Sulfide catabolism ameliorates hypoxic brain injury

The mammalian brain is highly vulnerable to oxygen deprivation, yet the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. Hypoxia induces accumulation of hydrogen sulfide, a gas that inhibits mitochondrial respiration. Here, we show that, in mice, rats, and nat...

Full description

Saved in:
Bibliographic Details
Published inNature communications Vol. 12; no. 1; pp. 3108 - 19
Main Authors Marutani, Eizo, Morita, Masanobu, Hirai, Shuichi, Kai, Shinichi, Grange, Robert M H, Miyazaki, Yusuke, Nagashima, Fumiaki, Traeger, Lisa, Magliocca, Aurora, Ida, Tomoaki, Matsunaga, Tetsuro, Flicker, Daniel R, Corman, Benjamin, Mori, Naohiro, Yamazaki, Yumiko, Batten, Annabelle, Li, Rebecca, Tanaka, Tomohiro, Ikeda, Takamitsu, Nakagawa, Akito, Atochin, Dmitriy N, Ihara, Hideshi, Olenchock, Benjamin A, Shen, Xinggui, Nishida, Motohiro, Hanaoka, Kenjiro, Kevil, Christopher G, Xian, Ming, Bloch, Donald B, Akaike, Takaaki, Hindle, Allyson G, Motohashi, Hozumi, Ichinose, Fumito
Format Journal Article
LanguageEnglish
Published England Nature Publishing Group 25.05.2021
Nature Publishing Group UK
Nature Portfolio
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:The mammalian brain is highly vulnerable to oxygen deprivation, yet the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. Hypoxia induces accumulation of hydrogen sulfide, a gas that inhibits mitochondrial respiration. Here, we show that, in mice, rats, and naturally hypoxia-tolerant ground squirrels, the sensitivity of the brain to hypoxia is inversely related to the levels of sulfide:quinone oxidoreductase (SQOR) and the capacity to catabolize sulfide. Silencing SQOR increased the sensitivity of the brain to hypoxia, whereas neuron-specific SQOR expression prevented hypoxia-induced sulfide accumulation, bioenergetic failure, and ischemic brain injury. Excluding SQOR from mitochondria increased sensitivity to hypoxia not only in the brain but also in heart and liver. Pharmacological scavenging of sulfide maintained mitochondrial respiration in hypoxic neurons and made mice resistant to hypoxia. These results illuminate the critical role of sulfide catabolism in energy homeostasis during hypoxia and identify a therapeutic target for ischemic brain injury.
ISSN:2041-1723
2041-1723
DOI:10.1038/s41467-021-23363-x