Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Oncol Rep ; 35(6): 3471-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27035858

ABSTRACT

As targets for cancer therapy, endoplasmic reticulum (ER) stress and autophagy are closely linked. However, the signaling pathways responsible for induction of autophagy in response to ER stress and its cellular consequences appear to vary with cell type and stimulus. In the present study, we showed that dithiothreitol (DTT) induced ER stress in HeLa cells in a time-Ā and dose-dependent fashion. With increased ER stress, reactive oxygen species (ROS) production increased and autophagy flux, assessed by intracellular accumulation of LC3B-II and p62, was inhibited. N-acetyl-L-cysteine (NAC), a classic antioxidant, exacerbated cell death induced by 3.2Ā mM of DTT, but attenuated that induced by 6.4Ā mM DTT. Low cytotoxic doses of DTT transiently activated c-JNU N-terminal kinase (JNK) and p38, whereas high dose of DTT persistently activated JNK and p38 and simultaneously reduced extracellular signal-regulated kinase (ERK) activity. Combined treatment with DTT and U0126, an inhibitor of ERK upstream activators mitogen-activated protein kinase (MAPK) kinaseĀ 1 and 2 (MEK1/2), blocked autophagy flux in HeLa cells. This effect was similar to that caused by a combination of DTT and chloroquine (CQ). These data suggested that insufficient autophagy was accompanied by increased ROS production during DTT-induced ER stress. ROS appeared to regulate MAPK signaling, switching from a pro-survival to a pro-apoptotic signal as ER stress increased. ERK inhibition by ROS during severe ER stress blocked autophagic flux. Impaired autophagic flux, in turn, aggravated ER stress, ultimately leading to cell death. Taken together, our data provide the first reported evidence that ROS may control cell fate through regulating the MAPK pathways and autophagic flux during DTT-induced ER/oxidative stress.


Subject(s)
Autophagy/drug effects , Dithiothreitol/pharmacology , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , HeLa Cells , Humans , p38 Mitogen-Activated Protein Kinases/physiology
2.
Int J Oncol ; 49(2): 773-84, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27277143

ABSTRACT

The Bcl-2 antiapoptotic proteins are important cancer therapy targets; however, their role in cancer cell metabolism remains unclear. We found that the BH3-only protein mimetic S1, a novel pan Bcl-2 inhibitor, simultaneously interrupted glucose metabolism and induced apoptosis in human SKOV3 ovarian cancer cells, which was related to the activation of SIRT3, a stress-responsive deacetylase. S1 interrupted the cellular glucose metabolism mainly through causing damage to mitochondrial respiration and inhibiting glycolysis. Moreover, S1 upregulated the gene and protein expression of SIRT3, and induced the translocation of SIRT3 from the nucleus to mitochondria. SIRT3 silencing reversed the effects of S1 on glucose metabolism and apoptosis through increasing the level of HK-II localized to the mitochondria, while a combination of the glycolysis inhibitor 2-DG and S1 intensified the cytotoxicity through further upregulation of SIRT3 expression. This study underscores an essential role of SIRT3 in the antitumor effect of Bcl-2 inhibitors in human ovarian cancer through regulating both metabolism and apoptosis. The manipulation of Bcl-2 inhibitors combined with the use of classic glycolysis inhibitors may be rational strategies to improve ovarian cancer therapy.


Subject(s)
Ovarian Neoplasms/drug therapy , Peptide Fragments/administration & dosage , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/administration & dosage , Sirtuin 3/biosynthesis , Apoptosis/drug effects , Biomimetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose/metabolism , Humans , Mitochondria/drug effects , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA, Small Interfering/genetics , Sirtuin 3/antagonists & inhibitors
3.
Oncol Rep ; 34(2): 913-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26081590

ABSTRACT

Sanguinarine (SAN), an alkaloid isolated from plants of the Papaveraceae family, is a compound with multiple biological activities. In the present study, we explored the anticancer properties of SAN in lung cancer using the human lung adenocarcinoma cell line SPC-A1. Our results revealed that SAN inhibited SPC-A1 cell growth and induced apoptosis in a dose-dependent manner. We found that SAN triggered reactive oxygen species (ROS) production, while elimination of ROS by N-acetylcysteine (NAC) reversed the growth inhibition and apoptosis induced by SAN. SAN-induced endoplasmic reticulum (ER) stress resulted in the upregulation of many genes and proteins involved in the unfolded protein response (UPR) pathway, including glucose-regulated protein 78 (GRP78), p-protein kinase R (PKR)-like ER kinase (PERK), p-eukaryotic translation initiation factor 2α (eIF2α), activating transcription factor 4 (ATF4) and CCAAT/enhancer binding protein homologous protein (CHOP). Blocking ER stress with tauroursodeoxycholic acid (TUDCA) markedly reduced SAN-induced inhibition of growth and apoptosis. Furthermore, TUDCA decreased SAN-induced ROS production, and NAC attenuated SAN-induced GRP78 and CHOP expression. Overall, our data indicate that the anticancer effects of SAN in lung cancer cells depend on ROS production and ER stress and that SAN may be a potential agent against lung cancer.


Subject(s)
Adenocarcinoma/metabolism , Benzophenanthridines/pharmacology , Endoplasmic Reticulum Stress , Isoquinolines/pharmacology , Lung Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma of Lung , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy
4.
Biomed Res Int ; 2014: 234370, 2014.
Article in English | MEDLINE | ID: mdl-25177684

ABSTRACT

Tumor cells overexpress antiapoptotic proteins of the Bcl-2 (B-cell leukemia/lymphoma-2) family, which can lead to both escape from cell death and resistance to chemotherapeutic drugs. Recent studies suggest that the endoplasmic reticulum (ER) can produce proapoptotic signals, amplifying the apoptotic signaling cascade. The crosstalk between mitochondria and ER plays a decisive role in many cellular events but especially in cell death. Bcl-2 family proteins located in the ER and mitochondria can influence not only the function of the two organelles but also the interaction between them. Therefore, the Bcl-2 family of proteins may also be involved in the mechanism of tumor chemotherapy resistance by influencing crosstalk between the ER and mitochondria. In this review we will briefly discuss evidence to support this concept.


Subject(s)
Drug Resistance, Neoplasm , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor Cross-Talk , Animals , Antineoplastic Agents/therapeutic use , Humans , Models, Biological , Stress, Physiological
5.
Oncol Rep ; 30(6): 2677-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100381

ABSTRACT

SKOV3/DDP human ovarian cancer cells have been shown to be resistant to cisplatin. Although the BH3 mimetic S1 induces cell death in several types of tumor cells, it is unclear whether it induces death in drug-resistant cells. Herein, we found that S1 induced endoplasmic reticulum (ER) stress-associated apoptosis in both SKOV3 and SKOV3/DDP cells. S1 activated autophagy at early time points in SKOV3/DDP cells, and inhibition of autophagy increased ER stress-associated apoptosis. Collectively, our data indicate that autophagy plays a protective role, but it cannot protect against S1-induced cell death in cisplatin-resistant SKOV3/DDP cells.


Subject(s)
Biomimetics , Drug Resistance, Neoplasm/drug effects , Endoplasmic Reticulum Stress/drug effects , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Female , Humans , Mitochondrial Membrane Transport Proteins , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/chemistry
6.
Cancer Lett ; 323(2): 180-7, 2012 Oct 28.
Article in English | MEDLINE | ID: mdl-22579788

ABSTRACT

Previous results showed that a novel BH3 mimetic S1 could induce cell death in a wide range of cancer types in vitro through Bax/Bak-dependent apoptosis. We demonstrated that in addition to mitochondrial pathway apoptosis, endoplasmic reticulum (ER) stress-associated apoptosis was also induced by S1. Moreover, S1 can induce autophagy in U251 cells, which may occur through ER stress and disruption of the association of Bcl-2 and Beclin 1. Inhibition of autophagy by the autophagic inhibitors 3-methyladenine (3-MA) or chloroquine (CQ) increased S1-induced apoptosis. In conclusion, autophagy plays an important role in S1-induced U251 cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Brain Neoplasms/pathology , Endoplasmic Reticulum/drug effects , Glioma/pathology , Membrane Proteins/metabolism , Molecular Mimicry , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/pharmacology , Beclin-1 , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Glioma/immunology , Glioma/metabolism , Humans , In Situ Nick-End Labeling , Microscopy, Confocal , Microscopy, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL