International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Sickle cell anemia (SCA) is the most common form of sickle cell disease (SCD), a group of genetic disorders caused by defective hemoglobin genes (1). SCA is a prevalent hemoglobin anomaly worldwide and a major cause of child mortality in West Africa, accounting for 9-16% of deaths (2). It is caused by a point mutation on chromosome 11, where valine replaces glutamic acid in the β-globin chain, leading to hemoglobin S (HbS) formation. Under low oxygen conditions, HbS polymerizes, reducing red blood cell mobility, increasing fragility, and causing vaso-occlusive complications and hemolysis (3). Even though the molecular basis of SCA is well-known and the pathophysiology of the disease is better understood, there has been a substantial lag in its application to the development of safe and effective therapies. Recent studies underscore the therapeutic potential of reactivating fetal hemoglobin (HbF), which is mediated by epigenetic processes. The present review discusses the mechanisms through which targeting epigenetic regulators and transcriptional repressors can increase HbF and alleviate SCA pathology.
HbF, composed of two α- and two γ-globin chains produced by the γ-globin genes, is dominant throughout fetal life, but decreases postnatally due to hemoglobin switching (4). Adults have residual HbF levels (usually 1%) (5). The γ-globin genes (HBG1 and HBG2) are crucial in SCA pathophysiology, as an increased expression of HbF reduces disease severity, improves red blood cell lifespan and enhances erythropoiesis efficiency (6). Individuals with the hereditary persistence of HbF are often clinically asymptomatic (7). Thus, re-inducing HbF through γ-globin gene activation offers a promising therapeutic strategy.
Epigenetic mechanisms regulate gene expression without altering the deoxyribonucleic acid (DNA) sequence and include the following:
The methylation of CpG islands in the γ-globin promoter inhibits its expression (8). This suppression is carried out by DNA methyltransferases (DNMTs). DNMT inhibitors can demethylate the γ-globin promoter and restore HbF expression (9).
The acetylation of lysine residues via histone acetyltransferases (HATs) reduces histone-DNA interactions, making chromatin more accessible to transcription factors and promoting gene expression, whereas deacetylation via histone deacetylases (HDACs) compacts chromatin and represses gene expression (10). Histone methylation by histone methyltransferases (HMTs) or demethylation by lysine-specific demethylase 1 (LSD1) also influences γ-globin transcription (11). Protein arginine methyltransferase 5 (PRMT5) and H3K27me3 are associated with γ-globin silencing (12). Adenosine-20,30-dialdehyde, a methyltransferase inhibitor, was previously demonstrated to stimulate γ-globin gene expression in human early-stage cultured red blood cell precursors, suggesting that PRMT5 catalytic activity may be mechanically associated with γ-globin gene suppression (13). Transcriptionally active chromatin is associated with the trimethylation of histone H3, specifically lysine at position 4 (H3K4me3), while H3K27me3 and H3K9Me3 result in tight chromatin, which suppresses gene expression (14).
B-cell lymphoma/leukemia 11A (BCL11A) and zinc finger and BTB domain containing 7A (ZBTB7A) are key transcriptional repressors of the γ-globin gene. To maintain a suppressed chromatin state, they enlist the nucleosome remodeling and deacetylase complex/chromodomain helicase DNA-binding protein 4 (NuRD/CHD4) (15). Other elements that control these repressors and function as indirect targets for HbF induction include zinc finger protein 410 (ZNF410) and hypermethylated in cancer 2 (HIC2) (16). Furthermore, epigenetic control is reversible and dynamic, rendering it a potential target for therapeutic applications (17).
Investigations into certain HDAC antagonists, such as short-chain fatty acids, have demonstrated potential for triggering the production of HbF (18). The clinical development of phosphodiesterase 9 inhibitors (PF-04447943; ClinicalTrials.gov Identifier: NCT02114203-completed), mechanistic target of rapamycin (mTOR) inhibitors, such as sirolimus (Sirthalaclin trial; ClinicalTrials.gov Identifier: NCT03877809-completed; mechanistic proof of HbF induction relevant to sickle cell disease), and 3,4-dihydroxyphenylalanine DOPA decarboxylase inhibitors such as benserazide (BENeFiTS trial; ClinicalTrials.gov Identifier: NCT04432623-ongoing) has demonstrated enhanced γ-globin or HbF expression, although none has established long-term transfusion independence in hemoglobinopathies (19-21). The molecular links between the alleged mechanisms of action of these agents and epigenetic modifications that may influence HbF production remain unclear. The selective erythroid deletion of BCL11A in mice has shown promise in the treatment of SCA (22), leading to clinical studies using genome modification with promising outcomes (23,24).
Single BCL11A or ZBTB7A knockout (KO) leads to 40-50% HbF expression in human erythroid progenitors, whereas dual KO results in ~90-100% HbF, indicating their independent contribution to repression. While adult-stage ZBTB7A deletion causes moderate macrocytic anemia, complete knockout results in embryonic mortality due to severe anemia (15). In hematopoietic stem and progenitor cell (HSPC)-derived erythroblasts, ZBTB7A reduction also postpones erythroid development. Future research is required to identify an optimal therapeutic strategy where selectively inhibiting ZBTB7A in adults induces HbF without adverse effects. Expanding upon the fundamental knowledge of transcription factors linked to HbF repression, the following paragraphs discuss other epigenetic regulators that provide novel therapeutic options for the reactivation of γ-globin expression.
ZNF410 is a transcription factor with a single known target, CHD4, to stimulate the transcription of γ-globin (HBG1/2) genes, rendering it a suitable target for HbF reactivation (25). The deletion of ZNF410 does not affect erythroid development, and leads to a marked increase in the levels of HBG1/2, and a corresponding decrease in the level of β-globin gene in various cell models and mice. Dual BCL11A and ZNF410 knockdown enhances HbF production more effectively than targeting BCL11A alone, rendering ZNF410 a promising therapeutic target for SCD and β-thalassemia (26).
HIC2 is a zinc finger transcription factor that plays a critical role in gene regulation and development. It functions as a transcriptional repressor for BCL11A, thereby promoting HbF production by reducing chromatin accessibility at BCL11A enhancers (16). Additionally, widely interspaced zinc finger (WIZ) degradation via molecular glue degraders has been explored as a strategy to induce HbF (27). OTQ923, a genome-edited hematopoietic stem and progenitor cell product, was evaluated in a first-in-patient phase I/II clinical study to decrease the biological activity of BCL11A, boost HbF and decrease SCD-related complications. However, that study was discontinued for commercial reasons rather than safety concerns (ClinicalTrials.gov Identifier: NCT04443907). Presently under investigation is a multi-center, open-label, non-randomized, phase 2 research (GRASP, BMT CTN 2001) that targets BCL11A to raise HbF in patients with SCD (ClinicalTrials.gov Identifier: NCT05353647).
Inhibitors of DNMT (such as decitabine), HDAC (such as butyrate, vorinostat and givinostat), and LSD1 have been demonstrated to increase HbF (28). Hydroxyurea, however, not epigenetic in effect, remains a key component of therapy due to its potential to produce HbF (29). Through DNA hypomethylation, DNMT inhibitors, such as decitabine and 5-azacytidine can restore the expression of γ-globin genes (30). However, the clinical development of 5-azacytidine for hemoglobinopathies was discontinued due to severe effects, although the medication is still used for specific blood-related cancers (31). The clinical success of hydroxyurea is mostly owing to its ability to produce HbF, possibly through epigenetic modifications (32).
In SCA, hydroxyurea raises HbF levels via two distinct processes: i) Ribonucleotide reductase inhibition, which encourages the selection of erythroid precursors with a high HbF expression; and ii) the direct selection of HbF cell formation by inhibition of soluble guanylate cyclase (28).
The most widely accepted theory is that it triggers ‘stress erythropoiesis’, which recruits primitive erythroid precursors that have the capacity to produce HbF, resulting in elevated HbF levels and a corresponding decrease in the proportion of HbS (32). Hydroxyurea alters methylation and demethylation processes at the γ-globin gene promoter to boost γ-globin expression; however, these changes do not significantly increase fetal globin reactivation (33). Another proposed mechanism involves hydroxyurea-driven nitric oxide production, which increases intracellular cyclic guanosine monophosphate-protein kinase G, ultimately inducing HbF synthesis. However, the exact mechanisms involved remain unknown.
HDAC1 and HDAC2 inhibitors have shown promise in the treatment of SCA by inducing HbF production. Non-selective HDAC inhibitors, such as butyrates, have been shown to be effective (34). However, side-effects and gene expression issues need to be addressed for long-term effective treatments, including the selective suppression of HDAC-1 or HDAC-2(35). The selective inhibition of HDAC1 and HDAC2 inhibitors modifies the chromatin structure of the β-globin gene, producing HbF without directly affecting bone marrow (36). HDAC inhibitors enhance histone acetylation, opening chromatin and increasing γ-globin transcription.
According to Di Micco et al (37), HDAC-1 and HDAC-2 share 85% identity and 93% similarity, rendering selective chemicals challenging. However, they regulate gene expression for γ-globin induction, eliminating the need for selectivity (37). Acetylon Pharmaceuticals, Inc. developed ACY-957, a chemical with a zinc-chelating benzamide group, that increases histone acetylation and GATA2 binding in cells. It suppresses HDAC1/2 and enhances γ-globin mRNA expression (38). The targeted inhibition of HDAC1 and HDAC2 in adult human red blood cells increases HbF production without altering the cell cycle (39). Romidepsin (40), the HDAC1/3 inhibitor, MS-275 (entinostat) (41), and givinostat, a hydroxamate inhibitor, also promote γ-globin gene expression without disrupting cell growth (42). The genetic knockdown of HDAC1/2 increases HbF levels, and combination therapy with hydroxyurea enhances γ-globin synthesis (43). This is further enhanced by MS-275(43). A phase 1 trial of panobinostat, a pan-HDAC inhibitor, is currently underway in adult patients with SCD (ClinicalTrials.gov identifier: NCT01245179). CT-101, a class I HDAC inhibitor, has been reported to selectively activate γ-globin transcription, leading to elevated F-cells and HbF levels with minimal cytotoxicity. In combination with hydroxyurea, it results in an even greater increase in HbF levels.
These results highlight the potential of these agents as pharmacological inducers of HbF. However, despite promising preclinical findings, there is still a dearth of human clinical proof (44). A summary of the aforementioned pharmacologic HbF inducers, their mechanisms of action and current clinical status is presented in Table I.
The CRISPR/Cas9 technique can be used to edit the epigenome and modify methylation patterns of DNA or histone changes in the γ-globin gene locus. It enables precision genome editing to disrupt HbF repressors or mimic the hereditary persistence of fetal hemoglobin (45). Preclinical and early clinical research has shown that targeting BCL11A or its enhancer, ZBTB7A, or altering γ-globin promoters can successfully regenerate HbF (46). Gene therapy is probably safer than allogenic hematopoietic stem cell transplant, though it is associated with adverse effects and is expensive (47).
CRISPR/Cas9 identified nuclear factor 1A and nuclear factor 1X as HBG1/2 repressors that are cooperative and expressed at higher levels in adult erythroid cells than in fetal cells. These transcription factors inhibit HBG1/2 and promote BCL11A expression, playing a vital role in the fetal-to-adult hemoglobin transition (48). A CRISPR/Cas9 screen demonstrated that globin gene switching is affected by ubiquitin-proteasome components, with von Hippel-Lindau ubiquitin ligase (VHL E3) depletion stabilizing hypoxia-inducible factor 1α, which initiates the transcription of the γ-globin gene through enhanced chromatin accessibility and transcriptional activators. This implies that prolyl hydroxylase domain enzyme inhibition or hypoxia may be used as therapy for β-hemoglobinopathy (49).
Frati et al (50) employed CRISPR/Cas9 gene editing to target lymphoma-related factor (LRF) repressor sites in γ-globin promoters, effectively reactivating HbF expression. This genome editing strategy in patient-derived HSPCs and healthy donor cells results in a significant HbF production and a high disruption of lymphoma-related factor binding sites (LRF BS). In SCD HSPCs, LRF BS disruption was more effective but also decreased myeloid bias and engraftment. Genes linked to inflammatory reactions and DNA damage were upregulated during the editing process (50). Furthermore, the USA and UK authorized the commercial gene therapy product, Casgevy, a CRISPR-based treatment that targets BCL11A, for use in 2023(51). Despite this achievement, a number of obstacles still limit its clinical application: Patients must be at least 12 years old to be eligible; there are safety concerns about potential mosaicism and off-target effects; and the sustained effectiveness of HbF induction over time is unclear. Moreover, high costs and infrastructure demands pose substantial obstacles to worldwide access, especially in sub-Saharan Africa, where the prevalence of SCA is high. Additionally, ethical concerns have been raised, particularly in light of the possibility of germline editing, which might bring heritable modifications into the human genome (52).
Large-scale, long-term clinical trials are required to validate preclinical results and prove efficacy and safety in humans. Ethical issues also need to be addressed, particularly in relation to fair access, the possible unintended consequences of genome editing, and the necessity of clearly outlining the advantages, disadvantages, and available options when treating pediatric patients. Future studies should therefore ensure safety, improve selectivity, optimize delivery strategies, lower expenses and guarantee access to innovative treatments. Researchers, clinicians and policymakers must work together to make sure that discoveries result in real benefits for patients with SCA worldwide.
The present review is based on preclinical studies and early-phase trials. The lack of specificity in a number of epigenetic agents poses safety issues. Furthermore, accessibility and cost barriers restrict the potential of CRISPR treatments. The regulatory network that controls γ-globin is still not fully known, and there is still no explanation for the variation in patient response.
Although hydroxyurea is a key therapy, its variable efficacy and side-effects pose a challenge. DNMT and HDAC inhibitors are hindered by their toxicity, and CRISPR-based therapies such as Casgevy hold promise; however, high costs and complexity persist. These translational hurdles need to be overcome to demonstrate clinical effectiveness, particularly in areas with limited resources and high prevalence of the disease.
In conclusion, epigenetic modification provides a compelling approach for the treatment of SCA via HbF reactivation. Advances in pharmacology and genome editing have sped up clinical translation. Selective HDAC1/2 inhibition and CRISPR/Cas9-based treatments are particularly promising. To establish HbF reactivation as a safe, efficient and widely available treatment for SCA, further research on precision gene editing and specific epigenetic modulators is mandatory.
Not applicable.
Funding: No funding was received.
Not applicable.
SAA was involved in the conceptualization of the study, and in the drafting and editing of the manuscript. FB and AYA were involved the reviewing and editing of the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Chauhan W and Zennadi R: Keap1-Nrf2 heterodimer: A therapeutic target to ameliorate sickle cell disease. Antioxidants (Basel). 12(740)2023.PubMed/NCBI View Article : Google Scholar | |
|
Acharya B, Mishra DP, Barik B, Mohapatra RK and Sarangi AK: Recent progress in the treatment of sickle cell disease: An up-to-date review. Beni-Suef Univ J Basic Appl Sci. 12(38)2023. | |
|
Demers M, Sturtevant S, Guertin KR, Gupta D, Desai K, Vieira BF, Li W, Hicks A, Ismail A, Gonçalves BP, et al: MetAP2 inhibition modifies hemoglobin S to delay polymerization and improves blood flow in sickle cell disease. Blood Adv. 5:1388–1402. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Fontana L, Alahouzou Z, Miccio A and Antoniou P: Epigenetic regulation of β-globin genes and the potential to treat hemoglobinopathies through epigenome editing. Genes (Basel). 14(577)2023.PubMed/NCBI View Article : Google Scholar | |
|
Costa D, Capuano M, Sommese L and Napoli C: Impact of epigenetic mechanisms on therapeutic approaches of hemoglobinopathies. Blood Cells Mol Dis. 55:95–100. 2015.PubMed/NCBI View Article : Google Scholar | |
|
El Hoss S, Cochet S, Godard A, Yan H, Dussiot M, Frati G, Boutonnat-Faucher B, Laurance S, Renaud O, Joseph L, et al: Fetal hemoglobin rescues ineffective erythropoiesis in sickle cell disease. Hematologica. 106:2707–2719. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Hara Y, Kawabata E, Lemgart VT, Bronson PG, Hicks A, Peters R, Krishnamoorthy S, Ribeil JA, Schmunk LJ, Eglinton J, et al: Genomic discovery and functional validation of MRP1 as a novel fetal hemoglobin modulator and potential therapeutic target in sickle cell disease. medRxiv, 2023. | |
|
Bao X, Zuo Y, Chen D and Zhao C: DNA methylation patterns of β-globin cluster in β-thalassemia patients. Clin Epigenetics. 12(187)2020.PubMed/NCBI View Article : Google Scholar | |
|
Gilmartin AG, Groy A, Gore ER, Atkins C, Long ER, Montoute MN, Wu Z, Halsey W, McNulty DE, Ennulat D, et al: In vitro and in vivo induction of fetal hemoglobin with a reversible and selective DNMT1 inhibitor. Hematologica. 106:1979–1987. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Shvedunova M and Akhtar A: Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol. 23:329–349. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Pavan AR, Lopes JR and Dos Santos JL: The state of the art of fetal hemoglobin-inducing agents. Expert Opin Drug Discov. 17:1279–1293. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Zhao Q, Rank G, Tan YT, Li H, Moritz RL, Simpson RJ, Cerruti L, Curtis DJ, Patel DJ, Allis CD, et al: PRMT5-mediated methylation of histone H4R3 recruits DNMT3A, coupling histone and DNA methylation in gene silencing. Nat Struct Mol Biol. 16:304–311. 2009.PubMed/NCBI View Article : Google Scholar | |
|
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. 11(14)2013.PubMed/NCBI View Article : Google Scholar | |
|
Wu X, Xu H, Xia E, Gao L, Hou Y, Sun L, Zhang H and Cheng Y: Histone modifications in the regulation of erythropoiesis. Ann Med. 57(2490824)2025.PubMed/NCBI View Article : Google Scholar | |
|
Masuda T, Wang X, Maeda M, Canver MC, Sher F, Funnell APW, Fisher C, Suciu M, Martyn GE, Norton LJ, et al: Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science. 351:285–289. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Huang P, Peslak SA, Ren R, Khandros E, Qin K, Keller CA, Giardine B, Bell HW, Lan X, Sharma M, et al: HIC2 controls developmental hemoglobin switching by repressing BCL11A transcription. Nat Genet. 54:1417–1426. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Wu YL, Lin ZJ, Li CC, Lin X, Shan SK, Guo B, Zheng MH, Li F, Yuan LQ and Li ZH: Epigenetic regulation in metabolic diseases: Mechanisms and advances in clinical study. Signal Transduct Target Ther. 8(98)2023.PubMed/NCBI View Article : Google Scholar | |
|
Fathallah H, Weinberg RS, Galperin Y, Sutton M and Atweh GF: Role of epigenetic modifications in normal globin gene regulation and butyrate-mediated induction of fetal hemoglobin. Blood. 110:3391–3397. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Charnigo RJ, Beidler D, Rybin D, Pittman DD, Tan B, Howard J, Michelson AD, Frelinger AL III and Clarke N: PF-04447943, a phosphodiesterase 9A inhibitor, in stable sickle cell disease patients: A phase Ib randomized, placebo-controlled study. Clin Transl Sci. 12:180–188. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Zuccato C, Cosenza LC, Zurlo M, Gasparello J, Papi C, D'Aversa E, Breveglieri G, Lampronti I, Finotti A, Borgatti M, et al: Expression of γ-globin genes in β-thalassemia patients treated with sirolimus: Results from a pilot clinical trial (sirthalaclin). Ther Adv Hematol. 13(20406207221100648)2022.PubMed/NCBI View Article : Google Scholar | |
|
National Library of Medicine: The BENeFiTS Trial in Beta Thalassemia Intermedia (PB04-001). Clinicaltrials.gov identifier: NCT04432623. https://clinicaltrials.gov/study/NCT04432623?cond=%22Beta%20thalassemia%22&rank=5. Accessed November 1, 2025. | |
|
Xu J, Peng C, Sankaran VG, Shao Z, Esrick EB, Chong BG, Ippolito GC, Fujiwara Y, Ebert BL, Tucker PW and Orkin SH: Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science. 334:993–996. 2011.PubMed/NCBI View Article : Google Scholar | |
|
Esrick EB, Lehmann L, Biffi A, Achebe M, Brendel C, Ciuculescu MF, Daley H, MacKinnon B, Morris E, Federico A, et al: Post-transcriptional genetic silencing of BCL11A to treat sickle cell disease. N Engl J Med. 384:205–215. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Fu B, Liao S, Chen S, Li W, Wang Q, Hu J, Yang F, Hsiao S, Jiang Y, Wang L, et al: CRISPR-Cas9-mediated gene editing of the BCL11A enhancer for pediatric β0/β0 transfusion-dependent β-thalassemia. Nat Med. 28:1573–1580. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Stamatoyannopoulos G: Control of globin gene expression during development and erythroid differentiation. Exp Hematol. 33:259–271. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Liu B, Brendel C, Vinjamur DS, Zhou Y, Harris C, McGuinness M, Manis JP, Bauer DE, Xu H and Williams DA: Development of a double shmiR lentivirus effectively targeting both BCL11A and ZNF410 for enhanced induction of fetal hemoglobin to treat β-hemoglobinopathies. Mol Ther. 30:2693–2708. 2022. | |
|
Ting PY, Borika S, Kerrigan J, Thomsen NM, Aghania E, Hinman AE, Reyes A, Pizzato N, Fodor BD, Wu F, et al: A molecular glue degrader of the WIZ transcription factor for fetal hemoglobin induction. Science. 385:91–99. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Bou-Fakhredin R, De Franceschi L, Motta I, Cappellini MD and Taher AT: Pharmacological induction of fetal hemoglobin in β-thalassemia and sickle cell disease: An updated perspective. Pharmaceuticals (Basel). 15(753)2022.PubMed/NCBI View Article : Google Scholar | |
|
Ginder GD: Epigenetic regulation of fetal globin gene expression in adult erythroid cells. Transl Res. 165:115–125. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Lavelle D, Engel JD and Saunthararajah Y: Fetal hemoglobin induction by epigenetic drugs. Semin Hematol. 55:60–67. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Charache S, Dover G, Smith K, Talbot CC Jr, Moyer M and Boyer S: Treatment of sickle cell anemia with 5-azacytidine results in increased fetal hemoglobin production and is associated with nonrandom hypomethylation of DNA around the gamma-delta-beta-globin gene complex. Proc Natl Acad Sci USA. 80:4842–4846. 1983.PubMed/NCBI View Article : Google Scholar | |
|
López Rubio M and Argüello Marina M: The current role of hydroxyurea in the treatment of sickle cell anemia. J Clin Med. 13(6404)2024.PubMed/NCBI View Article : Google Scholar | |
|
Pule GD, Mowla S, Novitzky N, Wiysonge CS and Wonkam A: A systematic review of known mechanisms of hydroxyurea-induced fetal hemoglobin for treatment of sickle cell disease. Expert Rev Hematol. 8:669–679. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Weinberg RS, Ji X, Sutton M, Perrine S, Galperin Y, Li Q, Liebhaber SA, Stamatoyannopoulos G and Atweh GF: Butyrate increases the efficiency of translation of gamma-globin mRNA. Blood. 105:1807–1809. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Bradner JE, Mak R, Tanguturi SK, Mazitschek R, Haggarty SJ, Ross K, Chang CY, Bosco J, West N, Morse E, et al: Chemical genetic strategy identifies histone deacetylase 1 (HDAC1) and HDAC2 as therapeutic targets in sickle cell disease. Proc Natl Acad Sci USA. 107:12617–12622. 2010.PubMed/NCBI View Article : Google Scholar | |
|
Lohani N, Bhargava N, Munshi A and Ramalingam S: Pharmacological and molecular approaches for the treatment of β-hemoglobin disorders. J Cell Physiol. 233:4563–4577. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Di Micco S, Chini MG, Terracciano S, Bruno I, Riccio R and Bifulco G: Structural basis for the design and synthesis of selective HDAC inhibitors. Bioorg Med Chem. 21:3795–3807. 2013.PubMed/NCBI View Article : Google Scholar | |
|
Shearstone JR, Golonzhka O, Chonkar A, Tamang D, VanDuzer JH, Jones SS and Jarpe MB: Chemical inhibition of histone deacetylases 1 and 2 induces fetal hemoglobin through activation of GATA2. PLoS One. 11(e0153767)2016.PubMed/NCBI View Article : Google Scholar | |
|
Esrick EB, McConkey M, Lin K, Frisbee A and Ebert BL: Inactivation of HDAC1 or HDAC2 induces gamma globin expression without altering cell cycle or proliferation. Am J Hematol. 90:624–628. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Habibi H, Atashi A, Abroun S and Noruzinia M: Synergistic effect of simvastatin and romidepsin on gamma-globin gene induction. Cell J. 20:576–583. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Dai Y, Sangerman J, Luo HY, Fucharoen S, Chui DHK, Faller DV and Perrine SP: Therapeutic fetal-globin inducers reduce transcriptional repression in hemoglobinopathy erythroid progenitors through distinct mechanisms. Blood Cell Mol Dis. 56:62–69. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Ronzoni L, Sonzogni L, Fossati G, Modena D, Trombetta E, Porretti L and Cappellini MD: Modulation of gamma globin genes expression by histone deacetylase inhibitors: An in vitro study. Br J Hematol. 165:714–721. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Cisneros GS and Thein SL: Recent advances in the treatment of sickle cell disease. Front Physiol. 11(435)2020.PubMed/NCBI View Article : Google Scholar | |
|
Takezaki M, Li B, Xu H, Patel N, Lucas R, Cerbone RE, Koti S, Hendrick CL, Junker LH and Pace BS: The histone deacetylase inhibitor CT-101 flips the switch to fetal hemoglobin expression in sickle cell disease mice. PLoS One. 20(e0323550)2025.PubMed/NCBI View Article : Google Scholar | |
|
Traxler EA, Yao Y, Wang YD, Woodard KJ, Kurita R, Nakamura Y, Hughes JR, Hardison RC, Blobel GA, Li C and Weiss MJ: A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat Med. 22:987–990. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S, et al: CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells. Nature. 539:384–389. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Xu L, Lahiri P, Skowronski J, Bhatia N, Lattanzi A and Porteus MH: Molecular dynamics of genome editing with CRISPR-Cas9 and rAAV6 virus in human HSPCs to treat sickle cell disease. Mol Ther Methods Clin Dev. 30:317–331. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Qin K, Huang P, Feng R, Keller CA, Peslak SA, Khandros E, Saari MS, Lan X, Mayuranathan T, Doerfler PA, et al: Dual function NFI factors control fetal hemoglobin silencing in adult erythroid cells. Nat Genet. 54:874–884. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Feng R, Mayuranathan T, Huang P, Doerfler PA, Li Y, Yao Y, Zhang J, Palmer LE, Mayberry K, Christakopoulos GE, et al: Activation of γ-globin expression by hypoxia-inducible factor 1α. Nature. 610:783–790. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Frati G, Brusson M, Sartre G, Mlayah B, Felix T, Chalumeau A, Antoniou P, Hardouin G, Concordet JP, Romano O, et al: Safety and efficacy studies of CRISPR-Cas9 treatment of sickle cell disease highlights disease-specific responses. Mol Ther. 32:4337–4352. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Song X, Liu J, Chen T, Zheng T, Wang X and Guo X: Gene therapy and gene editing strategies in inherited blood disorders. J Genet Genomics. 51:1162–1172. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Singh A, Irfan H, Fatima E, Nazir Z, Verma A and Akilimali A: Revolutionary breakthrough: FDA approves CASGEVY, the first CRISPR/Cas9 gene therapy for sickle cell disease. Ann Med Surg (Lond). 86:4555–4559. 2024.PubMed/NCBI View Article : Google Scholar |