Abstract

β-Thalassaemia is caused by over 300 mutations in and around the β-globin gene that lead to impaired synthesis of β-globin. The expression of α-globin continues normally, resulting in an excess of α-globin chains within red blood cells and their precursors. These unpaired α-globin chains form unstable α-hemichromes that trigger cascades of events to generate reactive oxygen species, leading to ineffective erythropoiesis and haemolysis in patients with β-thalassaemia. The clinical genetic data reported over several decades have demonstrated how the coinheritance of α-thalassaemia ameliorates the disease phenotype of β-thalassaemia. Thus, it is evident that down-regulation of the α-globin gene expression in patients with β-thalassaemia could ameliorate or even cure β-thalassaemia. Over the last few years, significant progress has been made in utilising this pathway to devise a cure for β-thalassaemia. Most research has been done to alter the epigenetic landscape of the α-globin locus or the well-characterised distant enhancers of α-globin. In vitro, pre-clinical studies on primary human erythroid cells have unveiled inhibition of histone lysine demethylation and histone deacetylation as potential targets to achieve selective downregulation of α-globin through epigenetic drug targeting. CRISPR based genome editing has been successfully used in vitro to mutate α-globin genes or enhancers of α-goblin to achieve clinically significant knockdowns of α-globin to the levels beneficial for patients with β-thalassaemia. This review summarises the current knowledge on the regulation of human α-globin genes and the clinical genetic data supporting the pathway of targeting α-globin as a treatment for β-thalassaemia. It also presents the progress of epigenetic drug and genome editing approaches currently in development to treat β-thalassaemia.

Highlights

  • INTRODUCTIONΒ-Thalassaemia is one of the most common monogenic diseases affecting the red blood cells (RBC) (Taher et al, 2018)

  • Specialty section: This article was submitted to Genome Editing in Blood Disorders, a section of the journal Frontiers in Genome Editing

  • This review summarises the current knowledge on the regulation of human α-globin genes and the clinical genetic data supporting the pathway of targeting α-globin as a treatment for β-thalassaemia

Read more

Summary

INTRODUCTION

Β-Thalassaemia is one of the most common monogenic diseases affecting the red blood cells (RBC) (Taher et al, 2018). Another clinical trial (ClinicalTrials.gov Identifier NCT03655678) used CRISPR/Cas gene editing to disrupt an erythroid-specific enhancer region of BCL11A In this trial, a female patient with transfusion-dependent β-thalassaemia who received the autologous genome edited CD34+ HSC product CTX001 showed sustained elevations of HbF and remained transfusion independent (last reviewed after 18 months posttransplant) (Frangoul et al, 2021). A female patient with transfusion-dependent β-thalassaemia who received the autologous genome edited CD34+ HSC product CTX001 showed sustained elevations of HbF and remained transfusion independent (last reviewed after 18 months posttransplant) (Frangoul et al, 2021) In addition to these clinical trials, the disruption of a γ-globin gene promoter motif bound by BCL11A by CRISPR/Cas has induced HbF to potentially therapeutic levels in in vitro studies (Métais et al, 2019).

Synergistic Genome Editing
Future Challenges
Findings
CONCLUSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call