[HTML][HTML] Time-dependent stabilization of hypoxia inducible factor-1α by different intracellular sources of reactive oxygen species

M Calvani, G Comito, E Giannoni, P Chiarugi - PloS one, 2012 - journals.plos.org
M Calvani, G Comito, E Giannoni, P Chiarugi
PloS one, 2012journals.plos.org
Intratumoral hypoxia is a major obstacle in the development of effective cancer
chemotherapy, decreasing the efficacy of anti-neoplastic drugs in several solid tumours. The
hypoxic environment, through its master regulator hypoxia inducible factor-1 (HIF-1), is able
to maintain an anti-apoptotic potential through activation of critical genes associated with
drug resistance. Besides affecting metabolism and motility of tumour cells, hypoxia also
paradoxically increases production of reactive oxygen species (ROS), which contribute to …
Intratumoral hypoxia is a major obstacle in the development of effective cancer chemotherapy, decreasing the efficacy of anti-neoplastic drugs in several solid tumours. The hypoxic environment, through its master regulator hypoxia inducible factor-1 (HIF-1), is able to maintain an anti-apoptotic potential through activation of critical genes associated with drug resistance. Besides affecting metabolism and motility of tumour cells, hypoxia also paradoxically increases production of reactive oxygen species (ROS), which contribute to stabilize HIF-1 through a redox-mediated inhibition of its proteolysis. Here we reported that 1% O2 hypoxia increases the resistance of human metastatic melanoma cells to conventional chemotherapy with etoposide, and that the increase in chemoresistance strongly depends on ROS delivery due to hypoxia. We reported a biphasic redox-dependent role of HIF-1, involving mitochondrial complex III and NADPH oxidase as oxidants sources, synergising in enhancing survival to chemotherapy. The feed-forward loop engaged by hypoxia involves first an HIF-1-dependent vascular endothelial growth factor-A (VEGF-A) autocrine production and, in the later phase, activation of NADPH oxidase from VEGF/VEGFR2 interaction, finally leading to a further redox-dependent long lasting stabilization of HIF-1. We therefore identified a redox-dependent circuitry linking hypoxia-driven ROS to VEGF-A secretion and to enhanced melanoma cell survival to etoposide chemotherapy.
PLOS