Regulation of hypoxia-inducible factor 1α expression and function by the mammalian target of rapamycin

CC Hudson, M Liu, GG Chiang… - … and cellular biology, 2002 - Taylor & Francis
CC Hudson, M Liu, GG Chiang, DM Otterness, DC Loomis, F Kaper, AJ Giaccia…
Molecular and cellular biology, 2002Taylor & Francis
Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric transcription factor containing an
inducibly expressed HIF-1α subunit and a constititutively expressed HIF-1β subunit. Under
hypoxic conditions, the HIF-1α subunit accumulates due to a decrease in the rate of
proteolytic degradation, and the resulting HIF-1α-HIF-1β heterodimers undergo
posttranslational modifications that promote transactivation. Recent studies suggest that
amplified signaling through phosphoinositide 3-kinase, and its downstream target, mTOR …
Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric transcription factor containing an inducibly expressed HIF-1α subunit and a constititutively expressed HIF-1β subunit. Under hypoxic conditions, the HIF-1α subunit accumulates due to a decrease in the rate of proteolytic degradation, and the resulting HIF-1α-HIF-1β heterodimers undergo posttranslational modifications that promote transactivation. Recent studies suggest that amplified signaling through phosphoinositide 3-kinase, and its downstream target, mTOR, enhances HIF-1-dependent gene expression in certain cell types. In the present study, we have explored further the linkage between mTOR and HIF-1 in PC-3 prostate cancer cells treated with hypoxia or the hypoxia mimetic agent, CoCl2. Pretreatment of PC-3 cells with the mTOR inhibitor, rapamycin, inhibited both the accumulation of HIF-1α and HIF-1-dependent transcription induced by hypoxia or CoCl2. Transfection of these cells with wild-type mTOR enhanced HIF-1 activation by hypoxia or CoCl2, while expression of a rapamycin-resistant mTOR mutant rendered both HIF-1α stabilization and HIF-1 transactivating function refractory to inhibition by rapamycin. Studies with GAL4-HIF-1α fusion proteins pinpointed the oxygen-dependent degradation domain as a critical target for the rapamycin-sensitive, mTOR-dependent signaling pathway leading to HIF-1α stabilization by CoCl2. These studies position mTOR as an upstream activator of HIF-1 function in cancer cells and suggest that the antitumor activity of rapamycin is mediated, in part, through the inhibition of cellular responses to hypoxic stress.
Taylor & Francis Online