2015
DOI: 10.1074/jbc.m115.654913
|View full text |Cite
|
Sign up to set email alerts
|

Mitochondrial Respiratory Dysfunction Induces Claudin-1 Expression via Reactive Oxygen Species-mediated Heat Shock Factor 1 Activation, Leading to Hepatoma Cell Invasiveness

Abstract: Background: Increased claudin-1 (Cln-1) expression in human hepatoma cells with mitochondrial defects leads to high tumor cell invasiveness. Results: Cln-1-mediated hepatoma cell invasiveness occurs via heat shock factor 1 (HSF1) activation. Conclusion:The mitochondrial defect-ROS-HSF1 axis controls hepatoma cell invasiveness. Significance: HSF1 is a key mitochondrial retrograde-responsive transcription factor controlling hepatoma cell invasiveness.

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

1
10
0

Year Published

2017
2017
2023
2023

Publication Types

Select...
5
2
1

Relationship

0
8

Authors

Journals

citations
Cited by 18 publications
(11 citation statements)
references
References 47 publications
(49 reference statements)
1
10
0
Order By: Relevance
“…For example, Ma et al ( 67 ) described that clones of breast cancer cells generated by treatment with rotenone, exhibited mitochondrial respiratory defects, increased ROS levels, and high migration and invasion properties, which were inhibited by treatment with antioxidants such as NAC and mito-TEMPO, a mitochondria-targeted antioxidant ( 67 ). Similar effects have also been observed in hepatoma cells ( 66 ). Altogether, these data suggest that the inhibition of Complex I activity accompanied by ROS generation promotes migration, invasion, and metastasis.…”
Section: Role Of Complex I In Metastasis Of Cancer Cellssupporting
confidence: 81%
See 1 more Smart Citation
“…For example, Ma et al ( 67 ) described that clones of breast cancer cells generated by treatment with rotenone, exhibited mitochondrial respiratory defects, increased ROS levels, and high migration and invasion properties, which were inhibited by treatment with antioxidants such as NAC and mito-TEMPO, a mitochondria-targeted antioxidant ( 67 ). Similar effects have also been observed in hepatoma cells ( 66 ). Altogether, these data suggest that the inhibition of Complex I activity accompanied by ROS generation promotes migration, invasion, and metastasis.…”
Section: Role Of Complex I In Metastasis Of Cancer Cellssupporting
confidence: 81%
“…As mutations in mtDNA represent an early event during breast tumorigenesis, producing defective OXPHOS with a metabolic shift toward glycolysis could be used as a potential biomarker for early detection and prognosis ( 65 ). In further support, several reports indicate that the inhibition of Complex I activity by pharmacologic interventions using small molecules can increase ROS generation, promoting the migration and invasion of cancer cells ( 62 , 66 , 67 ). For example, Ma et al ( 67 ) described that clones of breast cancer cells generated by treatment with rotenone, exhibited mitochondrial respiratory defects, increased ROS levels, and high migration and invasion properties, which were inhibited by treatment with antioxidants such as NAC and mito-TEMPO, a mitochondria-targeted antioxidant ( 67 ).…”
Section: Role Of Complex I In Metastasis Of Cancer Cellsmentioning
confidence: 71%
“…However, from our knowledge, no evidence has been previously reported after Amph exposure regarding sensitized oxidative/inflammatory responses in brain microvessels. Furthermore, there is an established link between elevated ROS levels and further stimulating HSP70 synthesis as a protective response to apoptotic signaling (Horowitz & Robinson, ; Lee et al, ). Additionally, we found no sensitized response to the Amph challenge in brain microvessels obtained from animals pretreated with AT 1 ‐R blocker.…”
Section: Discussionmentioning
confidence: 99%
“…For instance, HSF1 has been shown to upregulate β-catenin via HuR [63] and loss of HSF1 reduced NFκB activation and suppressed tumor growth [16]. HSF1 activation in hepatoma cells led to HSF1-driven Cln-1 expression leading to cell invasion [64]. Loss of HSF1 in a lung tumor model also showed a decrease in migration associated with a decrease in vinculin, an actin-binding protein known to promote cell migration [60].…”
Section: Role Of Hsf1 In Tumor Progressionmentioning
confidence: 99%