IJMMS_2024v14n4

International Journal of Molecular Medical Science, 2024, Vol.14, No.4, 203-215 http://medscipublisher.com/index.php/ijmms 207 Moreover, the distribution of HbF among erythrocytes is a significant factor in its protective role. Patients with a higher proportion of HbF-containing erythrocytes experience fewer complications compared to those with a lower proportion, even if their total HbF levels are similar (Steinberg, 2020). This differential distribution underscores the importance of not just the quantity but also the cellular distribution of HbF in ameliorating SCA symptoms. 4.2 Epigenetic regulation of HbF expression The expression of HbF is regulated by several genetic and epigenetic mechanisms. Key genetic loci such as BCL11A, HBS1L-MYB, and the β-globin gene cluster play pivotal roles in modulating HbF levels (Bae et al., 2012; Sales et al., 2022). Epigenetic modifications, including DNA methylation and histone modifications, also significantly influence HbF expression. For example, the silencing of the BCL11A gene, a known repressor of γ-globin gene expression, can lead to increased HbF levels (Métais et al., 2019; Quagliano et al., 2022). MicroRNAs (miRNAs) are another layer of epigenetic regulation affecting HbF expression. For instance, miR-144 has been shown to silence the NRF2 gene, which in turn represses γ-globin transcription. Inhibition of miR-144 can reverse this effect, thereby increasing HbF levels (Li et al., 2019). These findings highlight the complex interplay of genetic and epigenetic factors in the regulation of HbF, offering multiple potential targets for therapeutic intervention. 4.3 Therapeutic strategies targeting HbF Several therapeutic strategies aim to increase HbF levels to treat SCA. Pharmacological agents like hydroxyurea have been widely used to induce HbF production. However, the response to hydroxyurea varies among patients, partly due to genetic polymorphisms that affect drug efficacy and toxicity. Genetic studies have identified single-nucleotide polymorphisms (SNPs) in genes such as BCL11A that influence the response to hydroxyurea, suggesting that personalized medicine approaches could optimize treatment outcomes (Sales et al., 2022). Gene editing technologies, particularly CRISPR/Cas9, offer promising avenues for permanently increasing HbF levels. By targeting and disrupting repressor elements in the γ-globin gene promoters, such as those bound by BCL11A, researchers have successfully induced therapeutic levels of HbF in preclinical models (Métais et al., 2019; Quagliano et al., 2022). These advances in gene editing not only provide a potential cure for SCA but also highlight the importance of understanding the genetic and epigenetic regulation of HbF for developing effective therapies. 5 Histone Modifications and Sickle Cell Anemia 5.1 Histone acetylation and deacetylation Histone acetylation and deacetylation are critical processes in the regulation of gene expression, impacting various diseases, including sickle cell anemia. Histone acetylation, typically associated with gene activation, is mediated by histone acetyltransferases (HATs), while histone deacetylation, associated with gene repression, is mediated by histone deacetylases (HDACs). The balance between these opposing activities determines the chromatin state and, consequently, gene expression patterns. In the context of sickle cell anemia, the modulation of histone acetylation has shown potential therapeutic benefits. For instance, HDAC inhibitors (HDIs) have been explored for their ability to reactivate fetal hemoglobin (HbF) production, which can ameliorate the symptoms of sickle cell disease by inhibiting the polymerization of sickle hemoglobin (HbS) (Kelly et al., 2010; Bajbouj et al., 2021). The therapeutic potential of HDIs is supported by their efficacy in other diseases, such as cancer and cardiovascular diseases, where they modulate gene expression to counteract disease progression (Pons et al., 2008; Knethen et al., 2020). 5.2 Histone methylation Histone methylation is another crucial epigenetic modification that can either activate or repress gene transcription, depending on the specific histone residues and the number of methyl groups added. This modification is carried out by histone methyltransferases (HMTs) and reversed by histone demethylases (HDMTs).

RkJQdWJsaXNoZXIy MjQ4ODYzNQ==