Congenital sideroblastic anemia model due to ALAS2 mutation is susceptible to ferroptosis

X-linked sideroblastic anemia (XLSA), the most common form of congenital sideroblastic anemia, is caused by a germline mutation in the erythroid-specific 5-aminolevulinate synthase (ALAS2) gene. In XLSA, defective heme biosynthesis leads to ring sideroblast formation because of excess mitochondrial...

Full description

Saved in:
Bibliographic Details
Published inScientific reports Vol. 12; no. 1; p. 9024
Main Authors Ono, Koya, Fujiwara, Tohru, Saito, Kei, Nishizawa, Hironari, Takahashi, Noriyuki, Suzuki, Chie, Ochi, Tetsuro, Kato, Hiroki, Ishii, Yusho, Onodera, Koichi, Ichikawa, Satoshi, Fukuhara, Noriko, Onishi, Yasushi, Yokoyama, Hisayuki, Yamada, Rie, Nakamura, Yukio, Igarashi, Kazuhiko, Harigae, Hideo
Format Journal Article
LanguageEnglish
Published England Nature Publishing Group 30.05.2022
Nature Publishing Group UK
Nature Portfolio
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:X-linked sideroblastic anemia (XLSA), the most common form of congenital sideroblastic anemia, is caused by a germline mutation in the erythroid-specific 5-aminolevulinate synthase (ALAS2) gene. In XLSA, defective heme biosynthesis leads to ring sideroblast formation because of excess mitochondrial iron accumulation. In this study, we introduced ALAS2 missense mutations on human umbilical cord blood-derived erythroblasts; hereafter, we refer to them as XLSA clones. XLSA clones that differentiated into mature erythroblasts showed an increased frequency of ring sideroblast formation with impaired hemoglobin biosynthesis. The expression profiling revealed significant enrichment of genes involved in ferroptosis, which is a form of regulated cell death induced by iron accumulation and lipid peroxidation. Notably, treatment with erastin, a ferroptosis inducer, caused a higher proportion of cell death in XLSA clones. XLSA clones exhibited significantly higher levels of intracellular lipid peroxides and enhanced expression of BACH1, a regulator of iron metabolism and potential accelerator of ferroptosis. In XLSA clones, BACH1 repressed genes involved in iron metabolism and glutathione synthesis. Collectively, defective heme biosynthesis in XLSA clones could confer enhanced BACH1 expression, leading to increased susceptibility to ferroptosis. The results of our study provide important information for the development of novel therapeutic targets for XLSA.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:2045-2322
2045-2322
DOI:10.1038/s41598-022-12940-9