首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
《Autophagy》2013,9(9):966-978
Quercetin, a dietary antioxidant present in fruits and vegetables, is a promising cancer chemopreventive agent that inhibits tumor promotion by inducing cell cycle arrest and promoting apoptotic cell death. In this study, we examined the biological activities of quercetin against gastric cancer. Our studies demonstrated that exposure of gastric cancer cells AGS and MKN28 to quercetin resulted in pronounced pro-apoptotic effect through activating the mitochondria pathway. Meanwhile, treatment with quercetin induced appearance of autophagic vacuoles, formation of acidic vesicular organelles (AVOs), conversion of LC3-I to LC3-II, recruitment of LC3-II to the autophagosomes as well as activation of autophagy genes, suggesting that quercetin initiates the autophagic progression in gastric cancer cells. Furthermore, either administration of autophagic inhibitor chloroquine or selective ablation of atg5 or beclin 1 using small interfering RNA (siRNA) could augment quercetin-induced apoptotic cell death, suggesting that autophagy plays a protective role against quercetin-induced apoptosis. Moreover, functional studies revealed that quercetin activated autophagy by modulation of Akt-mTOR signaling and hypoxia-induced factor 1α (HIF-1α) signaling. Finally, a xenograft model provided additional evidence for occurrence of quercetin-induced apoptosis and autophagy in vivo. Together, our studies provided new insights regarding the biological and anti-proliferative activities of quercetin against gastric cancer, and may contribute to rational utility and pharmacological study of quercetin in future anti-cancer research.  相似文献   

2.
Paclitaxel is one of the most effective chemotherapy drugs for advanced cervical cancer. However, acquired resistance of paclitaxel represents a major barrier to successful anticancer treatment. In this study, paclitaxel-resistant HeLa sublines (HeLa-R cell lines) were established by continuous exposure and increased autophagy level was observed in HeLa-R cells. 3-Methyladenine or ATG7 siRNA, autophagy inhibitors, could restore sensitivity of HeLa-R cells to paclitaxel compared with parental HeLa cells. To determine the underlying molecular mechanism, differentially expressed proteins between HeLa and HeLa-R cells were identified by two-dimensional gel electrophoresis coupled with electrospray ionization quadrupole time-of-flight MS/MS. We found glycolysis-associated proteins were upregulated in HeLa-R cell lines. Inhibition of glycolysis by 2-deoxy-D-glucose or koningic acid could decrease autophagy and enhance sensitivity of HeLa-R cells to paclitaxel. Moreover, glycolysis could activate HIF1-α. Downregulation of HIF1-α by specific siRNA could decrease autophagy and resensitize HeLa-R cells to paclitaxel. Taken together, a possible Warburg effect activated HIF1-α-mediated signaling-induced autophagic pathway is proposed, which may provide new insight into paclitaxel chemoresistance.Cervical cancer, a common malignant tumor, is an vital cause of morbidity and mortality among women worldwide.1 Paclitaxel, the targets of which are the microtubules of cancer cells, is one of the most useful anticancer drugs against cervical cancer.2 Paclitaxel can destroy the dynamic equilibrium of tubulin between soluble dimers and polymerized form to make the microtubule structure stable. In addition, it is a strong inhibitor of chromosomal replication by obstructing tumor cells in the mitotic phases or late G2.3 However, acquired chemoresistance to paclitaxel obviously limits the successful treatment of cervical cancer. One main explanation on tumor cell resistance to paclitaxel is the overexpression of P-glycoprotein (P-gp, MDR-1), which works as a drug efflux pump. However, clinical utility of P-gp inhibitors are often ineffective or toxic at the doses required to attenuate P-gp function.4, 5, 6 Other possible mechanisms of action contain alterations in the drug-binding affinity of the microtubules,7 changes of tubulin structure and cell cycle deregulation.8, 9, 10, 11 Thus, paclitaxel-resistant mechanisms are complicated and still not entirely clear until now.As a self-proteolysis procedure in almost all eukaryotic cells, autophagy activated by adverse cellular environment contributes to the breakdown of intracellular components within lysosomes to supply an alternative source of energy and thus sustain cell survival.12, 13 However, it has been shown that cell death could be inhibited by suppressing expression of some vital autophagy-associated genes, underscoring the functional role of autophagy in the cell death.14, 15 Several important autophagy-associated proteins, such as Beclin 1 and PtdIns3K class I, have important roles in the control of both autophagy and apoptosis.16, 17, 18 Thus, the function of autophagy has been described as a double-edged sword that can work both as a protector and killer of cells, which depends on the developmental stage of the disease or the surrounding microenvironment.19It has been reported that anticancer treatment (such as radiotherapy, chemotherapy and molecular targeted therapy) could induce autophagy in cancer cells. In addition, impaired autophagy could make cancer cells sensitize to these therapies, indicating a hopeful strategy to better the efficacy of cancer treatment.11, 20, 21, 22 However, few studies on the underlying molecular mechanism of chemoresistance-associated autophagy were carried out.In this study, increased levels of autophagy were observed in paclitaxel-resistant HeLa sublines (HeLa-R cell lines). 3-Methyladenine (3-MA) or ATG7 siRNA, autophagy inhibitors, restored sensitivity of HeLa-R cells to paclitaxel. In addition, a group of metabolic proteins with significant alteration were identified by proteomics, which may suggest the switch of cellular metabolism from tricarboxylic acid cycle to glycolysis. Moreover, inhibition of glycolysis by 2-deoxy-D-glucose (2-DG) or koningic acid (KA) could inhibit autophagy and enhance sensitivity of HeLa-R cells to paclitaxel. In addition, HIF1-α could be activated by glycolysis and HIF1-α siRNA could decrease autophagy and resensitize HeLa-R cells to paclitaxel. In conclusion, a possible Warburg effect activated HIF1-α-mediated signaling-induced autophagic pathway is proposed, which may provide new insight into paclitaxel chemoresistance.  相似文献   

3.
4.
5.
6.
7.
All-trans retinoic acid (ATRA), a pan-retinoic acid receptor (RAR) agonist, is, along with other retinoids, a promising therapeutic agent for the treatment of a variety of solid tumors. On the one hand, preclinical studies have shown promising anticancer effects of ATRA in breast cancer; on the other hand, resistances occurred. Autophagy is a cellular recycling process that allows the degradation of bulk cellular contents. Tumor cells may take advantage of autophagy to cope with stress caused by anticancer drugs. We therefore wondered if autophagy is activated by ATRA in mammary tumor cells and if modulation of autophagy might be a potential novel treatment strategy. Indeed, ATRA induces autophagic flux in ATRA-sensitive but not in ATRA-resistant human breast cancer cells. Moreover, using different RAR agonists as well as RARα-knockdown breast cancer cells, we demonstrate that autophagy is dependent on RARα activation. Interestingly, inhibition of autophagy in breast cancer cells by either genetic or pharmacological approaches resulted in significantly increased apoptosis under ATRA treatment and attenuated epithelial differentiation. In summary, our findings demonstrate that ATRA-induced autophagy is mediated by RARα in breast cancer cells. Furthermore, inhibition of autophagy results in enhanced apoptosis. This points to a potential novel treatment strategy for a selected group of breast cancer patients where ATRA and autophagy inhibitors are applied simultaneously.Macroautophagy (hereafter referred to as autophagy) is a conserved mechanism characterized by the formation of double-membrane structures. These so-called autophagosomes deliver cytoplasmic material to the lysosome for subsequent degradation.1 Basal autophagy requires tight regulation as alterations in autophagy have been associated with many pathological conditions, including cancer.2 In addition, autophagy has been linked to fundamental processes such as development and cellular differentiation. In these processes, autophagy contributes to cell remodeling as observed during erythrocyte, lymphocyte or adipocyte differentiation.3 In the context of cancer and cancer therapy, autophagy is a double-edged sword. Owing to its homeostatic role in the removal of potentially harmful damaged organelles and protein aggregates, it is suggested to be tumor suppressive under normal conditions.4 In cancer cells, however, autophagy can be oncogenic, enabling survival under stressful conditions.5 Hence, the role of autophagy in tumorigenesis is clearly dependent on the cellular context and the tumor stage. In some cases, therapeutic agents induce an autophagic response that can promote resistance to treatment. In other cases, autophagy contributes to the action of antitumor agents.6 Therefore, knowledge about the action exerted by autophagy in response to anticancer treatments is a prerequisite for the identification of patients benefiting from therapeutic strategies based on autophagy modulators.All-trans retinoic acid (ATRA), the active metabolite of vitamin A, exerts diverse functions in almost every cell and organ system. ATRA controls cell proliferation, differentiation as well as immune, and neuronal functions primarily via regulation of gene expression.7 Endogenous retinoid levels are altered in different diseases of the lung, kidney and central nervous system, and contribute to their pathophysiology.8 ATRA is successfully used in the treatment of acute promyelocytic leukemia (APL), where it induces granulocytic differentiation of the blast and subsequent cell death of the differentiated leukemic cells. Importantly, ATRA-induced differentiation of the APL cell line, NB4, involves induction of macroautophagy.9, 10, 11, 12 In addition to its cytodifferentiating capacity in APL, ATRA has been proposed as an antitumorigenic agent for other types of cancer. The antiproliferative, cytodifferentiating and proapoptotic effects of retinoids are predominantly mediated by the nuclear hormone retinoid acid receptors RARα, RARβ and RARγ.13, 14 In breast cancer, preclinical studies have shown that retinoids are promising therapeutic agents. However, the clinical trials conducted so far were somewhat disappointing, possibly as a consequence of the study designs.15 Breast cancer is a highly heterogeneous disease represented as a collection of diseases with distinct histopathological and molecular features. The most important clinical classification of this tumor is based on the determination of ER (estrogen receptor), PR (progesterone receptor) and HER2 (human epidermal growth factor receptor-2) receptors. ER-positive breast cancer patients are eligible for hormonal therapies, whereas HER2 oncogenic activity can be blocked using targeted therapies.16 Approximately 15–20% of breast carcinomas overexpress HER2, which is associated with poor prognosis.17 Owing to the development of resistance to current HER2-targeted treatments such as trastuzumab and lapatinib alternative therapeutic strategies are required.18, 19 ATRA was recently shown to exert strong antitumor activity in cell lines representing a subgroup of HER2-positive breast tumors characterized by coamplification of the ERBB2 and RARα genes.20 This antitumor activity is remarkably stimulated by simultaneous HER2 inhibition with lapatinib. In addition, autophagy is induced upon ATRA treatment of the APL-derived cell line NB49, 10, 11 and retinoids have clinical relevance in breast cancer. Thus, we investigated whether and how autophagy is induced in breast cancer cells. In addition, we evaluated whether autophagy modulation represents a potential therapeutic strategy for potentiating ATRA cytotoxicity in breast cancer cells.  相似文献   

8.
There are two possible outcomes when DNA damage occurs in normal mammalian cells: either induction of cell-cycle checkpoint which inhibits the progress of the cell cycles as well as activates DNA repair pathways, or activation of apoptosis to eliminate damaged cells. The p53 tumour-suppressor gene plays a key role in selecting these pathways. In our present works, the human gastric cancer cell line AGS was treated with tripchlorolide, a potent antitumor compound purified from a Chinese herb Tripterygium Wilfordii Hook. Single cell gel electrophoresis (Comet assay) showed that the treatment of tripchlorolide resulted in DNA damage in AGS cells. The damaged AGS cells went through apoptosis, which was time- and dose- dependent.  相似文献   

9.
10.
11.
Y Zhu  M Zhu  P Lance 《Experimental cell research》2012,318(19):2520-2530
COX-2 is a major inflammatory mediator implicated in colorectal inflammation and cancer. However, the exact origin and role of COX-2 on colorectal inflammation and carcinogenesis are still not well defined. Recently, we reported that COX-2 and iNOS signalings interact in colonic CCD18Co fibroblasts. In this article, we investigated whether activation of COX-2 signaling by IL1β in primary colonic fibroblasts obtained from normal and cancer patients play a critical role in regulation of proliferation and invasiveness of human colonic epithelial cancer cells. Our results demonstrated that COX-2 level was significantly higher in cancer associated fibroblasts than that in normal fibroblasts with or without stimulation of IL-1β, a powerful stimulator of COX-2. Using in vitro assays for estimating proliferative and invasive potential, we discovered that the proliferation and invasiveness of the epithelial cancer cells were much greater when the cells were co-cultured with cancer associated fibroblasts than with normal fibroblasts, with or without stimulation of IL1β. Further analysis indicated that the major COX-2 product, prostaglandin E(2), directly enhanced proliferation and invasiveness of the epithelial cancer cells in the absence of fibroblasts. Moreover, a selective COX-2 inhibitor, NS-398, blocked the proliferative and invasive effect of both normal and cancer associate fibroblasts on the epithelial cancer cells, with or without stimulation of IL-1β. Those results indicate that activation of COX-2 signaling in the fibroblasts plays a major role in promoting proliferation and invasiveness of the epithelial cancer cells. In this process, PKC is involved in the activation of COX-2 signaling induced by IL-1β in the fibroblasts.  相似文献   

12.
J Mao  S Fan  W Ma  P Fan  B Wang  J Zhang  H Wang  B Tang  Q Zhang  X Yu  L Wang  B Song  L Li 《Cell death & disease》2014,5(1):e1039
The Wnt1 protein, a secreted ligand that activates Wnt signaling pathways, contributes to the self-renewal of cancer stem cells (CSCs) and thus may be a major determinant of tumor progression and chemoresistance. In a series of gastric cancer specimens, we found strong correlations among Wnt1 expression, CD44 expression, and the grade of gastric cancer. Stable overexpression of Wnt1 increased AGS gastric cancer cells'' proliferation rate and spheroids formation, which expressed CSC surface markers Oct4 and CD44. Subcutaneous injection of nude mice with Wnt1-overexpressing AGS cells resulted in larger tumors than injection of control AGS cells. Salinomycin, an antitumor agent, significantly reduced the volume of tumor caused by Wnt1-overexpressing AGS cells in vivo. This is achieved by inhibiting the proliferation of CD44+Oct4+ CSC subpopulation, at least partly through the suppression of Wnt1 and β-catenin expression. Taken together, activation of Wnt1 signaling accelerates the proliferation of gastric CSCs, whereas salinomycin acts to inhibit gastric tumor growth by suppressing Wnt signaling in CSCs. These results suggest that Wnt signaling might have a critical role in the self-renewal of gastric CSCs, and salinomycin targeting Wnt signaling may have important clinical applications in gastric cancer therapy.  相似文献   

13.
Background and aims: Transforming growth factor-beta (TGFβ) is known to potently inhibit cell growth. Loss of responsiveness to TGFβ inhibition on cell growth is a hallmark of many types of cancer, yet its mechanism is not fully understood. Membrane-anchored heparin-binding EGF-like growth factor (proHB-EGF) ectodomain is cleaved by a disintegrin and metalloproteinase (ADAM) members and is implicated in epidermal growth factor receptor (EGFR) transactivation. Recently, nuclear translocation of the C-terminal fragment (CTF) of pro-HB-EGF was found to induce cell growth. We investigated the association between TGFβ and HB-EGF signal transduction via ADAM activation.Materials and methods: The CCK-8 assay in two gastric cancer cell lines was used to determine the effect for cell growth by TGFβ. The effect of two ADAM inhibitors was also evaluated. Induction of EGFR phosphorylation by TGFβ was analyzed and the effect of the ADAM inhibitors was also examined. Nuclear translocation of HB-EGF-CTF by shedding through ADAM activated by TGFβ was also analyzed. EGFR transactivation, HB-EGF-CTF nuclear translocation, and cell growth were examined under the condition of ADAM17 knockdown.Result: TGFβ-induced EGFR phosphorylation of which ADAM inhibitors were able to inhibit. TGFβ induced shedding of proHB-EGF allowing HB-EGF-CTF to translocate to the nucleus. ADAM inhibitors blocked this nuclear translocation. TGFβ enhanced gastric cancer cell growth and ADAM inhibitors suppressed this effect. EGFR phosphorylation, HB-EGF-CTF nuclear translocation, and cell growth were suppressed in ADAM17 knockdown cells.Conclusion: HB-EGF-CTF nuclear translocation and EGFR transactivation from proHB-EGF shedding mediated by ADAM17 activated by TGFβ might be an important pathway of gastric cancer cell proliferation by TGFβ.  相似文献   

14.

Background

The hydroxylated derivatives of cholesterol, such as the oxysterols, play important roles in lipid metabolism. In particular, 25-hydroxycholesterol (25HC) has been implicated in a variety of metabolic events including cholesterol homeostasis and atherosclerosis. 25HC is detectable in human plasma after ingestion of a meal rich in oxysterols and following a dietary cholesterol challenge. In addition, the levels of oxysterols, including 25HC, have been found to be elevated in hypercholesterolemic serum.

Methodology/Principal Findings

Here, we demonstrate that the estrogen receptor (ER) α mediates gene expression changes and growth responses induced by 25HC in breast and ovarian cancer cells. Moreover, 25HC exhibits the ERα-dependent ability like 17β-estradiol (E2) to inhibit the up-regulation of HIF-1α and connective tissue growth factor by hypoxic conditions in cardiomyocytes and rat heart preparations and to prevent the hypoxia-induced apoptosis.

Conclusions/Significance

The estrogen action exerted by 25HC may be considered as an additional factor involved in the progression of breast and ovarian tumors. Moreover, the estrogen-like activity of 25HC elicited in the cardiovascular system may play a role against hypoxic environments.  相似文献   

15.
Phagocytosis of naturally dying cells usually blocks inflammatory reactions in host cells. We have recently observed that clearance of cells dying through autophagy leads to a pro-inflammatory response in human macrophages. Investigating this response further, we found that during engulfment of MCF-7 or 293T cells undergoing autophagic death, but not apoptotic or anoikic ones, caspase-1 was activated and IL-1β was processed, then secreted in a MyD88-independent manner. Autophagic dying cells were capable of preventing some LPS-induced pro-inflammatory responses, such as TNFα, IL-6 and IL-8 induction, but synergized with LPS for IL-1β production. Caspase-1 inhibition prevented macrophage IL-1β release triggered by the dying cells and also other pro-inflammatory cytokines which were not formed in the presence of IL-1 receptor antagonist anakinra either. IL-1β secretion was also observed using calreticulin knock down or necrostatin treated autophagic MCF-7 cells and it required phagocytosis of the dying cells which led to ATP secretion from macrophages. Blocking K (+) efflux during phagocytosis, the presence of apyrase, adding an antagonist of the P2X7 receptor or silencing the NOD-like receptor protein NALP3 inhibited IL-1β secretion. These data suggest that during phagocytosis of autophagic dying cells ATP, acting through its receptor, initiates K (+) efflux, inflammasome activation and secretion of IL-1β, which initiates further pro-inflammatory events. Thus, autophagic death of malignant cells and their clearance may lead to immunogenic response.  相似文献   

16.
17.
18.
Gastric cancer is a common malignancy with high mortality. Long noncoding RNA (lncRNA) zinc finger antisense (ZFAS)1 is upregulated in gastric cancer specimens compared with the para-carcinoma tissues. The silencing of ZFAS1 inhibited the growth, proliferation, cell cycle progress, migration, invasion and epithelial-mesenchymal transition (EMT), and enhanced the sensitivity to cis-platinum or paclitaxel in SGC7901 cells, as evidenced by the expression changes of proliferating cell nuclear antigen, Cyclin D1, Cyclin E, Cyclin B1, E-cadherin, N-cadherin, vimentin, matrix metalloproteinase (MMP)-2 and MMP-14. The ZFAS1 also activated the Wnt/β-catenin signaling. Subsequently, the ZFAS1 knockdown-induced the inhibition of migration, invasion, EMT and resistance to chemotherapeutic reagens was reversed by the overexpression of β-catenin. In summary, the silencing of ZFAS1 inhibited the growth, proliferation, cell cycle progress, migration, invasion, EMT and chemotherapeutic tolerance by blocking the Wnt/β-catenin signaling in gastric cancer cells.  相似文献   

19.
Hepatic progenitor cells (HPCs) are activated in the chronic liver injury and are found to participate in the progression of liver fibrosis, while the precise role of HPCs in liver fibrosis remains largely elusive. In this study, by immunostaining of human liver sections, we confirmed that HPCs were activated in the cirrhotic liver and secreted transforming growth factor β (TGF-β) and connective tissue growth factor (CTGF), both of which were important inducers of liver fibrosis. Besides, we used HPC cell lines LE/6 and WB-F344 as in vitro models and found that TGF-β induced secretion of CTGF in HPCs. Moreover, TGF-β signaling was intracrine activated and contributed to autonomous secretion of CTGF in HPCs. Furthermore, we found that TGF-β induced expression of CTGF was not mediated by TGF-β activated Smad signaling but mediated by TGF-β activated Erk, JNK and p38 MAPK signaling. Taken together, our results provide evidence for the role of HPCs in liver fibrosis and suggest that the production of CTGF by TGF-β activated MAPK signaling in HPCs may be a therapeutic target of liver fibrosis.  相似文献   

20.
SIRT1, a highly conserved NAD(+)-dependent protein deacetylase, is a key metabolic sensor that directly links nutrient signals to animal metabolic homeostasis. Although SIRT1 has been implicated in a number of hepatic metabolic processes, the mechanisms by which hepatic SIRT1 modulates bile acid metabolism are still not well understood. Here we report that deletion of hepatic SIRT1 reduces the expression of farnesoid X receptor (FXR), a nuclear receptor that regulates bile acid homeostasis. We provide evidence that SIRT1 regulates the expression of FXR through hepatocyte nuclear factor 1α (HNF1α). SIRT1 deficiency in hepatocytes leads to decreased binding of HNF1α to the FXR promoter. Furthermore, we show that hepatocyte-specific deletion of SIRT1 leads to derangements in bile acid metabolism, predisposing the mice to development of cholesterol gallstones on a lithogenic diet. Taken together, our findings indicate that SIRT1 plays a vital role in the regulation of hepatic bile acid homeostasis through the HNF1α/FXR signaling pathway.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号