Expression, regulation and clinical relevance of the ATPase inhibitory factor 1 in human cancers

M Sánchez-Aragó, L Formentini, I Martínez-Reyes… - Oncogenesis, 2013 - nature.com
M Sánchez-Aragó, L Formentini, I Martínez-Reyes, J García-Bermudez, F Santacatterina…
Oncogenesis, 2013nature.com
Recent findings in colon cancer cells indicate that inhibition of the mitochondrial H+-
adenosine triphosphate (ATP) synthase by the ATPase inhibitory factor 1 (IF1) promotes
aerobic glycolysis and a reactive oxygen species (ROS)-mediated signal that enhances
proliferation and cell survival. Herein, we have studied the expression, biological relevance,
mechanism of regulation and potential clinical impact of IF1 in some prevalent human
carcinomas. We show that IF1 is highly overexpressed in most (> 90%) of the colon (n= 64) …
Abstract
Recent findings in colon cancer cells indicate that inhibition of the mitochondrial H+-adenosine triphosphate (ATP) synthase by the ATPase inhibitory factor 1 (IF1) promotes aerobic glycolysis and a reactive oxygen species (ROS)-mediated signal that enhances proliferation and cell survival. Herein, we have studied the expression, biological relevance, mechanism of regulation and potential clinical impact of IF1 in some prevalent human carcinomas. We show that IF1 is highly overexpressed in most (> 90%) of the colon (n= 64), lung (n= 30), breast (n= 129) and ovarian (n= 10) carcinomas studied as assessed by different approaches in independent cohorts of cancer patients. The expression of IF1 in the corresponding normal tissues is negligible. By contrast, the endometrium, stomach and kidney show high expression of IF1 in the normal tissue revealing subtle differences by carcinogenesis. The overexpression of IF1 also promotes the activation of aerobic glycolysis and a concurrent ROS signal in mitochondria of the lung, breast and ovarian cancer cells mimicking the activity of oligomycin. IF1-mediated ROS signaling activates cell-type specific adaptive responses aimed at preventing death in these cell lines. Remarkably, regulation of IF1 expression in the colon, lung, breast and ovarian carcinomas is exerted at post-transcriptional levels. We demonstrate that IF1 is a short-lived protein (t 1/2∼ 100 min) strongly implicating translation and/or protein stabilization as main drivers of metabolic reprogramming and cell survival in these human cancers. Analysis of tumor expression of IF1 in cohorts of breast and colon cancer patients revealed its relevance as a predictive marker for clinical outcome, emphasizing the high potential of IF1 as therapeutic target.
nature.com