Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
2.
Free Radic Biol Med ; 174: 157-170, 2021 10.
Article in English | MEDLINE | ID: mdl-34403740

ABSTRACT

Tumor hypoxia is a major biological factor that drives resistance to chemotherapy and radiotherapy. We previously demonstrated that the pro-oxidative drug dihydroartemisinin (DHA) efficiently targeted normoxic and hypoxic cancer cells. Although well studied in normoxia, the mechanism behind DHA-mediated cytotoxicity in hypoxia is insufficiently explored. Here, we analyzed the effect of DHA in HCT116 wild type (wt) cells and in HCT116 Bax-/-Baksh cells with a defective intrinsic apoptosis pathway. Normoxic HCT116 wt cells underwent apoptosis shortly after treatment with DHA. Autophagy-associated cell death contributes to short-term cytotoxicity of DHA in normoxia. These cells switched to an apoptosis- and autophagy-independent cell death after treatment with DHA in hypoxia and displayed similar long-term survival in response to DHA in normoxia and hypoxia. In HCT116 Bax-/-Baksh cells, DHA induced cell cycle arrest shortly after treatment irrespective of oxygen levels. Later, HCT116 Bax-/-Baksh cells induced a delayed cell death after treatment with DHA in hypoxia followed by return to normoxia, while treatment with DHA in normoxia was hardly toxic. We identified lower glutathione levels in hypoxic HCT116 cells which correlated with higher lipid peroxidation after treatment with DHA. Moreover, insufficient expression of Bax/Bak counteracted hypoxia-mediated downregulation of mitochondrial function, thereby adding to DHA-induced ROS production and lipid peroxidation in hypoxia. In summary, DHA-mediated cytotoxicity in normoxia depended on Bax/Bak expression, while cytotoxicity after treatment with DHA in hypoxia was regulated independently of Bax/Bak in HCT116 colorectal cancer cells.


Subject(s)
Apoptosis , Colorectal Neoplasms , Artemisinins , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , HCT116 Cells , Humans , Hypoxia , bcl-2-Associated X Protein/genetics
3.
Cell Death Dis ; 12(7): 694, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257274

ABSTRACT

Hypoxia, a characteristic of most human solid tumors, is a major obstacle to successful radiotherapy. While moderate acute hypoxia increases cell survival, chronic cycling hypoxia triggers adaptation processes, leading to the clonal selection of hypoxia-tolerant, apoptosis-resistant cancer cells. Our results demonstrate that exposure to acute and adaptation to chronic cycling hypoxia alters the balance of Bcl-2 family proteins in favor of anti-apoptotic family members, thereby elevating the apoptotic threshold and attenuating the success of radiotherapy. Of note, inhibition of Bcl-2 and Bcl-xL by BH3-mimetic ABT-263 enhanced the sensitivity of HCT116 colon cancer and NCI-H460 lung cancer cells to the cytotoxic action of ionizing radiation. Importantly, we observed this effect not only in normoxia, but also in severe hypoxia to a similar or even higher extent. ABT-263 furthermore enhanced the response of xenograft tumors of control and hypoxia-selected NCI-H460 cells to radiotherapy, thereby confirming the beneficial effect of combined treatment in vivo. Targeting the Bcl-2 rheostat with ABT-263, therefore, is a particularly promising approach to overcome radioresistance of cancer cells exposed to acute or chronic hypoxia with intermittent reoxygenation. Moreover, we found intrinsic as well as ABT-263- and irradiation-induced regulation of Bcl-2 family members to determine therapy sensitivity. In this context, we identified Mcl-1 as a resistance factor that interfered with apoptosis induction by ABT-263, ionizing radiation, and combinatorial treatment. Collectively, our findings provide novel insights into the molecular determinants of hypoxia-mediated resistance to apoptosis and radiotherapy and a rationale for future therapies of hypoxic and hypoxia-selected tumor cell fractions.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis , Colonic Neoplasms/radiotherapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrolides/metabolism , Mice, Nude , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Tumor Burden/drug effects , Tumor Burden/radiation effects , Tumor Hypoxia , Tumor Microenvironment , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
4.
Cancer Lett ; 439: 24-38, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30205167

ABSTRACT

High tumor heterogeneity and increased therapy resistance acquired in a hypoxic tumor microenvironment remain major obstacles to successful radiotherapy. Others and we have shown that adaptation of cancer cells to cycling severe hypoxia and intermittent reoxygenation stress (chronic-cycling hypoxia) increases cellular antioxidant capacity thereby supporting resistance to chemotherapy and radiotherapy. Here we explored the involvement of antioxidant-associated mitochondrial transport-systems for maintenance of redox-homeostasis in adaptation to chronic-cycling hypoxia and associated radioresistance. Genetic or pharmacological inhibition of the mitochondrial dicarboxylate carrier (SLC25A10) or the oxoglutarate-carrier (SLC25A11) increased the cytotoxic effects of ionizing radiation (IR). But only targeting of SLC25A10 was effective in overcoming chronic-cycling hypoxia-induced enhanced death resistance in vitro and in vivo by disturbing increased antioxidant capacity. Furthermore, in silico analysis revealed that overexpression of SLC25A10 but not SLC25A11 is associated with reduced overall survival in lung- and breast-cancer patients. Our study reveals a role of SLC25A10 in supporting both, redox- and energy-homeostasis, ensuring radioresistance of cancer cells with tolerance to chronic-cycling hypoxia thereby proposing a novel strategy to overcome a mechanism of hypoxia-induced therapy resistance with potential clinical relevance regarding decreased patient survival.


Subject(s)
Antioxidants/metabolism , Dicarboxylic Acid Transporters/genetics , Neoplasms/radiotherapy , RNA Interference , Radiation, Ionizing , Xenograft Model Antitumor Assays , Animals , Cell Hypoxia , Cell Line, Tumor , Dicarboxylic Acid Transporters/metabolism , Humans , Hypoxia , Kaplan-Meier Estimate , Malonates/pharmacology , Malonates/therapeutic use , Mice, Nude , Neoplasms/drug therapy , Neoplasms/genetics , Radiation Tolerance/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...