Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Weiqian Chen is active.

Publication


Featured researches published by Weiqian Chen.


Oncotarget | 2015

Auranofin induces apoptosis by ROS-mediated ER stress and mitochondrial dysfunction and displayed synergistic lethality with piperlongumine in gastric cancer.

Peng Zou; Minxiao Chen; Jiansong Ji; Weiqian Chen; Xi Chen; Shilong Ying; Junru Zhang; Ziheng Zhang; Zhiguo Liu; Shulin Yang; Guang Liang

Gastric cancer (GC) is one of the leading causes of cancer mortality in the world. In addressing the need of treatments for relapsed disease, we report the identification of an existing U.S. Food and Drug Administration-approved small-molecule drug to repurpose for GC treatment. Auranofin (AF), clinically used to treat rheumatic arthritis, but it exhibited preclinical efficacy in GC cells. By increasing intracellular reactive oxygen species (ROS) levels, AF induces a lethal endoplasmic reticulum stress response and mitochondrial dysfunction in cultured GC cells. Blockage of ROS production reversed AF-induced ER stress and mitochondrial pathways activation as well as apoptosis. In addition, AF displays synergistic lethality with an ROS-generating agent piperlongumine, which is a natural product isolated from the long pepper Piper longum L. Taken together, this work provides a novel anticancer candidate for the treatment of gastric cancer. More importantly, it reveals that increased ROS generation might be an effective strategy in treating human gastric cancer.


Redox biology | 2016

Synergistic antitumor activity of rapamycin and EF24 via increasing ROS for the treatment of gastric cancer

Weiqian Chen; Peng Zou; Zhongwei Zhao; Xi Chen; Xiaoxi Fan; Rajamanickam Vinothkumar; Ri Cui; Fazong Wu; Qianqian Zhang; Guang Liang; Jiansong Ji

Mechanistic/mammalian target of rapamycin (mTOR) has emerged as a new potential therapeutic target for gastric cancer. Rapamycin and rapamycin analogs are undergoing clinical trials and have produced clinical responses in a subgroup of cancer patients. However, monotherapy with rapamycin at safe dosage fails to induce cell apoptosis and tumor regression which has hampered its clinical application. This has led to the exploration of more effective combinatorial regimens to enhance the effectiveness of rapamycin. In our present study, we have investigated the combination of rapamycin and a reactive oxygen species (ROS) inducer EF24 in gastric cancer. We show that rapamycin increases intracellular ROS levels and displays selective synergistic antitumor activity with EF24 in gastric cancer cells. This activity was mediated through the activation of c-Jun N terminal kinase and endoplasmic reticulum stress (ER) pathways in cancer cells. We also show that inhibiting ROS accumulation reverses ER stress and prevents apoptosis induced by the combination of rapamycin and EF24. These mechanisms were confirmed using human gastric cancer xenografts in immunodeficient mice. Taken together, our work provides a novel therapeutic strategy for the treatment of gastric cancer. The work reveals that ROS generation could be an important target for the development of new combination therapies for cancer treatment.


Oncotarget | 2016

EF24 induces ROS-mediated apoptosis via targeting thioredoxin reductase 1 in gastric cancer cells

Peng Zou; Yiqun Xia; Weiqian Chen; Xi Chen; Shilong Ying; Zhiguo Feng; Tongke Chen; Qingqing Ye; Zhe Wang; Chenyu Qiu; Shulin Yang; Guang Liang

Gastric cancer (GC) is one of the leading causes of cancer mortality in the world, and finding novel agents for the treatment of advanced gastric cancer is of urgent need. Diphenyl difluoroketone (EF24), a molecule having structural similarity to curcumin, exhibits potent anti-tumor activities by arresting cell cycle and inducing apoptosis. Although EF24 demonstrates potent anticancer efficacy in numerous types of human cancer cells, the cellular targets of EF24 have not been fully defined. We report here that EF24 may interact with the thioredoxin reductase 1 (TrxR1), an important selenocysteine (Sec)-containing antioxidant enzyme, to induce reactive oxygen species (ROS)-mediated apoptosis in human gastric cancer cells. By inhibiting TrxR1 activity and increasing intracellular ROS levels, EF24 induces a lethal endoplasmic reticulum stress in human gastric cancer cells. Importantly, knockdown of TrxR1 sensitizes cells to EF24 treatment. In vivo, EF24 treatment markedly reduces the TrxR1 activity and tumor cell burden, and displays synergistic lethality with 5-FU against gastric cancer cells. Targeting TrxR1 with EF24 thus discloses a previously unrecognized mechanism underlying the biological activity of EF24, and reveals that TrxR1 is a good target for gastric cancer therapy.


Cancer Letters | 2016

Piperlongumine as a direct TrxR1 inhibitor with suppressive activity against gastric cancer

Peng Zou; Yiqun Xia; Jiansong Ji; Weiqian Chen; Jinsan Zhang; Xi Chen; Vinothkumar Rajamanickam; Gaozhi Chen; Zhe Wang; Lingfeng Chen; Yifeng Wang; Shulin Yang; Guang Liang

Piperlongumine (PL), a natural alkaloid isolated from the fruit of long pepper, is known to selectively kill tumor cells while sparing their normal counterparts. However, the cellular target and potent anticancer efficacy of PL in numerous types of human cancer cells have not been fully defined. We report here that PL may interact with the thioredoxin reductase 1 (TrxR1), an important selenocysteine (Sec)-containing antioxidant enzyme, to induce reactive oxygen species (ROS)-mediated apoptosis in human gastric cancer cells. By inhibiting TrxR1 activity and increasing intracellular ROS levels, PL induces a lethal endoplasmic reticulum stress and mitochondrial dysfunction in human gastric cancer cells. Importantly, knockdown of TrxR1 sensitizes cells to PL treatment, and PL displays synergistic lethality with GSH inhibitors (BSO and Erastin) against gastric cancer cells. In vivo, PL treatment markedly reduces the TrxR1 activity and tumor cell burden. Remarkably, TrxR1 was significantly overexpressed in gastric cancer cell lines and human gastric cancer tissues. Targeting TrxR1 with PL thus discloses a previously unrecognized mechanism underlying the biological activity of PL and provides an in-depth insight into the action of PL in the treatment of gastric cancer.


Oncotarget | 2016

Selective killing of gastric cancer cells by a small molecule via targeting TrxR1 and ROS-mediated ER stress activation

Weiqian Chen; Peng Zou; Zhongwei Zhao; Qiaoyou Weng; Xi Chen; Shilong Ying; Qingqing Ye; Zhe Wang; Jiansong Ji; Guang Liang

The thioredoxin reductase (TrxR) 1 is often overexpressed in numerous cancer cells. Targeting TrxR1 leads to a reduction in tumor progression and metastasis, making the enzyme an attractive target for cancer treatment. Our previous research revealed that the curcumin derivative B19 could induce cancer cell apoptosis via activation of endoplasmic reticulum (ER) stress. However, the upstream mechanism and molecular target of B19 is still unclear. In this study, we demonstrate that B19 directly inhibits TrxR1 enzyme activity to elevate oxidative stress and then induce ROS-mediated ER Stress and mitochondrial dysfunction, subsequently resulting in cell cycle arrest and apoptosis in human gastric cancer cells. A computer-assistant docking showed that B19 may bind TrxR1 protein via formation of a covalent bond with the residue Cys-498. Blockage of ROS production totally reversed B19-induced anti-cancer actions. In addition, the results of xenograft experiments in mice were highly consistent with in vitro studies. Taken together, targeting TrxR1 with B19 provides deep insight into the understanding of how B19 exerts its anticancer effects. More importantly, this work indicates that targeting TrxR1 and manipulating ROS levels are effective therapeutic strategy for the treatment of gastric cancer.


Cancer Chemotherapy and Pharmacology | 2016

Curcumin analog EF24 induces apoptosis via ROS-dependent mitochondrial dysfunction in human colorectal cancer cells.

Guodong He; Chen Feng; Rajamanickam Vinothkumar; Weiqian Chen; Xuanxuan Dai; Xi Chen; Qingqing Ye; Chenyu Qiu; Huiping Zhou; Guang Liang; Yubo Xie; Wei Wu

PurposeColorectal cancer is the most commonly diagnosed malignancy with high mortality rates worldwide. Improved therapeutic strategies with minimal adverse side effects are urgently needed. In this study, the anti-tumor effects of EF24, a novel analog of the natural compound curcumin, were evaluated in colorectal cancer cells.MethodsThe anti-tumor activity of EF24 on human colon cancer lines (HCT-116, SW-620, and HT-29) was determined by measures of cell cycle arrest, apoptosis, and mitochondrial function. The contribution of ROS in the EF24-induced anti-tumor activity was evaluated by measures of H2O2 and pretreatment with an ROS scavenger, NAC.ResultsThe findings indicated that EF24 treatment dose-dependently inhibited cell viability and caused cell cycle arrest at G2/M phase in all the tested colon cancer cell lines. Furthermore, we demonstrated that EF24 treatment induced apoptosis effectively via enhancing intracellular accumulation of ROS in both HCT-116 and SW-620 cells, but with moderate effects in HT-29 cells. We found that EF24 treatment decreased the mitochondrial membrane potential in the colon cancer cells, leading to the release of mitochondrial cytochrome c. Also, EF24 induced activation of caspases 9 and 3, causing decreased Bcl-2 protein expression and Bcl-2/Bax ratio. Pretreatment with NAC, a ROS scavenger, abrogated the EF24-induced cell death, apoptosis, cell cycle arrest, and mitochondrial dysfunction, suggesting an upstream ROS generation which was responsible for the anticancer effects of EF24.ConclusionsOur findings support an anticancer mechanism by which EF24 enhanced ROS accumulation in colon cancer cells, thereby resulting in mitochondrial membrane collapse and activated intrinsic apoptotic signaling. Thus, EF24 could be a potential candidate for therapeutic application of colon cancer.


Experimental Cell Research | 2017

Interactions between interleukin-6 and myeloid-derived suppressor cells drive the chemoresistant phenotype of hepatocellular cancer

Min Xu; Zhongwei Zhao; Jingjing Song; Xilin Lan; Siming Lu; Minjiang Chen; Zufei Wang; Weiqian Chen; Xiaoxi Fan; Fazong Wu; Li Chen; Jianfei Tu; Jiansong Ji

Abstract Emerging evidence implicates an important role for myeloid‐derived suppressor cells (MDSCs) in tumor growth, angiogenesis and metastasis. However, limited knowledge is known about the function of MDSCs in response to chemotherapies. In this study, we find that drug‐resistant hepatocellular cancer (HCC) cells‐derived conditioned medium significantly enhances the expansion and immunosuppressive function of MDSCs compared to their parental sensitive cells, which is demonstrated by increased level of arginase, nitric oxide (NO), and reactive oxygen species (ROS). Next, we reveal that drug‐resistant HCC cells‐derived IL‐6 activated MDSCs, which is demonstrated by using an anti‐IL‐6 neutralizing antibody that caused a reduced MDSC immunosuppressive activity. More importantly, the depletion of MDSC via the administration of anti‐Gr‐1 antibody or the blockade of IL‐6 signaling sensitized 5‐FU‐resistant H22 hepatoma to chemotherapy in the immunocompetent C57BL/6N mice. In primary human HCC, IL‐6 expression levels strongly correlate with an MDSC phenotype and chemotherapy response in HCC patients. In conclusion, these results describe a role of IL‐6 in the drug resistance in HCC chemotherapy and suggest that MDSC‐targeting treatments may be potential therapeutic strategy for HCC chemoresistance. Graphical abstract Figure. No Caption available. HighlightsDrug‐resistant HCC cells‐derived IL‐6 enhances the expansion and activity of MDSCs;MDSC depletion or IL‐6 blockade enhances the therapeutic efficacy of chemotherapy;IL‐6 expression correlates with the markers of MDSCs and chemotherapy response.


British Journal of Pharmacology | 2017

Curcuminoid EF24 enhances the anti-tumour activity of Akt inhibitor MK-2206 through ROS-mediated endoplasmic reticulum stress and mitochondrial dysfunction in gastric cancer

Xi Chen; Xuanxuan Dai; Peng Zou; Weiqian Chen; Vinothkumar Rajamanickam; Chen Feng; Weishan Zhuge; Chenyu Qiu; Qingqing Ye; Xiaohua Zhang; Guang Liang

Gastric cancer is one of the leading causes of morbidity and mortality worldwide. Akt is an anti‐apoptotic kinase that plays a dynamic role in cell survival and is implicated in the pathogenesis of gastric cancer. MK‐2206, the first allosteric inhibitor of Akt, is in clinical trials for a number of cancers. Although preclinical studies showed promise, clinical trials reported it had no effect when given alone at tolerated doses. The aim of our study was to delineate the effects of MK‐2206 on gastric cancer cells and explore the ability of combination treatments to enhance the anti‐tumour activity of MK‐2206.


British Journal of Pharmacology | 2017

Curcuminoid EF24 enhances the anti‐tumor activity of Akt inhibitor MK‐2206 through ROS‐mediated ER stress and mitochondrial dysfunction in gastric cancer

Xi Chen; Xuanxuan Dai; Peng Zou; Weiqian Chen; Vinothkumar Rajamanickam; Chen Feng; Weishan Zhuge; Chenyu Qiu; Qingqing Ye; Xiaohua Zhang; Guang Liang

Gastric cancer is one of the leading causes of morbidity and mortality worldwide. Akt is an anti‐apoptotic kinase that plays a dynamic role in cell survival and is implicated in the pathogenesis of gastric cancer. MK‐2206, the first allosteric inhibitor of Akt, is in clinical trials for a number of cancers. Although preclinical studies showed promise, clinical trials reported it had no effect when given alone at tolerated doses. The aim of our study was to delineate the effects of MK‐2206 on gastric cancer cells and explore the ability of combination treatments to enhance the anti‐tumour activity of MK‐2206.


Journal of Experimental & Clinical Cancer Research | 2017

(S)-crizotinib induces apoptosis in human non-small cell lung cancer cells by activating ROS independent of MTH1

Xuanxuan Dai; Gui-Long Guo; Peng Zou; Ri Cui; Weiqian Chen; Xi Chen; Changtian Yin; Wei He; Rajamanickam Vinothkumar; Fan Yang; Xiaohua Zhang; Guang Liang

BackgroundNon–small cell lung cancer (NSCLC) accounts for approximately 80–85% of all lung cancers and is usually diagnosed at an advanced stage with poor prognosis. Targeted therapy has produced unprecedented outcomes in patients with NSCLC as a number of oncogenic drivers have been found. Crizotinib, a selective small-molecule inhibitor, has been widely used for the treatment of NSCLC patients with ALK gene rearrangements. A recent study has also shown that (S)-enantiomer of crizotinib exhibits anticancer activity by targeting the protein mutT homologue (MTH1). Since this discovery, contradictory studies have cast a doubt on MTH1 as a therapeutic target of (S)-crizotinib.MethodsNCI-H460, H1975, and A549 cells and immunodeficient mice were chosen as a model to study the (S)-crizotinib treatment. The changes induced by (S)-crizotinib treatment in cell viability, apoptosis as well as ROS, and endoplasmic reticulum stress pathway in the cells were analyzed by MTT assay, FACSCalibur, Western blotting, ROS imaging and electron microscopy.ResultsHere, we report that MTH1 does not affect survival of NSCLC cells. We found that (S)-crizotinib induces lethal endoplasmic reticulum stress (ER) response in cultured NSCLC cells by increasing intracellular levels of reactive oxygen species (ROS). Blockage of ROS production markedly reversed (S)-crizotinib-induced ER stress and cell apoptosis, independent of MTH1. We confirmed these findings in NSCLC xenograft studies and showed that (S)-crizotinib-induced ER stress and cell apoptosis.ConclusionsOur results reveal a novel antitumor mechanism of (S)-crizotinib in NSCLC which involves activation of ROS-dependent ER stress apoptotic pathway and is independent of MTH1 inhibition.

Collaboration


Dive into the Weiqian Chen's collaboration.

Top Co-Authors

Avatar

Guang Liang

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Xi Chen

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Jiansong Ji

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Peng Zou

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Qingqing Ye

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chenyu Qiu

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Xiaoxi Fan

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Xuanxuan Dai

First Affiliated Hospital of Wenzhou Medical University

View shared research outputs
Top Co-Authors

Avatar

Chen Feng

Wenzhou Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge