2020
DOI: 10.15252/embr.202050827
|View full text |Cite
|
Sign up to set email alerts
|

OMA1 reprograms metabolism under hypoxia to promote colorectal cancer development

Abstract: Many cancer cells maintain enhanced aerobic glycolysis due to irreversible defective mitochondrial oxidative phosphorylation (OXPHOS). This phenomenon, known as the Warburg effect, is recently challenged because most cancer cells maintain OXPHOS. However, how cancer cells coordinate glycolysis and OXPHOS remains largely unknown. Here, we demonstrate that OMA1, a stress-activated mitochondrial protease, promotes colorectal cancer development by driving metabolic reprogramming. OMA1 knockout suppresses colorecta… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

1
45
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
8
1

Relationship

0
9

Authors

Journals

citations
Cited by 88 publications
(58 citation statements)
references
References 65 publications
1
45
0
Order By: Relevance
“…Under hypoxia, cancer cells could retain survival and stimulate tumor proliferation, migration, and invasion via glycolysis ( Huang and Zong, 2017 ). Wu et al reported that OMA, an ATP-dependent zinc metalloprotease, promotes cancer development by reprogramming metabolic colorectal cancer under hypoxia ( Wu et al, 2021b ). A recent study revealed that hypoxia downregulates the enzymes of pentose phosphate pathway and upregulates the enzymes of glycolysis in tumor and then stimulate tumor migration while inhibiting tumor proliferation ( Kathagen-Buhmann et al, 2016 ).…”
Section: Discussionmentioning
confidence: 99%
“…Under hypoxia, cancer cells could retain survival and stimulate tumor proliferation, migration, and invasion via glycolysis ( Huang and Zong, 2017 ). Wu et al reported that OMA, an ATP-dependent zinc metalloprotease, promotes cancer development by reprogramming metabolic colorectal cancer under hypoxia ( Wu et al, 2021b ). A recent study revealed that hypoxia downregulates the enzymes of pentose phosphate pathway and upregulates the enzymes of glycolysis in tumor and then stimulate tumor migration while inhibiting tumor proliferation ( Kathagen-Buhmann et al, 2016 ).…”
Section: Discussionmentioning
confidence: 99%
“…For example, cancer survival correlated with OMA1 gene expression levels in multiple studies. 9,58,59 The protooncogene MYC could thereby be responsible for OMA1's differential expression in tumors. 9 Another study placed OMA1 downstream of tumor protein p53.…”
Section: Discussionmentioning
confidence: 99%
“…On the contrary, OMA1 knockout is known to suppress CRC development. Upon activation by hypoxia, OMA1 increases mitochondrial ROS to stabilize HIF-1α, thus promoting glycolysis and suppressing OXPHOS in CRC cells [ 95 ]. These results suggest the crucial role of OMA1 in HIF-1α-mediated CRC development and a high potential as a target for CRC therapy.…”
Section: Associated Mitochondrial Mutations and Dysfunctionsmentioning
confidence: 99%