Cytosolic iron-sulfur assembly is evolutionarily tuned by a cancer-amplified ubiquitin ligase

JL Weon, SW Yang, PR Potts - Molecular cell, 2018 - cell.com
Molecular cell, 2018cell.com
The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway functions to incorporate
inorganic Fe-S cofactors into a variety of proteins, including several DNA repair enzymes.
However, the mechanisms regulating the CIA pathway are unknown. We describe here that
the MAGE-F1-NSE1 E3 ubiquitin ligase regulates the CIA pathway through ubiquitination
and degradation of the CIA-targeting protein MMS19. Overexpression or knockout of MAGE-
F1 altered Fe-S incorporation into MMS19-dependent DNA repair enzymes, DNA repair …
Summary
The cytosolic iron-sulfur (Fe-S) cluster assembly (CIA) pathway functions to incorporate inorganic Fe-S cofactors into a variety of proteins, including several DNA repair enzymes. However, the mechanisms regulating the CIA pathway are unknown. We describe here that the MAGE-F1-NSE1 E3 ubiquitin ligase regulates the CIA pathway through ubiquitination and degradation of the CIA-targeting protein MMS19. Overexpression or knockout of MAGE-F1 altered Fe-S incorporation into MMS19-dependent DNA repair enzymes, DNA repair capacity, sensitivity to DNA-damaging agents, and iron homeostasis. Intriguingly, MAGE-F1 has undergone adaptive pseudogenization in select mammalian lineages. In contrast, MAGE-F1 is highly amplified in multiple human cancer types and amplified tumors have increased mutational burden. Thus, flux through the CIA pathway can be regulated by degradation of the substrate-specifying MMS19 protein and its downregulation is a common feature in cancer and is evolutionarily controlled.
cell.com
以上显示的是最相近的搜索结果。 查看全部搜索结果

Google学术搜索按钮

example.edu/paper.pdf
查找
获取 PDF 文件
引用
References