Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 34(36): 4758-66, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-25531325

ABSTRACT

Cancer cells gain a growth advantage through the so-called Warburg effect by shifting glucose metabolism from oxidative phosphorylation to aerobic glycolysis. Hypoxia-inducible factor 1 (HIF-1) has been suggested to function in metabolic reprogramming; however, the underlying mechanism has not been fully elucidated. We found that the aberrant expression of wild-type isocitrate dehydrogenase 3α (IDH3α), a subunit of the IDH3 heterotetramer, decreased α-ketoglutarate levels and increased the stability and transactivation activity of HIF-1α in cancer cells. The silencing of IDH3α significantly delayed tumor growth by suppressing the HIF-1-mediated Warburg effect and angiogenesis. IDH3α expression was associated with the poor postoperative overall survival of lung and breast cancer patients. These results justify the exploitation of IDH3 as a novel target for the diagnosis and treatment of cancers.


Subject(s)
Breast Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Isocitrate Dehydrogenase/biosynthesis , Lung Neoplasms/genetics , Neovascularization, Pathologic/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Lung Neoplasms/pathology , Mice , Neovascularization, Pathologic/pathology , Oxidative Phosphorylation
2.
Oncogene ; 32(16): 2058-68, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-22710721

ABSTRACT

Cancer patients often suffer from local tumor recurrence after radiation therapy. Some intracellular and extracellular factors, such as activity of hypoxia-inducible factor 1 (HIF-1), cell cycle status and oxygen availability, have been suggested to affect DNA damage responses and eventual radioresistant characteristics of cancer cells. But when, where, and how these factors affect one another and induce cellular radioresistance is largely unknown. Here, we analyzed mechanistic and spatio-temporal relationships among them in highly heterogeneous tumor microenvironments. Experiments in vitro demonstrated that a decrease in the glucose concentration reduced the transcriptional activity of HIF-1 and expression of a downstream gene for the cell cycle regulator p27(Kip1) even under hypoxic conditions. Then, the proportion of cells in the radioresistant S phase increased, whereas that in the radiosensitive G1 phase decreased, significantly. Immunohistochemical analyses showed that cancer cells in perinecrotic hypoxic regions, which should be under low-glucose conditions, expressed little HIF-1α, and therefore, were mainly in S phase and less damaged by radiation treatment. Continuous administration of glucagon, which increases the blood glucose concentration and so improves glucose availability in perinecrotic hypoxic regions, induced HIF-1α expression and increased radiation-induced DNA damage. Taken all together, these results indicate that cancer cells in perinecrotic regions, which would be under low-glucose and hypoxic conditions, obtain radioresistance by decreasing the level of both HIF-1 activity and p27(Kip1) expression, and adjusting their cell cycle to the radioresistant S phase.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , Animals , Cell Growth Processes/physiology , Cell Growth Processes/radiation effects , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , G1 Phase/genetics , G1 Phase/physiology , G1 Phase/radiation effects , HEK293 Cells , HeLa Cells , Humans , Hypoxia-Inducible Factor 1/genetics , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Radiation Tolerance , S Phase/genetics , S Phase/physiology , S Phase/radiation effects , Xenograft Model Antitumor Assays
3.
Br J Cancer ; 100(5): 747-57, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19223896

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) has been reported to promote tumour radioresistance; therefore, it is recognised as an excellent target during radiation therapy. However, the inhibition of HIF-1 in unsuitable timing can suppress rather than enhance the effect of radiation therapy because its anti-angiogenic effect increases the radioresistant hypoxic fraction. In this study, we imaged changes of HIF-1 activity after treatment with radiation and/or an HIF-1 inhibitor, YC-1, and optimised their combination. Hypoxic tumour cells were reoxygenated 6 h postirradiation, leading to von Hippel-Lindau (VHL)-dependent proteolysis of HIF-1alpha and a resultant decrease in HIF-1 activity. The activity then increased as HIF-1alpha accumulated in the reoxygenated regions 24 h postirradiation. Meanwhile, YC-1 temporarily but significantly suppressed HIF-1 activity, leading to a decrease in microvessel density and an increase in tumour hypoxia. On treatment with YC-1 and then radiation, the YC-1-mediated increase in tumour hypoxia suppressed the effect of radiation therapy, whereas on treatment in the reverse order, YC-1 suppressed the postirradiation upregulation of HIF-1 activity and consequently delayed tumour growth. These results indicate that treatment regimen determines whether an HIF-1 inhibitor enhances or inhibits the therapeutic effect of radiation, and the suppression of the postirradiation upregulation of HIF-1 activity is important for the best therapeutic benefit.


Subject(s)
Hypoxia-Inducible Factor 1/antagonists & inhibitors , Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/therapeutic use , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Furans/pharmacology , Furans/therapeutic use , HeLa Cells , Humans , Hypoxia-Inducible Factor 1/metabolism , Indazoles/pharmacology , Indazoles/therapeutic use , Mice , Mice, SCID , Neoplasms/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/radiation effects , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...