temperley, I.J. A Genetically Determined Disorder with Features Both of Thalassaemia and Congenital Dyserythropoietic Anaemia. Br. J. Haematol. 1973, 24, 681–702. [Google Scholar] [CrossRef]
  47. Fernandes, Q. Therapeutic Strategies in Sickle Cell Anemia: The Past Present and Future. Life Sci. 2017, 178, 100–108. [Google Scholar] [CrossRef] [PubMed]
  48. Forget, B.G. Molecular Basis of Hereditary Persistence of Fetal Hemoglobin. Ann. N. Y Acad. Sci. 1998, 850, 38–44. [Google Scholar] [CrossRef]
  49. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef] [Green Version]
  50. Li, E.; Bestor, T.H.; Jaenisch, R. Targeted Mutation of the DNA Methyltransferase Gene Results in Embryonic Lethality. Cell 1992, 69, 915–926. [Google Scholar] [CrossRef]
  51. He, Y.-F.; Li, B.-Z.; Li, Z.; Liu, P.; Wang, Y.; Tang, Q.; Ding, J.; Jia, Y.; Chen, Z.; Li, L.; et al. Tet-Mediated Formation of 5-Carboxylcytosine and Its Excision by TDG in Mammalian DNA. Science 2011, 333, 1303–1307. [Google Scholar] [CrossRef] [Green Version]
  52. Ito, S.; Shen, L.; Dai, Q.; Wu, S.C.; Collins, L.B.; Swenberg, J.A.; He, C.; Zhang, Y. Tet Proteins Can Convert 5-Methylcytosine to 5-Formylcytosine and 5-Carboxylcytosine. Science 2011, 333, 1300–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ruiz, M.A.; Rivers, A.; Ibanez, V.; Vaitkus, K.; Mahmud, N.; DeSimone, J.; Lavelle, D. Hydroxymethylcytosine and Demethylation of the γ-Globin Gene Promoter during Erythroid Differentiation. Epigenetics 2015, 10, 397–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Charlet, J.; Duymich, C.E.; Lay, F.D.; Mundbjerg, K.; Dalsgaard Sørensen, K.; Liang, G.; Jones, P.A. Bivalent Regions of Cytosine Methylation and H3K27 Acetylation Suggest an Active Role for DNA Methylation at Enhancers. Mol. Cell 2016, 62, 422–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Mabaera, R.; Richardson, C.A.; Johnson, K.; Hsu, M.; Fiering, S.; Lowrey, C.H. Developmental- and Differentiation-Specific Patterns of Human Gamma- and Beta-Globin Promoter DNA Methylation. Blood 2007, 110, 1343–1352. [Google Scholar] [CrossRef] [Green Version]
  56. Bao, X.; Zuo, Y.; Chen, D.; Zhao, C. DNA Methylation Patterns of β-Globin Cluster in β-Thalassemia Patients. Clin. Epigenetics 2020, 12, 187. [Google Scholar] [CrossRef]
  57. Lessard, S.; Beaudoin, M.; Benkirane, K.; Lettre, G. Comparison of DNA Methylation Profiles in Human Fetal and Adult Red Blood Cell Progenitors. Genome. Med. 2015, 7, 1. [Google Scholar] [CrossRef] [Green Version]
  58. Goren, A.; Simchen, G.; Fibach, E.; Szabo, P.E.; Tanimoto, K.; Chakalova, L.; Pfeifer, G.P.; Fraser, P.J.; Engel, J.D.; Cedar, H. Fine Tuning of Globin Gene Expression by DNA Methylation. PLoS ONE 2006, 1, e46. [Google Scholar] [CrossRef] [Green Version]
  59. Gong, Y.; Zhang, X.; Zhang, Q.; Zhang, Y.; Ye, Y.; Yu, W.; Shao, C.; Yan, T.; Huang, J.; Zhong, J.; et al. A Natural DNMT1 Mutation Elevates the Fetal Hemoglobin Level via Epigenetic Derepression of the γ-Globin Gene in β-Thalassemia. Blood 2021, 137, 1652–1657. [Google Scholar] [CrossRef]
  60. Tan, M.; Luo, H.; Lee, S.; Jin, F.; Yang, J.S.; Montellier, E.; Buchou, T.; Cheng, Z.; Rousseaux, S.; Rajagopal, N.; et al. Identification of 67 Histone Marks and Histone Lysine Crotonylation as a New Type of Histone Modification. Cell 2011, 146, 1016–1028. [Google Scholar] [CrossRef] [Green Version]
  61. Zhou, V.W.; Goren, A.; Bernstein, B.E. Charting Histone Modifications and the Functional Organization of Mammalian Genomes. Nat. Rev. Genet. 2011, 12, 7–18. [Google Scholar] [CrossRef]
  62. Romano, O.; Petiti, L.; Felix, T.; Meneghini, V.; Portafax, M.; Antoniani, C.; Amendola, M.; Bicciato, S.; Peano, C.; Miccio, A. GATA Factor-Mediated Gene Regulation in Human Erythropoiesis. iScience 2020, 23, 101018. [Google Scholar] [CrossRef] [PubMed]
  63. Allfrey, V.G.; Faulkner, R.; Mirsky, A.E. Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis. Proc. Natl. Acad. Sci. USA 1964, 51, 786–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Rank, G.; Cerruti, L.; Simpson, R.J.; Moritz, R.L.; Jane, S.M.; Zhao, Q. Identification of a PRMT5-Dependent Repressor Complex Linked to Silencing of Human Fetal Globin Gene Expression. Blood 2010, 116, 1585–1592. [Google Scholar] [CrossRef]
  65. Chang, K.-H.; Fang, X.; Wang, H.; Huang, A.; Cao, H.; Yang, Y.; Bonig, H.; Stamatoyannopoulos, J.A.; Papayannopoulou, T. Epigenetic Modifications and Chromosome Conformations of the Beta Globin Locus throughout Development. Stem. Cell Rev. Rep. 2013, 9, 397–407. [Google Scholar] [CrossRef] [Green Version]
  66. Xu, J.; Shao, Z.; Glass, K.; Bauer, D.E.; Pinello, L.; Van Handel, B.; Hou, S.; Stamatoyannopoulos, J.A.; Mikkola, H.K.A.; Yuan, G.-C.; et al. Combinatorial Assembly of Developmental Stage-Specific Enhancers Controls Gene Expression Programs during Human Erythropoiesis. Dev. Cell 2012, 23, 796–811. [Google Scholar] [CrossRef] [Green Version]
  67. Hsu, M.; Richardson, C.A.; Olivier, E.; Qiu, C.; Bouhassira, E.E.; Lowrey, C.H.; Fiering, S. Complex Developmental Patterns of Histone Modifications Associated with the Human Beta-Globin Switch in Primary Cells. Exp. Hematol. 2009, 37, 799–806.e4. [Google Scholar] [CrossRef] [Green Version]
  68. Kim, A.; Kiefer, C.M.; Dean, A. Distinctive Signatures of Histone Methylation in Transcribed Coding and Noncoding Human Beta-Globin Sequences. Mol. Cell Biol. 2007, 27, 1271–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Shi, L.; Cui, S.; Engel, J.D.; Tanabe, O. Lysine-Specific Demethylase 1 Is a Therapeutic Target for Fetal Hemoglobin Induction. Nat. Med. 2013, 19, 291–294. [Google Scholar] [CrossRef] [Green Version]
  70. Renneville, A.; Van Galen, P.; Canver, M.C.; McConkey, M.; Krill-Burger, J.M.; Dorfman, D.M.; Holson, E.B.; Bernstein, B.E.; Orkin, S.H.; Bauer, D.E.; et al. EHMT1 and EHMT2 Inhibition Induces Fetal Hemoglobin Expression. Blood 2015, 126, 1930–1939. [Google Scholar] [CrossRef] [Green Version]
  71. Krivega, I.; Byrnes, C.; de Vasconcellos, J.F.; Lee, Y.T.; Kaushal, M.; Dean, A.; Miller, J.L. Inhibition of G9a Methyltransferase Stimulates Fetal Hemoglobin Production by Facilitating LCR/γ-Globin Looping. Blood 2015, 126, 665–672. [Google Scholar] [CrossRef] [Green Version]
  72. Nualkaew, T.; Khamphikham, P.; Pongpaksupasin, P.; Kaewsakulthong, W.; Songdej, D.; Paiboonsukwong, K.; Sripichai, O.; Engel, J.D.; Hongeng, S.; Fucharoen, S.; et al. UNC0638 Induces High Levels of Fetal Hemoglobin Expression in β-Thalassemia/HbE Erythroid Progenitor Cells. Ann. Hematol. 2020, 99, 2027–2036. [Google Scholar] [CrossRef]
  73. Zhao, Q.; Rank, G.; Tan, Y.T.; Li, H.; Moritz, R.L.; Simpson, R.J.; Cerruti, L.; Curtis, D.J.; Patel, D.J.; Allis, C.D.; et al. PRMT5-Mediated Methylation of Histone H4R3 Recruits DNMT3A, Coupling Histone and DNA Methylation in Gene Silencing. Nat. Struct. Mol. Biol. 2009, 16, 304–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ju, J.; Wang, Y.; Liu, R.; Zhang, Y.; Xu, Z.; Wang, Y.; Wu, Y.; Liu, M.; Cerruti, L.; Zou, F.; et al. Human Fetal Globin Gene Expression Is Regulated by LYAR. Nucleic Acids Res. 2014, 42, 9740–9752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Shvedunova, M.; Akhtar, A. Modulation of Cellular Processes by Histone and Non-Histone Protein Acetylation. Nat. Rev. Mol. Cell Biol. 2022, 23, 329–349. [Google Scholar] [CrossRef]
  76. López-Bañuelos, L.; Vega, L. Inhibition of Acetylation, Is It Enough to Fight Cancer? Crit. Rev. Oncol. Hematol. 2022, 176, 103752. [Google Scholar] [CrossRef] [PubMed]
  77. Forsberg, E.C.; Downs, K.M.; Christensen, H.M.; Im, H.; Nuzzi, P.A.; Bresnick, E.H. Developmentally Dynamic Histone Acetylation Pattern of a Tissue-Specific Chromatin Domain. Proc. Natl. Acad. Sci. USA 2000, 97, 14494–14499. [Google Scholar] [CrossRef] [Green Version]
  78. Im, H.; Grass, J.A.; Christensen, H.M.; Perkins, A.; Bresnick, E.H. Histone Deacetylase-Dependent Establishment and Maintenance of Broad Low-Level Histone Acetylation within a Tissue-Specific Chromatin Domain. Biochemistry 2002, 41, 15152–15160. [Google Scholar] [CrossRef]
  79. Schübeler, D.; Francastel, C.; Cimbora, D.M.; Reik, A.; Martin, D.I.; Groudine, M. Nuclear Localization and Histone Acetylation: A Pathway for Chromatin Opening and Transcriptional Activation of the Human Beta-Globin Locus. Genes Dev. 2000, 14, 940–950. [Google Scholar] [CrossRef]
  80. Yin, W.; Barkess, G.; Fang, X.; Xiang, P.; Cao, H.; Stamatoyannopoulos, G.; Li, Q. Histone Acetylation at the Human Beta-Globin Locus Changes with Developmental Age. Blood 2007, 110, 4101–4107. [Google Scholar] [CrossRef] [Green Version]
  81. Dai, Y.; Chen, T.; Ijaz, H.; Cho, E.H.; Steinberg, M.H. SIRT1 Activates the Expression of Fetal Hemoglobin Genes. Am. J. Hematol. 2017, 92, 1177–1186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Kang, Y.; Kim, Y.W.; Kang, J.; Kim, A. Histone H3K4me1 and H3K27ac Play Roles in Nucleosome Eviction and ERNA Transcription, Respectively, at Enhancers. FASEB J. 2021, 35, e21781. [Google Scholar] [CrossRef] [PubMed]
  83. Madzo, J.; Liu, H.; Rodriguez, A.; Vasanthakumar, A.; Sundaravel, S.; Caces, D.B.D.; Looney, T.J.; Zhang, L.; Lepore, J.B.; Macrae, T.; et al. Hydroxymethylation at Gene Regulatory Regions Directs Stem/Early Progenitor Cell Commitment during Erythropoiesis. Cell. Rep. 2014, 6, 231–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. He, Y.; Rank, G.; Zhang, M.; Ju, J.; Liu, R.; Xu, Z.; Brown, F.; Cerruti, L.; Ma, C.; Tan, R.; et al. Induction of Human Fetal Hemoglobin Expression by Adenosine-2’,3’-Dialdehyde. J. Transl. Med. 2013, 11, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Blobel, G.A.; Nakajima, T.; Eckner, R.; Montminy, M.; Orkin, S.H. CREB-Binding Protein Cooperates with Transcription Factor GATA-1 and Is Required for Erythroid Differentiation. Proc. Natl. Acad. Sci. USA 1998, 95, 2061–2066. [Google Scholar] [CrossRef] [Green Version]
  86. Desai, M.A.; Webb, H.D.; Sinanan, L.M.; Scarsdale, J.N.; Walavalkar, N.M.; Ginder, G.D.; Williams, D.C. An Intrinsically Disordered Region of Methyl-CpG Binding Domain Protein 2 (MBD2) Recruits the Histone Deacetylase Core of the NuRD Complex. Nucleic Acids Res. 2015, 43, 3100–3113. [Google Scholar] [CrossRef]
  87. Cui, S.; Kolodziej, K.E.; Obara, N.; Amaral-Psarris, A.; Demmers, J.; Shi, L.; Engel, J.D.; Grosveld, F.; Strouboulis, J.; Tanabe, O. Nuclear Receptors TR2 and TR4 Recruit Multiple Epigenetic Transcriptional Corepressors That Associate Specifically with the Embryonic β-Type Globin Promoters in Differentiated Adult Erythroid Cells▿. Mol. Cell Biol. 2011, 31, 3298–3311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Deng, W.; Lee, J.; Wang, H.; Miller, J.; Reik, A.; Gregory, P.D.; Dean, A.; Blobel, G.A. Controlling Long-Range Genomic Interactions at a Native Locus by Targeted Tethering of a Looping Factor. Cell 2012, 149, 1233–1244. [Google Scholar] [CrossRef] [Green Version]
  89. Guo, X.; Plank-Bazinet, J.; Krivega, I.; Dale, R.K.; Dean, A. Embryonic Erythropoiesis and Hemoglobin Switching Require Transcriptional Repressor ETO2 to Modulate Chromatin Organization. Nucleic Acids Res. 2020, 48, 10226–10240. [Google Scholar] [CrossRef]
  90. Saunthararajah, Y.; Hillery, C.A.; Lavelle, D.; Molokie, R.; Dorn, L.; Bressler, L.; Gavazova, S.; Chen, Y.-H.; Hoffman, R.; DeSimone, J. Effects of 5-Aza-2′-Deoxycytidine on Fetal Hemoglobin Levels, Red Cell Adhesion, and Hematopoietic Differentiation in Patients with Sickle Cell Disease. Blood 2003, 102, 3865–3870. [Google Scholar] [CrossRef]
  91. Park, J.W.; Han, J.-W. Targeting Epigenetics for Cancer Therapy. Arch. Pharm. Res. 2019, 42, 159–170. [Google Scholar] [CrossRef] [Green Version]
  92. Grimmer, M.R.; Stolzenburg, S.; Ford, E.; Lister, R.; Blancafort, P.; Farnham, P.J. Analysis of an Artificial Zinc Finger Epigenetic Modulator: Widespread Binding but Limited Regulation. Nucleic Acids Res. 2014, 42, 10856–10868. [Google Scholar] [CrossRef]
  93. Durai, S.; Mani, M.; Kandavelou, K.; Wu, J.; Porteus, M.H.; Chandrasegaran, S. Zinc Finger Nucleases: Custom-Designed Molecular Scissors for Genome Engineering of Plant and Mammalian Cells. Nucleic Acids Res. 2005, 33, 5978–5990. [Google Scholar] [CrossRef]
  94. Moore, M.; Klug, A.; Choo, Y. Improved DNA Binding Specificity from Polyzinc Finger Peptides by Using Strings of Two-Finger Units. Proc. Natl. Acad. Sci. USA 2001, 98, 1437–1441. [Google Scholar] [CrossRef] [Green Version]
  95. Segal, D.J.; Barbas, C.F. Custom DNA-Binding Proteins Come of Age: Polydactyl Zinc-Finger Proteins. Curr. Opin. Biotechnol. 2001, 12, 632–637. [Google Scholar] [CrossRef] [PubMed]
  96. Zhang, L.; Spratt, S.K.; Liu, Q.; Johnstone, B.; Qi, H.; Raschke, E.E.; Jamieson, A.C.; Rebar, E.J.; Wolffe, A.P.; Case, C.C. Synthetic Zinc Finger Transcription Factor Action at an Endogenous Chromosomal Site. J. Biol. Chem. 2000, 275, 33850–33860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Liu, P.-Q.; Rebar, E.J.; Zhang, L.; Liu, Q.; Jamieson, A.C.; Liang, Y.; Qi, H.; Li, P.-X.; Chen, B.; Mendel, M.C.; et al. Regulation of an Endogenous Locus Using a Panel of Designed Zinc Finger Proteins Targeted to Accessible Chromatin Regions. J. Biol. Chem. 2001, 276, 11323–11334. [Google Scholar] [CrossRef] [Green Version]
  98. Wilber, A.; Tschulena, U.; Hargrove, P.W.; Kim, Y.-S.; Persons, D.A.; Barbas, C.F.; Nienhuis, A.W. A Zinc-Finger Transcriptional Activator Designed to Interact with the γ-Globin Gene Promoters Enhances Fetal Hemoglobin Production in Primary Human Adult Erythroblasts. Blood 2010, 115, 3033–3041. [Google Scholar] [CrossRef] [Green Version]
  99. Huisman, C.; van der Wijst, M.G.; Falahi, F.; Overkamp, J.; Karsten, G.; Terpstra, M.M.; Kok, K.; van der Zee, A.G.; Schuuring, E.; Wisman, G.B.A.; et al. Prolonged Re-Expression of the Hypermethylated Gene EPB41L3 Using Artificial Transcription Factors and Epigenetic Drugs. Epigenetics 2015, 10, 384–396. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Huisman, C.; van der Wijst, M.G.P.; Schokker, M.; Blancafort, P.; Terpstra, M.M.; Kok, K.; van der Zee, A.G.J.; Schuuring, E.; Wisman, G.B.A.; Rots, M.G. Re-Expression of Selected Epigenetically Silenced Candidate Tumor Suppressor Genes in Cervical Cancer by TET2-Directed Demethylation. Mol. Ther. 2016, 24, 536–547. [Google Scholar] [CrossRef] [Green Version]
  101. Maeder, M.L.; Angstman, J.F.; Richardson, M.E.; Linder, S.J.; Cascio, V.M.; Tsai, S.Q.; Ho, Q.H.; Sander, J.D.; Reyon, D.; Bernstein, B.E.; et al. Targeted DNA Demethylation and Activation of Endogenous Genes Using Programmable TALE-TET1 Fusion Proteins. Nat. Biotechnol. 2013, 31, 1137–1142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Siddique, A.N.; Nunna, S.; Rajavelu, A.; Zhang, Y.; Jurkowska, R.Z.; Reinhardt, R.; Rots, M.G.; Ragozin, S.; Jurkowski, T.P.; Jeltsch, A. Targeted Methylation and Gene Silencing of VEGF-A in Human Cells by Using a Designed Dnmt3a–Dnmt3L Single-Chain Fusion Protein with Increased DNA Methylation Activity. J. Mol. Biol. 2013, 425, 479–491. [Google Scholar] [CrossRef]
  103. Hilton, I.B.; D’Ippolito, A.M.; Vockley, C.M.; Thakore, P.I.; Crawford, G.E.; Reddy, T.E.; Gersbach, C.A. Epigenome Editing by a CRISPR-Cas9-Based Acetyltransferase Activates Genes from Promoters and Enhancers. Nat. Biotechnol. 2015, 33, 510–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Gregory, D.J.; Zhang, Y.; Kobzik, L.; Fedulov, A.V. Specific Transcriptional Enhancement of Inducible Nitric Oxide Synthase by Targeted Promoter Demethylation. Epigenetics 2013, 8, 1205–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Juárez-Moreno, K.; Erices, R.; Beltran, A.S.; Stolzenburg, S.; Cuello-Fredes, M.; Owen, G.I.; Qian, H.; Blancafort, P. Breaking through an Epigenetic Wall: Re-Activation of Oct4 by KRAB-Containing Designer Zinc Finger Transcription Factors. Epigenetics 2013, 8, 164–176. [Google Scholar] [CrossRef] [Green Version]
  106. Mussolino, C.; Cathomen, T. On Target? Tracing Zinc-Finger-Nuclease Specificity. Nat. Methods 2011, 8, 725–726. [Google Scholar] [CrossRef]
  107. Chaikind, B.; Kilambi, K.P.; Gray, J.J.; Ostermeier, M. Targeted DNA Methylation Using an Artificially Bisected M.HhaI Fused to Zinc Fingers. PLoS ONE 2012, 7, e44852. [Google Scholar] [CrossRef] [Green Version]
  108. Joung, J.K.; Sander, J.D. TALENs: A Widely Applicable Technology for Targeted Genome Editing. Nat Rev Mol Cell Biol 2013, 14, 49–55. [Google Scholar] [CrossRef] [Green Version]
  109. Moscou, M.J.; Bogdanove, A.J. A Simple Cipher Governs DNA Recognition by TAL Effectors. Science 2009, 326, 1501. [Google Scholar] [CrossRef]
  110. Jankele, R.; Svoboda, P. TAL Effectors: Tools for DNA Targeting. Brief Funct. Genom. 2014, 13, 409–419. [Google Scholar] [CrossRef]
  111. Boch, J.; Scholze, H.; Schornack, S.; Landgraf, A.; Hahn, S.; Kay, S.; Lahaye, T.; Nickstadt, A.; Bonas, U. Breaking the Code of DNA Binding Specificity of TAL-Type III Effectors. Science 2009, 326, 1509–1512. [Google Scholar] [CrossRef]
  112. Rogers, J.M.; Barrera, L.A.; Reyon, D.; Sander, J.D.; Kellis, M.; Keith Joung, J.; Bulyk, M.L. Context Influences on TALE–DNA Binding Revealed by Quantitative Profiling. Nat. Commun. 2015, 6, 7440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Juillerat, A.; Dubois, G.; Valton, J.; Thomas, S.; Stella, S.; Maréchal, A.; Langevin, S.; Benomari, N.; Bertonati, C.; Silva, G.H.; et al. Comprehensive Analysis of the Specificity of Transcription Activator-like Effector Nucleases. Nucleic Acids Res. 2014, 42, 5390–5402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Zhang, F.; Cong, L.; Lodato, S.; Kosuri, S.; Church, G.M.; Arlotta, P. Efficient Construction of Sequence-Specific TAL Effectors for Modulating Mammalian Transcription. Nat. Biotechnol. 2011, 29, 149–153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gao, X.; Yang, J.; Tsang, J.C.H.; Ooi, J.; Wu, D.; Liu, P. Reprogramming to Pluripotency Using Designer TALE Transcription Factors Targeting Enhancers. Stem Cell Rep. 2013, 1, 183–197. [Google Scholar] [CrossRef] [Green Version]
  116. Hu, J.; Lei, Y.; Wong, W.-K.; Liu, S.; Lee, K.-C.; He, X.; You, W.; Zhou, R.; Guo, J.-T.; Chen, X.; et al. Direct Activation of Human and Mouse Oct4 Genes Using Engineered TALE and Cas9 Transcription Factors. Nucleic Acids Res. 2014, 42, 4375–4390. [Google Scholar] [CrossRef]
  117. Groner, A.C.; Meylan, S.; Ciuffi, A.; Zangger, N.; Ambrosini, G.; Dénervaud, N.; Bucher, P.; Trono, D. KRAB–Zinc Finger Proteins and KAP1 Can Mediate Long-Range Transcriptional Repression through Heterochromatin Spreading. PLoS Genet. 2010, 6, e1000869. [Google Scholar] [CrossRef] [Green Version]
  118. Cong, L.; Zhou, R.; Kuo, Y.; Cunniff, M.; Zhang, F. Comprehensive Interrogation of Natural TALE DNA-Binding Modules and Transcriptional Repressor Domains. Nat. Commun. 2012, 3, 968. [Google Scholar] [CrossRef] [Green Version]
  119. Zhang, Z.; Wu, E.; Qian, Z.; Wu, W.-S. A Multicolor Panel of TALE-KRAB Based Transcriptional Repressor Vectors Enabling Knockdown of Multiple Gene Targets. Sci. Rep. 2015, 4, 7338. [Google Scholar] [CrossRef] [Green Version]
  120. Masuda, J.; Kawamoto, H.; Strober, W.; Takayama, E.; Mizutani, A.; Murakami, H.; Ikawa, T.; Kitani, A.; Maeno, N.; Shigehiro, T.; et al. Transient Tcf3 Gene Repression by TALE-Transcription Factor Targeting. Appl. Biochem. Biotechnol. 2016, 180, 1559–1573. [Google Scholar] [CrossRef]
  121. Konermann, S.; Brigham, M.D.; Trevino, A.E.; Hsu, P.D.; Heidenreich, M.; Le, C.; Platt, R.J.; Scott, D.A.; Church, G.M.; Zhang, F. Optical Control of Mammalian Endogenous Transcription and Epigenetic States. Nature 2013, 500, 472–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Ou, K.; Yu, M.; Moss, N.G.; Wang, Y.J.; Wang, A.W.; Nguyen, S.C.; Jiang, C.; Feleke, E.; Kameswaran, V.; Joyce, E.F.; et al. Targeted Demethylation at the CDKN1C/P57 Locus Induces Human β Cell Replication. J. Clin. Investig. 2018, 129, 209–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Bernstein, D.L.; Le Lay, J.E.; Ruano, E.G.; Kaestner, K.H. TALE-Mediated Epigenetic Suppression of CDKN2A Increases Replication in Human Fibroblasts. J. Clin. Invest. 2015, 125, 1998–2006. [Google Scholar] [CrossRef] [PubMed]
  124. Mendenhall, E.M.; Williamson, K.E.; Reyon, D.; Zou, J.Y.; Ram, O.; Joung, J.K.; Bernstein, B.E. Locus-Specific Editing of Histone Modifications at Endogenous Enhancers. Nat. Biotechnol. 2013, 31, 1133–1136. [Google Scholar] [CrossRef] [Green Version]
  125. Amabile, A.; Migliara, A.; Capasso, P.; Biffi, M.; Cittaro, D.; Naldini, L.; Lombardo, A. Inheritable Silencing of Endogenous Genes by Hit-and-Run Targeted Epigenetic Editing. Cell 2016, 167, 219–232.e14. [Google Scholar] [CrossRef] [Green Version]
  126. Mlambo, T.; Nitsch, S.; Hildenbeutel, M.; Romito, M.; Müller, M.; Bossen, C.; Diederichs, S.; Cornu, T.I.; Cathomen, T.; Mussolino, C. Designer Epigenome Modifiers Enable Robust and Sustained Gene Silencing in Clinically Relevant Human Cells. Nucleic Acids Res. 2018, 46, 4456–4468. [Google Scholar] [CrossRef]
  127. Tesson, L.; Usal, C.; Ménoret, S.; Leung, E.; Niles, B.J.; Remy, S.; Santiago, Y.; Vincent, A.I.; Meng, X.; Zhang, L.; et al. Knockout Rats Generated by Embryo Microinjection of TALENs. Nat. Biotechnol. 2011, 29, 695–696. [Google Scholar] [CrossRef]
  128. Guilinger, J.P.; Pattanayak, V.; Reyon, D.; Tsai, S.Q.; Sander, J.D.; Joung, J.K.; Liu, D.R. Broad Specificity Profiling of TALENs Results in Engineered Nucleases with Improved DNA-Cleavage Specificity. Nat. Methods 2014, 11, 429–435. [Google Scholar] [CrossRef] [Green Version]
  129. Mali, P.; Aach, J.; Stranges, P.B.; Esvelt, K.M.; Moosburner, M.; Kosuri, S.; Yang, L.; Church, G.M. CAS9 Transcriptional Activators for Target Specificity Screening and Paired Nickases for Cooperative Genome Engineering. Nat. Biotechnol. 2013, 31, 833–838. [Google Scholar] [CrossRef]
  130. Polstein, L.R.; Perez-Pinera, P.; Kocak, D.D.; Vockley, C.M.; Bledsoe, P.; Song, L.; Safi, A.; Crawford, G.E.; Reddy, T.E.; Gersbach, C.A. Genome-Wide Specificity of DNA Binding, Gene Regulation, and Chromatin Remodeling by TALE- and CRISPR/Cas9-Based Transcriptional Activators. Genome Res. 2015, 25, 1158–1169. [Google Scholar] [CrossRef] [Green Version]
  131. Cuculis, L.; Zhao, C.; Abil, Z.; Zhao, H.; Shukla, D.; Schroeder, C.M. Divalent Cations Promote TALE DNA-Binding Specificity. Nucleic Acids Res. 2020, 48, 1406–1422. [Google Scholar] [CrossRef]
  132. Gilbert, L.A.; Larson, M.H.; Morsut, L.; Liu, Z.; Brar, G.A.; Torres, S.E.; Stern-Ginossar, N.; Brandman, O.; Whitehead, E.H.; Doudna, J.A.; et al. CRISPR-Mediated Modular RNA-Guided Regulation of Transcription in Eukaryotes. Cell 2013, 154, 442–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Maeder, M.L.; Linder, S.J.; Cascio, V.M.; Fu, Y.; Ho, Q.H.; Joung, J.K. CRISPR RNA–Guided Activation of Endogenous Human Genes. Nat. Methods 2013, 10, 977–979. [Google Scholar] [CrossRef] [Green Version]
  134. Moreno, A.M.; Fu, X.; Zhu, J.; Katrekar, D.; Shih, Y.-R.V.; Marlett, J.; Cabotaje, J.; Tat, J.; Naughton, J.; Lisowski, L.; et al. In Situ Gene Therapy via AAV-CRISPR-Cas9-Mediated Targeted Gene Regulation. Mol. Ther. 2018, 26, 1818–1827. [Google Scholar] [CrossRef] [Green Version]
  135. Moses, C.; Hodgetts, S.I.; Nugent, F.; Ben-Ary, G.; Park, K.K.; Blancafort, P.; Harvey, A.R. Transcriptional Repression of PTEN in Neural Cells Using CRISPR/DCas9 Epigenetic Editing. Sci. Rep. 2020, 10, 11393. [Google Scholar] [CrossRef]
  136. Perez-Pinera, P.; Ousterout, D.G.; Brunger, J.M.; Farin, A.M.; Glass, K.A.; Guilak, F.; Crawford, G.E.; Hartemink, A.J.; Gersbach, C.A. Synergistic and Tunable Human Gene Activation by Combinations of Synthetic Transcription Factors. Nat. Methods 2013, 10, 239–242. [Google Scholar] [CrossRef]
  137. Gao, X.; Tsang, J.C.H.; Gaba, F.; Wu, D.; Lu, L.; Liu, P. Comparison of TALE Designer Transcription Factors and the CRISPR/DCas9 in Regulation of Gene Expression by Targeting Enhancers. Nucleic Acids Res. 2014, 42, e155. [Google Scholar] [CrossRef] [Green Version]
  138. Cheng, A.W.; Wang, H.; Yang, H.; Shi, L.; Katz, Y.; Theunissen, T.W.; Rangarajan, S.; Shivalila, C.S.; Dadon, D.B.; Jaenisch, R. Multiplexed Activation of Endogenous Genes by CRISPR-on, an RNA-Guided Transcriptional Activator System. Cell Res. 2013, 23, 1163–1171. [Google Scholar] [CrossRef] [PubMed]
  139. Chavez, A.; Scheiman, J.; Vora, S.; Pruitt, B.W.; Tuttle, M.; Iyer, E.P.R.; Lin, S.; Kiani, S.; Guzman, C.D.; Wiegand, D.J.; et al. Highly Efficient Cas9-Mediated Transcriptional Programming. Nat. Methods 2015, 12, 326–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Zhang, X.; Lv, S.; Luo, Z.; Hu, Y.; Peng, X.; Lv, J.; Zhao, S.; Feng, J.; Huang, G.; Wan, Q.-L.; et al. MiniCAFE, a CRISPR/Cas9-Based Compact and Potent Transcriptional Activator, Elicits Gene Expression in Vivo. Nucleic Acids Res. 2021, 49, 4171–4185. [Google Scholar] [CrossRef]
  141. Kantor, B.; Tagliafierro, L.; Gu, J.; Zamora, M.E.; Ilich, E.; Grenier, C.; Huang, Z.Y.; Murphy, S.; Chiba-Falek, O. Downregulation of SNCA Expression by Targeted Editing of DNA Methylation: A Potential Strategy for Precision Therapy in PD. Mol. Ther. 2018, 26, 2638–2649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Stepper, P.; Kungulovski, G.; Jurkowska, R.Z.; Chandra, T.; Krueger, F.; Reinhardt, R.; Reik, W.; Jeltsch, A.; Jurkowski, T.P. Efficient Targeted DNA Methylation with Chimeric DCas9–Dnmt3a–Dnmt3L Methyltransferase. Nucleic Acids Res. 2017, 45, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
  143. Nuñez, J.K.; Chen, J.; Pommier, G.C.; Cogan, J.Z.; Replogle, J.M.; Adriaens, C.; Ramadoss, G.N.; Shi, Q.; Hung, K.L.; Samelson, A.J.; et al. Genome-Wide Programmable Transcriptional Memory by CRISPR-Based Epigenome Editing. Cell 2021, 184, 2503–2519.e17. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, X.S.; Wu, H.; Ji, X.; Stelzer, Y.; Wu, X.; Czauderna, S.; Shu, J.; Dadon, D.; Young, R.A.; Jaenisch, R. Editing DNA Methylation in the Mammalian Genome. Cell 2016, 167, 233–247.e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, X.S.; Wu, H.; Krzisch, M.; Wu, X.; Graef, J.; Muffat, J.; Hnisz, D.; Li, C.H.; Yuan, B.; Xu, C.; et al. Rescue of Fragile X Syndrome Neurons by DNA Methylation Editing of the FMR1 Gene. Cell 2018, 172, 979–992.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Choudhury, S.R.; Cui, Y.; Lubecka, K.; Stefanska, B.; Irudayaraj, J. CRISPR-DCas9 Mediated TET1 Targeting for Selective DNA Demethylation at BRCA1 Promoter. Oncotarget 2016, 7, 46545–46556. [Google Scholar] [CrossRef] [Green Version]
  147. Wilk, C.; Effenberg, L.; Abberger, H.; Steenpass, L.; Hansen, W.; Zeschnigk, M.; Kirschning, C.; Buer, J.; Kehrmann, J. CRISPR/Cas9-Mediated Demethylation of FOXP3-TSDR toward Treg-Characteristic Programming of Jurkat T Cells. Cell. Immunol. 2022, 371, 104471. [Google Scholar] [CrossRef]
  148. Okada, M.; Kanamori, M.; Someya, K.; Nakatsukasa, H.; Yoshimura, A. Stabilization of Foxp3 Expression by CRISPR-DCas9-Based Epigenome Editing in Mouse Primary T Cells. Epigenetics Chromatin. 2017, 10, 24. [Google Scholar] [CrossRef] [Green Version]
  149. Fang, S.; Cui, D.; Hong, T.; Guo, L.; Lee, Y.-T.; Lee, M.; Isgandarova, S.; Martinez-Moczygemba, M.; Zhou, Y.; Li, J.; et al. Ten-Eleven Translocation Ablation Impairs Cardiac Differentiation of Mouse Embryonic Stem Cells. Stem. Cells 2022, 40, 260–272. [Google Scholar] [CrossRef]
  150. Kabadi, A.M.; Machlin, L.; Dalal, N.; Lee, R.E.; McDowell, I.; Shah, N.N.; Drowley, L.; Randell, S.H.; Reddy, T.E. Epigenome Editing of the CFTR-Locus for Treatment of Cystic Fibrosis. J. Cyst. Fibros. 2022, 21, 164–171. [Google Scholar] [CrossRef]
  151. Kwon, D.Y.; Zhao, Y.-T.; Lamonica, J.M.; Zhou, Z. Locus-Specific Histone Deacetylation Using a Synthetic CRISPR-Cas9-Based HDAC. Nat. Commun. 2017, 8, 15315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Kearns, N.A.; Pham, H.; Tabak, B.; Genga, R.M.; Silverstein, N.J.; Garber, M.; Maehr, R. Functional Annotation of Native Enhancers with a Cas9–Histone Demethylase Fusion. Nat. Methods 2015, 12, 401–403. [Google Scholar] [CrossRef] [Green Version]
  153. Williams, R.M.; Senanayake, U.; Artibani, M.; Taylor, G.; Wells, D.; Ahmed, A.A.; Sauka-Spengler, T. Genome and Epigenome Engineering CRISPR Toolkit for in Vivo Modulation of Cis -Regulatory Interactions and Gene Expression in the Chicken Embryo. Development 2018, 145, dev.160333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. O’Geen, H.; Ren, C.; Nicolet, C.M.; Perez, A.A.; Halmai, J.; Le, V.M.; Mackay, J.P.; Farnham, P.J.; Segal, D.J. DCas9-Based Epigenome Editing Suggests Acquisition of Histone Methylation Is Not Sufficient for Target Gene Repression. Nucleic Acids Res. 2017, 45, 9901–9916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. O’Geen, H.; Tomkova, M.; Combs, J.A.; Tilley, E.K.; Segal, D.J. Determinants of Heritable Gene Silencing for KRAB-DCas9 + DNMT3 and Ezh2-DCas9 + DNMT3 Hit-and-Run Epigenome Editing. Nucleic Acids Res. 2022, 50, 3239–3253. [Google Scholar] [CrossRef] [PubMed]
  156. Pattanayak, V.; Lin, S.; Guilinger, J.P.; Ma, E.; Doudna, J.A.; Liu, D.R. High-Throughput Profiling of off-Target DNA Cleavage Reveals RNA-Programmed Cas9 Nuclease Specificity. Nat. Biotechnol. 2013, 31, 839–843. [Google Scholar] [CrossRef]
  157. Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-Fidelity CRISPR–Cas9 Nucleases with No Detectable Genome-Wide off-Target Effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Liu, J.-J.; Orlova, N.; Oakes, B.L.; Ma, E.; Spinner, H.B.; Baney, K.L.M.; Chuck, J.; Tan, D.; Knott, G.J.; Harrington, L.B.; et al. CasX Enzymes Comprise a Distinct Family of RNA-Guided Genome Editors. Nature 2019, 566, 218–223. [Google Scholar] [CrossRef]
  159. Kiani, S.; Chavez, A.; Tuttle, M.; Hall, R.N.; Chari, R.; Ter-Ovanesyan, D.; Qian, J.; Pruitt, B.W.; Beal, J.; Vora, S.; et al. Cas9 GRNA Engineering for Genome Editing, Activation and Repression. Nat. Methods 2015, 12, 1051–1054. [Google Scholar] [CrossRef] [Green Version]
  160. Liu, J.; Sun, M.; Cho, K.B.; Gao, X.; Guo, B. A CRISPR-Cas9 Repressor for Epigenetic Silencing of KRAS. Pharmacol. Res. 2021, 164, 105304. [Google Scholar] [CrossRef]
  161. Zheng, Y.; Shen, W.; Zhang, J.; Yang, B.; Liu, Y.-N.; Qi, H.; Yu, X.; Lu, S.-Y.; Chen, Y.; Xu, Y.-Z.; et al. CRISPR Interference-Based Specific and Efficient Gene Inactivation in the Brain. Nat. Neurosci. 2018, 21, 447–454. [Google Scholar] [CrossRef]
  162. Mussolino, C. Precise Epigenome Editing on the Stage: A Novel Approach to Modulate Gene Expression. Epigenet. Insights 2018, 11, 2516865718818838. [Google Scholar] [CrossRef] [Green Version]
  163. Kiefer, C.M.; Hou, C.; Little, J.A.; Dean, A. Epigenetics of Beta-Globin Gene Regulation. Mutat. Res. 2008, 647, 68–76. [Google Scholar] [CrossRef] [Green Version]
  164. Kalantri, S.A.; Ray, R.; Chattopadhyay, A.; Bhattacharjee, S.; Biswas, A.; Bhattacharyya, M. Efficacy of Decitabine as Hemoglobin F Inducer in HbE/β-Thalassemia. Ann. Hematol. 2018, 97, 1689–1694. [Google Scholar] [CrossRef]
  165. Molokie, R.; Lavelle, D.; Gowhari, M.; Pacini, M.; Krauz, L.; Hassan, J.; Ibanez, V.; Ruiz, M.A.; Ng, K.P.; Woost, P.; et al. Oral Tetrahydrouridine and Decitabine for Non-Cytotoxic Epigenetic Gene Regulation in Sickle Cell Disease: A Randomized Phase 1 Study. PLoS Med. 2017, 14, e1002382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Gilmartin, A.G.; Groy, A.; Gore, E.R.; Atkins, C.; Long, E.R.; Montoute, M.N.; Wu, Z.; Halsey, W.; McNulty, D.E.; Ennulat, D.; et al. In Vitro and in Vivo Induction of Fetal Hemoglobin with a Reversible and Selective DNMT1 Inhibitor. Haematologica 2021, 106, 1979–1987. [Google Scholar] [CrossRef] [PubMed]
  167. Okam, M.M.; Esrick, E.B.; Mandell, E.; Campigotto, F.; Neuberg, D.S.; Ebert, B.L. Phase 1/2 Trial of Vorinostat in Patients with Sickle Cell Disease Who Have Not Benefitted from Hydroxyurea. Blood 2015, 125, 3668–3669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Junker, L.H.; Li, B.; Zhu, X.; Koti, S.; Cerbone, R.E.; Hendrick, C.L.; Sangerman, J.; Perrine, S.; Pace, B.S. Novel Histone Deacetylase Inhibitor CT-101 Induces γ-Globin Gene Expression in Sickle Erythroid Progenitors with Targeted Epigenetic Effects. Blood Cells Mol. Dis. 2022, 93, 102626. [Google Scholar] [CrossRef]
  169. Cui, S.; Lim, K.-C.; Shi, L.; Lee, M.; Jearawiriyapaisarn, N.; Myers, G.; Campbell, A.; Harro, D.; Iwase, S.; Trievel, R.C.; et al. The LSD1 Inhibitor RN-1 Induces Fetal Hemoglobin Synthesis and Reduces Disease Pathology in Sickle Cell Mice. Blood 2015, 126, 386–396. [Google Scholar] [CrossRef]
  170. Rivers, A.; Vaitkus, K.; Ruiz, M.A.; Ibanez, V.; Jagadeeswaran, R.; Kouznetsova, T.; DeSimone, J.; Lavelle, D. RN-1, a Potent and Selective Lysine-Specific Demethylase 1 Inhibitor, Increases γ-Globin Expression, F Reticulocytes, and F Cells in a Sickle Cell Disease Mouse Model. Exp. Hematol. 2015, 43, 546–553.e1–3. [Google Scholar] [CrossRef] [PubMed]
  171. Rivers, A.; Vaitkus, K.; Ibanez, V.; Ruiz, M.A.; Jagadeeswaran, R.; Saunthararajah, Y.; Cui, S.; Engel, J.D.; DeSimone, J.; Lavelle, D. The LSD1 Inhibitor RN-1 Recapitulates the Fetal Pattern of Hemoglobin Synthesis in Baboons (P. Anubis). Haematologica 2016, 101, 688–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Matson, D.; Xie, K.; Roth, M.; Stuart, B.; Bruno, P.; Efremov, I.; Thompson, L.; Silver, S.; Moxham, C. Ftx-6058 Induces Fetal Hemoglobin Production and Ameliorates Disease Pathology in Sickle Cell Mice. Blood 2021, 138, 2018. [Google Scholar] [CrossRef]
  173. Sher, F.; Hossain, M.; Seruggia, D.; Schoonenberg, V.A.C.; Yao, Q.; Cifani, P.; Dassama, L.M.K.; Cole, M.A.; Ren, C.; Vinjamur, D.S.; et al. Rational Targeting of a NuRD Subcomplex Guided by Comprehensive in Situ Mutagenesis. Nat. Genet. 2019, 51, 1149–1159. [Google Scholar] [CrossRef]
  174. Ginder, G.D. Epigenetic Regulation of Fetal Globin Gene Expression in Adult Erythroid Cells. Transl. Res. 2015, 165, 115–125. [Google Scholar] [CrossRef] [Green Version]
  175. Amendola, M.; Brusson, M.; Miccio, A. CRISPRthripsis: The Risk of CRISPR/Cas9-Induced Chromothripsis in Gene Therapy. Stem Cells Transl. Med. 2022, 11, 1003–1009. [Google Scholar] [CrossRef]
  176. Costa, F.C.; Fedosyuk, H.; Neades, R.; de Los Rios, J.B.; Barbas, C.F.; Peterson, K.R. Induction of Fetal Hemoglobin In Vivo Mediated by a Synthetic γ -Globin Zinc Finger Activator. Anemia 2012, 2012, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Thakore, P.I.; D’Ippolito, A.M.; Song, L.; Safi, A.; Shivakumar, N.K.; Kabadi, A.M.; Reddy, T.E.; Crawford, G.E.; Gersbach, C.A. Highly Specific Epigenome Editing by CRISPR-Cas9 Repressors for Silencing of Distal Regulatory Elements. Nat. Methods 2015, 12, 1143–1149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Li, K.; Liu, Y.; Cao, H.; Zhang, Y.; Gu, Z.; Liu, X.; Yu, A.; Kaphle, P.; Dickerson, K.E.; Ni, M.; et al. Interrogation of Enhancer Function by Enhancer-Targeting CRISPR Epigenetic Editing. Nat. Commun. 2020, 11, 485. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Human α- and β-globin loci and hemoglobin switching. (A) Schematic representation of the α- and β-globin loci on chromosomes 16 and 11, respectively. α-like (HBZ, HBA2 and HBA1) and β-like (HBE1, HBG2, HBG1, HBD and HBB) globin genes are depicted with colored boxes in 5′ to 3′ orientation. The globin encoded by each gene is reported below each box. HBBP1 pseudogene is depicted with a white box. The HSs are presented with grey boxes. (B) Diagrammatic representation of the human hemoglobin embryonic-to-fetal and fetal-to-adult switching, describing the expression pattern of α-like (α and ζ) and β-like (ε, γ, β and δ) globin chains, from conception to birth, and to adult life. (C) Composition of hemoglobin for different developmental stages (embryonic, fetal and adult Hb). Chromosome (Chr); multispecies conserves sequences (MCS); locus control region (LCR); hypersensitive site (HS); hemoglobin (Hb); fetal hemoglobin (HbF); adult hemoglobin (HbA); adult hemoglobin 2 (HbA2). Created with BioRender.com.
Figure 1. Human α- and β-globin loci and hemoglobin switching. (A) Schematic representation of the α- and β-globin loci on chromosomes 16 and 11, respectively. α-like (HBZ, HBA2 and HBA1) and β-like (HBE1, HBG2, HBG1, HBD and HBB) globin genes are depicted with colored boxes in 5′ to 3′ orientation. The globin encoded by each gene is reported below each box. HBBP1 pseudogene is depicted with a white box. The HSs are presented with grey boxes. (B) Diagrammatic representation of the human hemoglobin embryonic-to-fetal and fetal-to-adult switching, describing the expression pattern of α-like (α and ζ) and β-like (ε, γ, β and δ) globin chains, from conception to birth, and to adult life. (C) Composition of hemoglobin for different developmental stages (embryonic, fetal and adult Hb). Chromosome (Chr); multispecies conserves sequences (MCS); locus control region (LCR); hypersensitive site (HS); hemoglobin (Hb); fetal hemoglobin (HbF); adult hemoglobin (HbA); adult hemoglobin 2 (HbA2). Created with BioRender.com.
Genes 14 00577 g001
Figure 2. Regulation of β-like globin gene expression. Schematic model of β-like globin gene regulation in the adult (upper panel) and fetal stages, in individuals with hereditary persistence of fetal hemoglobin (HPFH) mutations, or in hemoglobinopathy patients, or in low O2 conditions (lower panel). Numerous transcription factors have been identified to regulate γ-globin expression in fetal/adult stages or HPFH conditions, either by directly silencing (NFIA/X [15], ERF [16]) or activating (Coup-TFII [17], IGF2BP1 [18], HIF1α/β [19], NF-Y [20,21], GATA1 [20] and KLF1 [22]) γ-globin genes, or by co-acting with or indirectly regulating the expression of the two major γ-globin repressors BCL11A (KLF1 [23], NFIA/X [15], ATF4 [24,25], SOX6 [26]) and LRF (KLF1 [27]) or other key-players such as KLF1 (MYB [28]), ATF4 (HRI [24,25]), MYB (ATF4 [29]) and CHD4 (ZNF410 [30,31]). Ovals represent proteins with repressive (red), activating (green), repressive/activating (yellow) activity, or epigenetic modifying activity (blue and purple). Dotted green lines show indirect activation and dotted black ovals indicate protein heterodimers. Created with BioRender.com.
Figure 2. Regulation of β-like globin gene expression. Schematic model of β-like globin gene regulation in the adult (upper panel) and fetal stages, in individuals with hereditary persistence of fetal hemoglobin (HPFH) mutations, or in hemoglobinopathy patients, or in low O2 conditions (lower panel). Numerous transcription factors have been identified to regulate γ-globin expression in fetal/adult stages or HPFH conditions, either by directly silencing (NFIA/X [15], ERF [16]) or activating (Coup-TFII [17], IGF2BP1 [18], HIF1α/β [19], NF-Y [20,21], GATA1 [20] and KLF1 [22]) γ-globin genes, or by co-acting with or indirectly regulating the expression of the two major γ-globin repressors BCL11A (KLF1 [23], NFIA/X [15], ATF4 [24,25], SOX6 [26]) and LRF (KLF1 [27]) or other key-players such as KLF1 (MYB [28]), ATF4 (HRI [24,25]), MYB (ATF4 [29]) and CHD4 (ZNF410 [30,31]). Ovals represent proteins with repressive (red), activating (green), repressive/activating (yellow) activity, or epigenetic modifying activity (blue and purple). Dotted green lines show indirect activation and dotted black ovals indicate protein heterodimers. Created with BioRender.com.
Genes 14 00577 g002
Figure 3. Epigenetic regulation of the β- and γ- globin gene expression. Schematic representation of the epigenetic profile of HBG1/2 and HBB promoters (represented by squares) in the fetal or fetal-like stages (fetal life, individuals with HPFH mutations and β-hemoglobinopathy patients; upper panel) and in the adult stage (healthy individuals; middle panel). The presence of DNA methylation and histone modifications (activating acetylation marks and activating or repressive methylation marks) is associated with an open or closed chromatin conformation that influences the accessibility of RNA polymerase II (RNAPII) and, consequently, the transcription rate as a whole. Green arrows indicate active transcription. CpG sites that are specifically methylated in the HBG1/2 genes in the adult stage are indicated and named after their distance from the transcription start site (e.g., −162, −53, −50, +6, +17, and +50). Created with BioRender.com.
Figure 3. Epigenetic regulation of the β- and γ- globin gene expression. Schematic representation of the epigenetic profile of HBG1/2 and HBB promoters (represented by squares) in the fetal or fetal-like stages (fetal life, individuals with HPFH mutations and β-hemoglobinopathy patients; upper panel) and in the adult stage (healthy individuals; middle panel). The presence of DNA methylation and histone modifications (activating acetylation marks and activating or repressive methylation marks) is associated with an open or closed chromatin conformation that influences the accessibility of RNA polymerase II (RNAPII) and, consequently, the transcription rate as a whole. Green arrows indicate active transcription. CpG sites that are specifically methylated in the HBG1/2 genes in the adult stage are indicated and named after their distance from the transcription start site (e.g., −162, −53, −50, +6, +17, and +50). Created with BioRender.com.
Genes 14 00577 g003
Figure 4. Overview of epigenome editors and artificial transcription factors: domains and epigenetic modifications. Schematic representation of epigenome editors and artificial transcription factors: (left panel). DNA-binding domains (DBDs) (zinc-finger, transcription activator-like effector (TALE) or dead Cas9 (dCas9)) can be fused with TFs or one or more epigenome modifiers. The epigenome modifier is an enzymatic domain that removes or adds epigenetic marks. The transcription factor recruits other activating/repressing co-factors depositing or removing epigenetic marks. The domains and related epigenetic modifications are listed in the table. Schematic representation of epigenetic modifications mediated directly or indirectly by epigenetic modifiers and TFs (right panel). Blue dots represent DNA methylation of CpG dinucleotides, and yellow dots represent histone methylation and orange dots represent histone acetylation. CpG methylation (CpGme); trimethylation of lysine 9 of histone 3 (H3K9me3); acetylation of lysine 9 of histone 3 (H3K9ac); methylation of lysine 4 of histone 3 (H3K4me); methylation of lysine 9 of histone 3 (H3K9me); acetylation of lysine 27 of histone 3 (H3K27ac); Histone deacetylase (HDAC); Ten-eleven translocation methylcytosine dioxygenase 1 (TET1); DNA (cytosine-5)-methyltransferase (DNMT); Krüppel associated box (KRAB); mSin3 interaction domain (SID); Lysine-specific histone demethylase 1A (KDM1A); histone acetyltransferase (HAT); Suppressor of variegation 3-9 homolog 1 (SUV39H1); Enhancer of zeste homolog 2 (EZH2). Created with BioRender.com.
Figure 4. Overview of epigenome editors and artificial transcription factors: domains and epigenetic modifications. Schematic representation of epigenome editors and artificial transcription factors: (left panel). DNA-binding domains (DBDs) (zinc-finger, transcription activator-like effector (TALE) or dead Cas9 (dCas9)) can be fused with TFs or one or more epigenome modifiers. The epigenome modifier is an enzymatic domain that removes or adds epigenetic marks. The transcription factor recruits other activating/repressing co-factors depositing or removing epigenetic marks. The domains and related epigenetic modifications are listed in the table. Schematic representation of epigenetic modifications mediated directly or indirectly by epigenetic modifiers and TFs (right panel). Blue dots represent DNA methylation of CpG dinucleotides, and yellow dots represent histone methylation and orange dots represent histone acetylation. CpG methylation (CpGme); trimethylation of lysine 9 of histone 3 (H3K9me3); acetylation of lysine 9 of histone 3 (H3K9ac); methylation of lysine 4 of histone 3 (H3K4me); methylation of lysine 9 of histone 3 (H3K9me); acetylation of lysine 27 of histone 3 (H3K27ac); Histone deacetylase (HDAC); Ten-eleven translocation methylcytosine dioxygenase 1 (TET1); DNA (cytosine-5)-methyltransferase (DNMT); Krüppel associated box (KRAB); mSin3 interaction domain (SID); Lysine-specific histone demethylase 1A (KDM1A); histone acetyltransferase (HAT); Suppressor of variegation 3-9 homolog 1 (SUV39H1); Enhancer of zeste homolog 2 (EZH2). Created with BioRender.com.
Genes 14 00577 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fontana, L.; Alahouzou, Z.; Miccio, A.; Antoniou, P. Epigenetic Regulation of β-Globin Genes and the Potential to Treat Hemoglobinopathies through Epigenome Editing. Genes 2023, 14, 577. https://doi.org/10.3390/genes14030577

AMA Style

Fontana L, Alahouzou Z, Miccio A, Antoniou P. Epigenetic Regulation of β-Globin Genes and the Potential to Treat Hemoglobinopathies through Epigenome Editing. Genes. 2023; 14(3):577. https://doi.org/10.3390/genes14030577

Chicago/Turabian Style

Fontana, Letizia, Zoe Alahouzou, Annarita Miccio, and Panagiotis Antoniou. 2023. "Epigenetic Regulation of β-Globin Genes and the Potential to Treat Hemoglobinopathies through Epigenome Editing" Genes 14, no. 3: 577. https://doi.org/10.3390/genes14030577

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop