Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Theranostics ; 10(4): 1833-1848, 2020.
Article in English | MEDLINE | ID: mdl-32042339

ABSTRACT

Purpose: To determine the role of UCH-L1 in regulating ERα expression, and to evaluate whether therapeutic targeting of UCH-L1 can enhance the efficacy of anti-estrogen therapy against breast cancer with loss or reduction of ERα. Methods: Expressions of UCH-L1 and ERα were examined in breast cancer cells and patient specimens. The associations between UCH-L1 and ERα, therapeutic response and prognosis in breast cancer patients were analyzed using multiple databases. The molecular pathways by which UCH-L1 regulates ERα were analyzed using immunoblotting, qRT-PCR, immunoprecipitation, ubiquitination, luciferase and ChIP assays. The effects of UCH-L1 inhibition on the efficacy of tamoxifen in ERα (-) breast cancer cells were tested both in vivo and in vitro. Results: UCH-L1 expression was conversely correlated with ERα status in breast cancer, and the negative regulatory effect of UCH-L1 on ERα was mediated by the deubiquitinase-mediated stability of EGFR, which suppresses ERα transcription. High expression of UCH-L1 was associated with poor therapeutic response and prognosis in patients with breast cancer. Up-regulation of ERα caused by UCH-L1 inhibition could significantly enhance the efficacy of tamoxifen and fulvestrant in ERα (-) breast cancer both in vivo and in vitro. Conclusions: Our results reveal an important role of UCH-L1 in modulating ERα status and demonstrate the involvement of UCH-L1-EGFR signaling pathway, suggesting that UCH-L1 may serve as a novel adjuvant target for treatment of hormone therapy-insensitive breast cancers. Targeting UCH-L1 to sensitize ER negative breast cancer to anti-estrogen therapy might represent a new therapeutic strategy that warrants further exploration.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/genetics , Ubiquitin Thiolesterase/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Down-Regulation/drug effects , ErbB Receptors/metabolism , Estrogen Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , Humans , Mice , Mice, Nude , Tamoxifen/therapeutic use , Ubiquitin Thiolesterase/metabolism , Up-Regulation/drug effects
2.
Curr Top Med Chem ; 19(23): 2143-2157, 2019.
Article in English | MEDLINE | ID: mdl-31456522

ABSTRACT

KRAS is a member of the murine sarcoma virus oncogene-RAS gene family. It plays an important role in the prevention, diagnosis and treatment of tumors during tumor cell growth and angiogenesis. KRAS is the most commonly mutated oncogene in human cancers, such as pancreatic cancers, colon cancers, and lung cancers. Detection of KRAS gene mutation is an important indicator for tracking the status of oncogenes, highlighting the developmental prognosis of various cancers, and the efficacy of radiotherapy and chemotherapy. However, the efficacy of different patients in clinical treatment is not the same. Since RNA interference (RNAi) technologies can specifically eliminate the expression of specific genes, these technologies have been widely used in the field of gene therapy for exploring gene function, infectious diseases and malignant tumors. RNAi refers to the phenomenon of highly specific degradation of homologous mRNA induced by double-stranded RNA (dsRNA), which is highly conserved during evolution. There are three classical RNAi technologies, including siRNA, shRNA and CRISPR-Cas9 system, and a novel synthetic lethal interaction that selectively targets KRAS mutant cancers. Therefore, the implementation of individualized targeted drug therapy has become the best choice for doctors and patients. Thus, this review focuses on the current status, future perspective and associated challenges in silencing of KRAS with RNAi technology.


Subject(s)
Biotechnology , Neoplasms/genetics , Neoplasms/therapy , Oncogene Protein p21(ras)/genetics , RNA Interference , Animals , Humans , Mutation , Neoplasms/pathology , Oncogene Protein p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics
3.
Int Immunopharmacol ; 17(3): 548-55, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23941769

ABSTRACT

Autophagy and necroptosis have been known to be interconnected, while the relationship between autophagy and necroptosis remains unclear. Here, we demonstrated that pan-caspase inhibitor z-VAD-fmk (zVAD) exacerbated TNFα-induced necroptosis and autophagy in murine fibrosarcoma L929 cells. And the RIP-1 inhibitor necrostatin-1 inhibited TNFα+zVAD-induced necroptosis and autophagy. Inhibition of autophagy by 3-methyladenine (3MA) or small interfering RNA (siRNA) against Beclin 1 augmented TNFα-induced necroptosis, while, autophagy inhibition did not influence TNFα+zVAD-induced necroptosis. These results suggested that autophagy was a downstream consequence of necroptosis, and had a negative-feedback function to necroptosis in TNFα-treated L929 cells, but not in the presence of zVAD. Subsequently, TNFα administration was accompanied with caspase-6 activation. Inhibition of caspase-6 activity by z-V-E(OMe)-I-D(OMe)-fmk (zVEID) or caspase-6 (p20) siRNA had no effect on necroptosis but promoted TNFα-induced autophagy. Meanwhile, autophagy inhibition further increased caspase-6 activation. Caspase-6 (p20) siRNA sequestered the increased necroptotic ratio by 3MA pretreatment in TNFα-treated L929 cells. In addition, caspase-6 activation induced by TNFα administration was inhibited by zVAD. Further, autophagy induced by higher concentration of zVAD did not negatively regulate necroptosis because caspase-6 was not activated. Collectively, our data indicated that autophagy was a downstream consequence of necroptosis, and negatively regulated necroptosis when caspase-6 was activated in TNFα-treated L929 cells.


Subject(s)
Caspase 6/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors/pharmacology , Cell Death/physiology , Imidazoles/pharmacology , Indoles/pharmacology , L Cells/drug effects , Mice , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
4.
Int Immunopharmacol ; 14(4): 674-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23000518

ABSTRACT

Tumor necrosis factor alpha (TNFα) induces necroptosis and autophagy; however, the detailed molecular mechanism is not fully understood. In this study, we found that TNFα administration caused mitochondrial dysfunction and reactive oxygen species (ROS) production, which led to necroptosis and autophagy in murine fibrosarcoma L929 cells. Notably, the RIP1 (serine-threonine kinase receptor-interacting protein 1, a main adaptor protein of necroptosis) specific inhibitor necrostatin-1 (Nec-1) recovered mitochondrial dysfunction and ROS production due to TNFα administration. Moreover, pan-caspase inhibitor z-VAD-fmk (zVAD) increased RIP1 expression and exacerbated TNFα-induced mitochondrial dysfunction and ROS production, indicating that RIP1 led to mitochondrial dysfunction and ROS production. In addition, cytochrome c release from mitochondria was accompanied with TNFα administration, and Nec-1 blocked the release of cytochrome c upon TNFα administration, while zVAD enhanced the release. These further suggested that RIP1 induced mitochondrial dysfunction accompanied with cytochrome c release. Furthermore, autophagy inhibitor 3-methyladenine (3MA) did not affect RIP1 expression as well as mitochondrial dysfunction and ROS production. Together with our previous publication that autophagy was a downstream consequence of necroptosis, we concluded that TNFα induced mitochondrial dysfunction accompanied with ROS production and cytochrome c release via RIP1, leading to necroptosis and resulting autophagic cell death.


Subject(s)
Fibroblasts/drug effects , Fibroblasts/physiology , GTPase-Activating Proteins/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antimycin A , Autophagy , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , GTPase-Activating Proteins/genetics , Gene Expression Regulation/drug effects , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Rotenone
5.
Free Radic Res ; 46(11): 1393-405, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22881126

ABSTRACT

Oridonin, a diterpenoid compound, extracted and purified from Rabdosia rubescen has been reported to have cytotoxic effect on tumour cells through apoptosis, and tyrosine kinase pathways are involved in these processes. A specific epidermal growth factor receptor (EGFR) inhibitor AG1478 was used to examine the relationship between EGFR signal pathways and oridonin-induced apoptosis and autophagy in EGFR abundant human epidermoid carcinoma A431 cells. Inhibition of EGFRaugmented oridonin-induced A431 cell apoptosis, while the changes of expression of downstream proteins, Bcl-2, Bcl-xL, Bax, cytochrome c, pro-caspase-3, Fas, FADD and pro-caspase-8 suggested that both the intrinsic and extrinsic apoptotic pathways are involved in these processes. Pretreatment with AG1478 aggravated oridonin-induced loss of mitochondrial membrane potential (MMP) and increased ROS generation in A431 cells, while a ROS scavenger, N-acetylcysteine (NAC) completely reversed oridonin- and AG1478-induced ROS generation and apoptosis. Therefore, AG1478 augmented oridonin-induced apoptosis by enhancing oxidative stress. Pretreatment with AG1478 decreased the expression of downstream MAPK proteins ERK, JNK and P38 and their phosphorylated forms to varying degrees compared with oridonin alone treatment. Then after administration of ERK, JNK and P38 inhibitors, only JNK inhibitor SP600125 effectively augmented oridonin-induced apoptosis and ROS generation. Therefore, in EGFR downstream pathways, JNK played a major role in preventing oridonin-induced apoptosis. Autophagy antagonised apoptosis and exerted a protective effect in A431 cells, and both AG1478 and SP600125 decreased oridonin-induced autophagy. Inhibition of EGFR augmented oridonin-induced apoptosis and this was caused by enhanced oxidative stress, and JNK played a major protective role by increasing autophagy, leading to antagonising apoptosis and ROS generation.


Subject(s)
Apoptosis/drug effects , Diterpenes, Kaurane/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Oxidative Stress/drug effects , Quinazolines/pharmacology , Tyrphostins/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology
6.
Acta Pharmacol Sin ; 33(8): 1055-61, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22842735

ABSTRACT

AIM: To investigate the role of nitric oxide (NO) in oridonin-induced apoptosis and autophagy in murine fibrosarcoma L929 cells and the underlying molecular mechanisms. METHODS: Cell viability was measured using MTT assay. Intracellular NO level, SubG(1) cell ratio and autophagy cell ratios were analyzed with flow cytometry after diaminofluorescein-2 diacetate (DAF-2DA), propidium iodide (PI) and monodansylcadaverine (MDC) staining, respectively. Protein expression was examined using Western blot analysis. RESULTS: Exposure of L929 cells to oridonin (50 µmol/L) for 24 h led to intracellular NO production. Pretreatment with NOS inhibitor 1400w or L-NAME inhibited oridonin-induced apoptosis and autophagy in L929 cells. The pretreatment decreased the apoptosis-related protein Bax translocation and cytochrome c release, increased Bcl-2 level, reversed the autophagy-associated protein Beclin 1 increase and conversion of LC3 I to LC3 II. Furthermore, pretreatment with NO scavenger DTT completely inhibited oridonin-induced apoptosis and autophagy in L929 cells. In addition, oridonin (50 µmol/L) activated ERK and p53 in L929 cells, and the interruption of ERK and p53 activation by PD 98059, pifithrin-α, or ERK siRNA decreased oridonin-induced apoptosis and autophagy. The inhibition of NO production reduced oridonin-induced ERK and p53 activation, and NO production was down-regulated by blocking ERK and p53 activation. CONCLUSION: NO played a pivotal role in oridonin-induced apoptosis and autophagy in L929 cells. Taken together with our previous finding that ERK contributes to p53 activation, it appears that NO, ERK, and p53 form a positive feedback loop. Consequently, we suggest that oridonin-induced apoptosis and autophagy are modulated by the NO-ERK-p53 molecular signaling mechanism in L929 cells.


Subject(s)
Diterpenes, Kaurane/pharmacology , Feedback, Physiological/physiology , Fibrosarcoma/metabolism , Genes, p53/physiology , MAP Kinase Signaling System/physiology , Nitric Oxide/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Autophagy/drug effects , Autophagy/physiology , Cell Line, Tumor , Diterpenes, Kaurane/therapeutic use , Feedback, Physiological/drug effects , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , MAP Kinase Signaling System/drug effects , Mice
7.
J Pharmacol Sci ; 119(2): 150-9, 2012.
Article in English | MEDLINE | ID: mdl-22673163

ABSTRACT

The pharmacological activity of SU11274 is primarily due to its inhibition of hepotocyte growth factor receptor (c-Met) kinase overexpression. In this study, we demonstrated that the pathway involved in SU11274-induced autophagy was presumably through inhibition of c-Met and its down-stream pathways, including phosphatidylinositol 3-kinases ­ Akt (PI3K­Akt) and the growth factor receptor bound protein-2 / son of sevenless ­ Ras ­ p38 MAPK (Grb2/SOS­Ras­p38) pathway. SU11274 time-dependently induced the generation of superoxide anion (O2(•−)) and hydrogen peroxide (H2O2). There is a negative feedback loop between reactive oxygen species (ROS) induction and SU11274. Then, we investigated the role of ROS in protecting cells against SU11274-induced autophagic cell death in A549 cells. O2(•−) and H2O2 generation activated c-Met­PI3K­Akt and c-Met­Grb2/SOS­Ras­p38 signaling pathways, which were suppressed by O2(•−) scavenger superoxide dismutase (SOD) and H2O2 scavenger catalase. In conclusion, O2(•−) and H2O2 evoked cell resistance to SU11274 via activating c-Met­PI3K­Akt and c-Met­Grb2/SOS­Ras­p38 pathways in A549 cells. SU11274 also induced ROS generation in Caenorhabditis elegans.


Subject(s)
Antineoplastic Agents/pharmacology , Hydrogen Peroxide/metabolism , Indoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Sulfonamides/pharmacology , Superoxides/metabolism , Animals , Autophagy/drug effects , Caenorhabditis elegans , Catalase/metabolism , Cell Line, Tumor , Cell Survival/drug effects , GRB2 Adaptor Protein/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Son of Sevenless Protein, Drosophila/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism
8.
J Pharmacol Sci ; 118(4): 423-32, 2012.
Article in English | MEDLINE | ID: mdl-22466960

ABSTRACT

SU11274, a small molecule inhibitor of c-Met, was reported to induce apoptosis in human non-small-cell lung cancer (NSCLC) cells. However, SU11274-mediated autophagy in NSCLC cells has rarely been reported. The aim of this study was to elucidate the molecular mechanisms mediating SU11274-induced autophagy in NSCLC A549 cells. Here we reported that SU11274-induced autophagy was accompanied with an increase in the conversion of LC3-I to LC3-II and up-regulation of Beclin-1 expression. Subsequently, we also found that small interfering RNA against c-Met induced A549 cell autophagy while promotion of c-Met by hepatocyte growth factor (HGF) suppressed A549 cell autophagy. Inhibition of autophagy by 3-methyladenine (3-MA) suppressed SU11274-induced cell death, suggesting that SU11274-induced autophagy caused cell death. Further study showed that ERK and p53 were activated after SU11274 treatment. Interruption of ERK and p53 activities decreased SU11274-induced autophagy, and blocking of ERK by the specific inhibitor PD98059 suppressed SU11274-induced p53 activation. Moreover, ERK activation upregulated Beclin-1 expression through induction of Bcl-2 phosphorylation, but p53 did not induce Bcl-2 phosphorylation. In conclusion, inhibition of c-Met induced autophagic cell death, which was associated with ERK-p53 activation and ERK-mediated Bcl-2 phosphorylation in A549 cells.


Subject(s)
Autophagy/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Indoles/pharmacology , Lung Neoplasms/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/metabolism , Autophagy/physiology , Cell Line, Tumor , Enzyme Activation/physiology , Humans , Lung Neoplasms/enzymology , Phosphorylation/drug effects , Phosphorylation/physiology , Receptor Protein-Tyrosine Kinases/physiology
9.
J Pharmacol Sci ; 117(3): 160-9, 2011.
Article in English | MEDLINE | ID: mdl-22027097

ABSTRACT

Tumor necrosis factor alpha (TNFα) has been reported to induce necroptosis and autophagy, but its mechanisms remain unclear. In this study, we found that TNFα significantly induced necroptosis and autophagy in murine fibrosarcoma L929 cells. The necroptosis inhibitor necrostatin-1 (Nec-1) completely blocked TNFα-induced necroptosis and autophagy, but inhibition of autophagy with 3-methyladenine (3MA) or Beclin 1 small interfering RNA (siRNA) promoted necroptosis, indicating that autophagy acted as a negative regulator of TNFα-induced necroptosis. The cytotoxicity of TNFα was accompanied by decreased expressions of phosphorylated p38 mitogen-activated protein kinase (p-p38) and nuclear factor-kappa B (NF-κB), and inhibition of p38 and NF-κB activation by chemical inhibitors or siRNA augmented these necroptotic and autophagic responses to TNFα in the cells. The pan-caspase inhibitor z-VAD-fmk (zVAD) exacerbated TNFα-induced necroptosis and autophagy. Combined treatment with TNFα and zVAD further decreased the expressions of p-p38 and NF-κB compared with TNFα alone treatment. Consequently, these results indicated that suppression of the p38-NF-κB survivial signaling pathway promoted necroptotic and autophagic cell death in TNFα-treated L929 cells.


Subject(s)
Cell Death , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Autophagy , Caspase Inhibitors , Cell Line, Tumor , Mice , NF-kappa B/antagonists & inhibitors , Proline/analogs & derivatives , Proline/pharmacology , Protein Kinase Inhibitors/pharmacology , Thiocarbamates/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Arch Biochem Biophys ; 490(1): 70-5, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19699177

ABSTRACT

Oridonin was reported to induce L929 cell apoptosis via ROS-mediated mitochondrial and ERK pathways; however, the precise mechanisms by which oridonin induces cell death remain unclear. Herein, we found that oridonin treatment induced an increase in G(2)/M phase cell percentage. And, G(2)/M phase arrest was associated with down-regulation of cell cycle related cdc2, cdc25c and cyclinB levels, as well as up-regulation of p21 and p-cdc2 levels. In addition, we discovered that interruption of p53 activation decreased oridonin-induced apoptosis, and blocking ERK by specific inhibitors or siRNA suppressed oridonin-induced p53 activation. Moreover, inhibition of PTK, protein kinase C, Ras, Raf or JNK activation increased oridonin-induced apoptosis. Also, the level of Ras, Raf or JNK was down-regulated by oridonin, and the inhibition of PTK, Ras, Raf activation decreased p-JNK level. In conclusion, oridonin induces L929 cell G(2)/M arrest and apoptosis, which is regulated by promoting ERK-p53 apoptotic pathway and suppressing PTK-mediated survival pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diterpenes, Kaurane/pharmacology , Fibrosarcoma/metabolism , G2 Phase/drug effects , Animals , Cell Division/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Fibrosarcoma/genetics , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , RNA, Small Interfering/metabolism , Time Factors , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , ras Proteins/antagonists & inhibitors
11.
FEBS J ; 276(5): 1291-306, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19187231

ABSTRACT

Autophagy and apoptosis have been known to be interconnected positively or negatively; however, the molecular mechanisms mediating these two cellular processes are not fully understood. In the present study, we demonstrated that the exposure of L929 cells to oridonin led to intracellular reactive oxygen species generation, followed by lipid peroxidation, as well as decreases in superoxide dismutase and glutathione activities. The reactive oxygen species scavenger N-acetyl-cysteine resulted in the complete inhibition of oridonin-induced apoptosis and mitochondrial membrane potential collapse. We showed that reactive oxygen species triggered apoptosis by Bax translocation, cytochrome c release and extracellular signal-regulated kinase activation. Further data confirmed that oridonin also induced L929 cell autophagy, as demonstrated by extensive autophagic vacuolization and the punctuate distribution of monodansylcadaverine staining and GFP-LC3, as well as the LC3-II/LC3-I proportion and Beclin 1 activation. Subsequently, we found that inhibition of autophagy by 3-methyladenine or small interfering RNA against LC3 and Beclin 1 promoted oridonin-induced cell apoptosis. The effects of p38 and nuclear factor-kappa B in oridonin-induced apoptosis and autophagy were further examined. Interruption of p38 and nuclear factor-kappa B activation by specific inhibitors or small interfering RNAs promoted apoptosis and reactive oxygen species generation, but decreased autophagy. Moreover, we showed that inhibition of autophagy reduced oridonin-induced activation of p38. Additionally, nuclear factor-kappa B activation was inhibited by blocking the p38 pathway. Consequently, these findings indicate that oridonin-induced L929 cell apoptosis is regulated by reactive oxygen species-mediated signaling pathways, and that oridonin-induced autophagy may block apoptosis by up-regulating p38 and nuclear factor-kappa B activation.


Subject(s)
Autophagy , Diterpenes, Kaurane/pharmacology , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , Fibrosarcoma , Mice , Microscopy, Electron, Transmission , NF-kappa B/antagonists & inhibitors , Signal Transduction , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...