Yongmei Xie
Sichuan University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Yongmei Xie.
Cell Death and Disease | 2015
Fangfang Yang; M Hu; Qian Lei; Yong Xia; Yongxia Zhu; Yali Li; H Jie; C Liu; Ying Xiong; Z Zuo; A Zeng; Luoting Yu; G Shen; Dong Wang; Yongmei Xie; Tinghong Ye; Yuquan Wei
Breast carcinoma is the most common female cancer with considerable metastatic potential. Signal transducers and activators of the transcription 3 (Stat3) signaling pathway is constitutively activated in many cancers including breast cancer and has been validated as a novel potential anticancer target. Here, we reported our finding with nifuroxazide, an antidiarrheal agent identified as a potent inhibitor of Stat3. The potency of nifuroxazide on breast cancer was assessed in vitro and in vivo. In this investigation, we found that nifuroxazide decreased the viability of three breast cancer cell lines and induced apoptosis of cancer cells in a dose-dependent manner. In addition, western blot analysis demonstrated that the occurrence of its apoptosis was associated with activation of cleaved caspases-3 and Bax, downregulation of Bcl-2. Moreover, nifuroxazide markedly blocked cancer cell migration and invasion, and the reduction of phosphorylated-Stat3Tyr705, matrix metalloproteinase (MMP) MMP-2 and MMP-9 expression were also observed. Furthermore, in our animal experiments, intraperitoneal administration of 50 mg/kg/day nifuroxazide suppressed 4T1 tumor growth and blocked formation of pulmonary metastases without detectable toxicity. Meanwhile, histological and immunohistochemical analyses revealed a decrease in Ki-67-positive cells, MMP-9-positive cells and an increase in cleaved caspase-3-positive cells upon nifuroxazide. Notably, nifuroxazide reduced the number of myeloid-derived suppressor cell in the lung. Our data indicated that nifuroxazide may potentially be a therapeutic agent for growth and metastasis of breast cancer.
Breast Cancer Research and Treatment | 2014
Tinghong Ye; Xiawei Wei; Tao Yin; Yong Xia; Deliang Li; Bin Shao; Sisi He; Min Luo; Xiang Gao; Zhiyao He; Can Luo; Ying Xiong; Ningyu Wang; Jun Zeng; Lifeng Zhao; Guobo Shen; Yongmei Xie; Luoting Yu; Yuquan Wei
Aberrant fibroblast growth factor (FGF) and FGF receptor (FGFR) system have been associated with breast cancer. The objectives of our study were to investigate the effects and mechanisms of FGFR inhibition on tumor growth and metastasis on breast cancer. Our studies showed that the FGFR inhibitor PD173074 decreased the viability of several human breast cancer cells, as well as 4T1 murine mammary tumor cells. Therefore, we chose 4T1 cells to study PD173074’s antitumor mechanism. Flow cytometry showed that PD173074 induced 4T1 cell apoptosis in a concentration-dependent manner. Western blot demonstrated that PD173074-induced apoptosis was correlated with the inhibition of Mcl-1 and survivin. Moreover, PD173074 also significantly increased the ratio of Bax/Bcl-2. PD173074 could also block 4T1 cell migration and invasion in vitro. In 4T1 tumor-bearing mice, PD173074 significantly inhibited tumor growth without obvious side effects. Meanwhile, PD173074 functionally reduced microvessel density and proliferation index and induced tumor apoptosis. Importantly, we found that FGFR inhibition by PD173074 reduced myeloid-derived suppressor cells (MDSCs) in the blood, spleens and tumors, accompanied by the increased infiltration of CD4+ and CD8+ T cells in the spleens and tumors. Furthermore, PD173074 significantly inhibited breast tumor metastasis to the lung of inoculated 4T1 breast cancer cells, which was accompanied by a reduction in MDSCs. Our findings suggested that FGFR inhibition could delay breast tumor progression, impair lung metastasis and break immunosuppression by effecting on tumor microenvironment, which may provide a promising therapeutic approach for breast cancer patient.
International Journal of Pharmaceutics | 2008
Yongmei Xie; Ping Xie; Xin Song; Xiao-Hai Tang; Hang Song
Esomeprazole zinc (EZ) is a poorly water-soluble substance. In order to increase its dissolution rate and bioavailability, solid dispersions of esomeprazole zinc (SDEZ) in polyethylene glycol 4000 (PEG4000) with different EZ to PEG4000 ratios were prepared by solvent method. Our studies showed that dissolution rate of EZ were distinctively increased in the solid dispersion system compared to that in pure EZ or physical mixtures. The increase of dissolution rate was obviously related to the ratio of EZ to PEG4000. The solid dispersion system (EZ/PEG4000=1/8, w/w) gave the highest dissolution rate: about 14.7-fold higher than that of the pure EZ. EZ was proved to be in amorphous state in this solid dispersion by using differential scanning calorimetry (DSC) and scanning electron microscopy (SEM) techniques. In vivo administration studies, SDEZ in enteric capsule (SDEZ-EC) has a lower Cmax and a longer Tmax than that of esomeprazole magnesium enteric-coated tablet (Nexium), and the differences of Cmax and Tmax between SDEZ-EC and Nexium are significant. This result suggests SDEZ-EC has a lower absorption rate than Nexium and corresponds with the in vitro dissolution.
Scientific Reports | 2016
Yongxia Zhu; Tinghong Ye; Xi Yu; Qian Lei; Fangfang Yang; Yong Xia; Li Liu; Hongxia Deng; Tiantao Gao; Cui-Ting Peng; Wei-Qiong Zuo; Ying Xiong; Lidan Zhang; Ning-Yu Wang; Lifeng Zhao; Yongmei Xie; Luoting Yu; Yuquan Wei
Melanoma is a highly malignant neoplasm of melanocytes with considerable metastatic potential and drug resistance, explaining the need for new candidates that inhibit tumor growth and metastasis. The signal transducer and activator of the transcription 3 (Stat3) signaling pathway plays an important role in melanoma and has been validated as promising anticancer target for melanoma therapy. In this study, nifuroxazide, an antidiarrheal agent identified as an inhibitor of Stat3, was evaluated for its anti-melanoma activity in vitro and in vivo. It had potent anti-proliferative activity against various melanoma cell lines and could induce G2/M phase arrest and cell apoptosis. Moreover, nifuroxazide markedly impaired melanoma cell migration and invasion by down-regulating phosphorylated-Src, phosphorylated-FAK, and expression of matrix metalloproteinase (MMP) -2, MMP-9 and vimentin. It also significantly inhibited tumor growth without obvious side effects in the A375-bearing mice model by inducing apoptosis and reducing cell proliferation and metastasis. Notably, nifuroxazide significantly inhibited pulmonary metastases, which might be associated with the decrease of myeloid-derived suppressor cells (MDSCs). These findings suggested that nifuroxazide might be a potential agent for inhibiting the growth and metastasis of melanoma.
Bioorganic & Medicinal Chemistry | 2010
Xiao-Hai Tang; Ping Xie; Yi Ding; Liang-Yin Chu; Jing-Ping Hou; Jinliang Yang; Xin Song; Yongmei Xie
Adriamycin (ADM) has been widely used in the treatment of many types of solid malignant tumor. However, cardiotoxicity, multidrug resistance and a short half-life in vivo are significant problems that limit its clinical application. To resolve these problems, a novel pectin-adriamycin conjugate (PAC) was synthesized by attaching ADM to low-methoxylated pectin via an amide linkage. The ADM content and weight-average molecular weight (Mw) of PAC were greater than 25% (w/w) and 50,360 g/mol, respectively. PAC was highly stable in plasma, but 33.2% of ADM was released from PAC after incubation for 30 h with lysosomes derived from rat liver. PAC was distributed uniformly in the cytoplasm of most A549 cells and accumulated in the nucleus of a few A549 cells after incubation for 30 h. At concentrations equivalent to 0.125-1.000 microg of ADM/mL, PAC did not inhibit the growth of either A594 or B16 cells to the same extent as free ADM or a mixture of ADM and pectin. Interestingly, at all concentrations, PAC inhibited the growth of 2780cp cells in vitro significantly more effectively than ADM or the mixture of ADM and pectin. The anticancer effect of PAC in vivo was evaluated with C57BL/6 mice bearing pulmonary metastases of B16 cells. Compared with ADM and the mixture of ADM and pectin, PAC suppressed tumor growth significantly and prolonged the mean survival time of the B16-inoculated mice. PAC has great potential for development as a tumor targeting polymer-drug.
International Journal of Pharmaceutics | 2013
Qing Ma; Bo Li; Yiyi Yu; Ying Zhang; Yang Wu; Wen Ren; Yu Zheng; Jun He; Yongmei Xie; Xiangrong Song; Gu He
A novel biomaterial poly(ethylene glycol)-block-poly(γ-cholesterol-l-glutamate) (mPEG-PCHLG) was designed and synthesized by introducing cholesterol side chains into this pegylated poly(amino acid) copolymers to enlarge the core space to increase the drug capacity. Paclitaxel (PTX) loaded mPEG-PCHLG nanoparticles (PTX-mPEG-PCHLG-Nps) were developed for the first time. The preparation method of nanoparticles was screened and optimized systemically. The optimal PTX-mPEG-PCHLG-Nps with the average diameter of 213.71 nm were constructed through the O/W single-emulsion solvent evaporation method. The entrapment efficiency and drug loading was 38.02 ± 4.51% and 93.90 ± 4.56%, respectively. PTX-mPEG-PCHLG-Nps were spherical and well-dispersed and displayed a dramatic sustained-release property. The in vitro cytotoxicity experiments demonstrated that the blank mPEG-PCHLG nanoparticles had no cytotoxicities on four tumor cell lines including A549, HepG-2, MCF-7 and C26, which implied that mPEG-PCHLG might be biocompatible. PTX-mPEG-PCHLG-Nps obtained the same cell growth inhibition activities as free PTX when incubated with the above tumor cells for 48h. It can be inferred that PTX-mPEG-PCHLG-Nps could probably have higher anticancer efficacy due to the inadequate release of PTX from nanoparticles. PTX-mPEG-PCHLG-Nps achieved the highest antitumor activity in A549 rather than HepG-2, MCF-7 and C26, thus PTX-mPEG-PCHLG-Nps could have a potential application in lung cancer therapy. All the data indicated that mPEG-PCHLG was one of biocompatible biomaterials and worth being widely investigated as hydrophobic antitumor drug carrier.
Biomedicine & Pharmacotherapy | 2015
Fangfang Yang; Yang Yu; Qian Lei; Anqi Zeng; Yali Li; Yongmei Xie; Tinghong Ye; Yuquan Wei
Melanoma is highly resistant to most conventional treatment, and the incidence and mortality rates are increasing rapidly worldwide. The objective of this study was to determine the anticancer effects of lobaplatin on the melanoma carcinoma cell line B16-F10 in vitro, and explored its mechanisms of action. Our results have shown that lobaplatin inhibited cell proliferation in human melanoma A375 and CHL-1 cells and murine melanoma B16-F10 cells in a concentration- and time-dependent manner. Flow cytometry assay confirmed that lobaplatin affected B16-F10 cell survival by blocking cell cycle progression in G2/M phase and inducing apoptosis in a concentration-dependent manner. In addition, the apoptosis was associated with downregulation of anti-apoptotic protein Bcl-2 while upregulation of pro-apoptotic protein Bax. Lobaplatin could also decrease the mitochondrial membrane potential, indicating that lobaplatin may induce apoptosis via mitochondria-mediated apoptotic pathway. Furthermore, lobaplatin blocked B16-F10 cell migration and invasion in vitro. These results suggested that lobaplatin could be an effective chemotherapeutic agent in melanoma treatment by inhibiting proliferation, inducing apoptosis, cell cycle arrest and blocking cell migration and invasion.
Cell Death and Disease | 2017
Tinghong Ye; Fangfang Yang; Yongxia Zhu; Yali Li; Qian Lei; Yong Xia; Ying Xiong; Lidan Zhang; Ningyu Wang; Lifeng Zhao; Hongfeng Gou; Yongmei Xie; Sheng-Yong Yang; Luoting Yu; Li Yang; Yuquan Wei
Colorectal carcinoma (CRC) is the one of the most common cancers with considerable metastatic potential, explaining the need for new drug candidates that inhibit tumor metastasis. The signal transducers and activators of the transcription 3 (Stat3) signaling pathway has an important role in CRC and has been validated as a promising anticancer target for CRC therapy. In the present study, we report our findings on nifuroxazide, an antidiarrheal agent identified as an inhibitor of Stat3. Our studies showed that nifuroxazide decreased the viability of three CRC cell lines and induced apoptosis of cancer cells in a concentration-dependent manner. Moreover, western blot analysis demonstrated that the occurrence of its apoptosis was correlated with the activation of Bax and cleaved caspase-3, and decreased the expression of Bcl-2. In addition, nifuroxazide markedly impaired CRC cell migration and invasion by downregulating phosphorylated-Stat3Tyr705, and also impaired the expression of matrix metalloproteinases (MMP-2 and MMP-9). Furthermore, our studies showed that nifuroxazide also significantly inhibited the tumor metastasis in lung and abdomen metastasis models of colon cancer. Meanwhile, nifuroxazide functionally reduced the proliferation index, induced tumor apoptosis and impaired metastasis. Notably, nifuroxazide reduced the number of myeloid-derived suppressor cells in the blood, spleens and tumors, accompanied by the increased infiltration of CD8+ T cells in the tumors. Importantly, a marked decrease in the number of M2-type macrophages in tumor in the abdomen metastasis model was also observed. Taken together, our results indicated that nifuroxazide could effectively inhibit tumor metastasis by mediating Stat3 pathway and it might have a therapeutic potential for the treatment of CRC.
Biomedicine & Pharmacotherapy | 2017
Yuanle Deng; Yali Li; Fangfang Yang; Anqi Zeng; Shuping Yang; Yi Luo; Yiwen Zhang; Yongmei Xie; Tinghong Ye; Yong Xia; Wenya Yin
Prostate cancer is a big threat to male for its poor prognosis and high mortality rate. Natural compounds are important resources of many anticancer drugs. Pomegranate is a kind of antioxidant-rich fruit and its peel and seed has potential anticancer activities. In this study, we aimed to investigate the effects of pomegranate peel extract (PoPx) on the apoptosis and metastasis of prostate cancer cells and the related mechanism. We found that PoPx showed growth inhibition on prostate cancer cells. Nuclei morphological and flow cytometer (FCM) analysis indicated that PoPx could induce prostate cancer apoptosis. Further investigation indicated that mitochondrial mediated intrinsic pathway is involved in the apoptosis. Exposure to PoPx led to loss of mitochondrial transmembrane potential (Δym), accumulation of reactive oxygen species (ROS). Western blot analysis showed that PoPx could increase the expression ratio of Bax/Bcl2 and activation of apoptosis executor caspase 3. Wound healing assay and transwell migration and invasion assay implied that PoPx has the potential to inhibit migration and invasion, two critical steps in prostate cancer metastasis. Downregulation of MMP2/MMP9 and upregulation of TIMP2 showed accordance with the inhibition of migration and invasion. In summary, the present data showed that PoPx could be a promising drug candidate to treat prostate cancer, showing us a better way to develop novel drugs from natural compounds.
Biomedicine & Pharmacotherapy | 2017
Kai Yu; Tingting Wang; Yujue Li; Chun Wang; Xia Wang; Mei Zhang; Yongmei Xie; S. Li; Zhenmei An; Tinghong Ye
The morbidity of thyroid cancer has been rising obviously throughout the world during the past years. Classic treatment procedure is generally curable for low risk differentiated thyroid cancer, but may lead to many postoperative complications. And low-level of thyroid stimulating hormone after surgery has side effects on both cardiovascular system and skeletal system. Furthermore effective treatment approaches for more aggressive differentiated thyroid cancer, poorly differentiated thyroid cancer and anaplastic thyroid cancer are absent, thus new candidates that can inhibit tumor growth and metastasis are urgently needed. In this study, niclosamide, an FDA approved anthelminthic drug, was evaluated for its anti-thyroid cancer activity in vitro. Niclosamide potently inhibited cell proliferation and induced apoptosis in human papillary thyroid cancer cell lines TPC-1 and BCPAP, as well as anaplastic thyroid cancer cell line ACT-1. In addition, the occurrence of TPC-1 apoptosis was correlated with activation of Bax and cleaved caspases-3, and inhibition of Bcl-2 and the mitochondrial membrane potential (ΔYm), indicating that niclosamide may induce apoptosis through a mitochondria-mediated intrinsic apoptotic pathway. Moreover, niclosamide markedly impaired TPC-1 cells and ACT-1 cells invasion. And we further found the inhibitory effect of TPC-1 was closely related with down-regulating of matrix metalloproteinase (MMP)-2 and -9 and up-regulating of tissue inhibitor of metalloproteinase (TIMP)-2. Taken together, these results demonstrated that niclosamide could be a potential agent for inhibiting the growth and metastasis of thyroid cancer.