Network


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

Hotspot


Dive into the research topics where Yinghui Xu is active.

Publication


Featured researches published by Yinghui Xu.


Journal of the National Cancer Institute | 2010

Essential Role of TRPC6 Channels in G2/M Phase Transition and Development of Human Glioma

Xia Ding; Zhuohao He; Kechun Zhou; Ju Cheng; Hailan Yao; Dongliang Lu; Rong Cai; Yening Jin; Bin Dong; Yinghui Xu; Yizheng Wang

BACKGROUND Patients with glioblastoma multiforme, the most aggressive form of glioma, have a median survival of approximately 12 months. Calcium (Ca(2+)) signaling plays an important role in cell proliferation, and some members of the Ca(2+)-permeable transient receptor potential canonical (TRPC) family of channel proteins have demonstrated a role in the proliferation of many types of cancer cells. In this study, we investigated the role of TRPC6 in cell cycle progression and in the development of human glioma. METHODS TRPC6 protein and mRNA expression were assessed in glioma (n = 33) and normal (n = 17) brain tissues from patients and in human glioma cell lines U251, U87, and T98G. Activation of TRPC6 channels was tested by platelet-derived growth factor-induced Ca(2+) imaging. The effect of inhibiting TRPC6 activity or expression using the dominant-negative mutant TRPC6 (DNC6) or RNA interference, respectively, was tested on cell growth, cell cycle progression, radiosensitization of glioma cells, and development of xenografted human gliomas in a mouse model. The green fluorescent protein (GFP) and wild-type TRPC6 (WTC6) were used as controls. Survival of mice bearing xenografted tumors in the GFP, DNC6, and WTC6 groups (n = 13, 15, and 13, respectively) was compared using Kaplan-Meier analysis. All statistical tests were two-sided. RESULTS Functional TRPC6 was overexpressed in human glioma cells. Inhibition of TRPC6 activity or expression attenuated the increase in intracellular Ca(2+) by platelet-derived growth factor, suppressed cell growth and clonogenic ability, induced cell cycle arrest at the G2/M phase, and enhanced the antiproliferative effect of ionizing radiation. Cyclin-dependent kinase 1 activation and cell division cycle 25 homolog C expression regulated the cell cycle arrest. Inhibition of TRPC6 activity also reduced tumor volume in a subcutaneous mouse model of xenografted human tumors (P = .014 vs GFP; P < .001 vs WTC6) and increased mean survival in mice in an intracranial model (P < .001 vs GFP or WTC6). CONCLUSIONS In this preclinical model, TRPC6 channels were essential for glioma development via regulation of G2/M phase transition. This study suggests that TRPC6 might be a new target for therapeutic intervention of human glioma.


Oncology Reports | 2011

β-elemene inhibits proliferation of human glioblastoma cells through the activation of glia maturation factor β and induces sensitization to cisplatin

Tingzhun Zhu; Yinghui Xu; Bin Dong; Jianing Zhang; Zhenqing Wei; Yousong Xu; Yiqun Yao

β-elemene, a natural drug extracted from Curcuma wenyujin, strongly inhibits glioblastoma growth. However, the mechanism of β-elemene antitumor action remains unclear. Glia maturation factor β (GMFβ) regulates cellular growth, fission, differentiation and apoptosis. It has been reported that overexpression of GMFβ inhibits the growth of glioblastoma cells and decreases tumor volume. To illustrate the role of GMFβ in the anti-proliferative effect of β-elemene in glioblastoma, U87 cells were treated with β-elemene at various doses and for different periods of time, and levels of phospho-GMFβ (p-GMFβ) and total GMFβ were determined by immunoprecipitation and western blot analysis. Upon GMFβ silencing using RNA interference, the antitumor action of β-elemene was evaluated in a methyl thiazolyl tetrazolium assay and by semi-quantitative western blot analysis of MKK3/6 and p-MKK3/6 expression. Finally, chemosensitization to cisplatin by β-elemene was examined using a cell counting array, and the cell growth inhibitory rate was calculated. The results showed that β-elemene inhibits U87 cell viability through the activation of the GMFβ signaling pathway. Conversely, silencing the expression of GMFβ reversed the antitumor effect of β-elemene and impaired the phosphorylation of MKK3/6. Furthermore, β-elemene increased the sensitivity of U87 glioblastoma cells to the chemotherapeutic agent cisplatin. Taken together, these results suggest that activation of the GMFβ pathway mediates the antitumor effect of β-elemene in glioblastoma. GMFβ is a putative molecular target for glioblastoma therapy.


International Journal of Oncology | 2011

ß-Elemene inhibits proliferation of human glioblastoma cells and causes cell-cycle G0/G1 arrest via mutually compensatory activation of MKK3 and MKK6

Ting-Zhun Zhu; Yongshun Zhao; Jianing Zhang; Longjie Li; Li-Juan Zou; Yiqun Yao; Yinghui Xu

ß-elemene, a natural plant drug extracted from Curcuma wenyujin, has shown a strong anti-glioblastoma effect. However, the antitumor mechanism of ß-elemene remains unclear. Mitogen-activated protein kinase kinase-3 (MKK3) and -6 (MKK6) can regulate cellular growth, fission, differentiation and apoptosis. To illustrate the role of MKK3 and MKK6 in the anti-glioblastoma proliferation effect of ß-elemene, U87 cells were treated with ß-elemene at various doses or for different times, and then phosphorylated MKK3 (p-MKK3), phosphorylated MKK6 (p-MKK6), MKK3 and MKK6 were detected by Western blot assay. After transient transfection with dominant-negative mutant plasmids of MKK3 and MKK6, cell viability and cell cycle stage were determined by methyl thiazolyl tetrazolium assay and flow cytometry, respectively. Results showed that ß-elemene inhibited the proliferation of U87 glioblastoma cells and arrested them in G0/G1 phase through up-regulating p-MKK3 and p-MKK6 levels. In contrast, inhibition of MKK3 and MKK6 reversed the antitumor effect of ß-elemene. Furthermore, when either MKK3 or MKK6 was inhibited by a dominant-negative plasmid, the other was compensatorily activated in the presence of ß-elemene. Taken together, our findings indicate that mutually compensatory activation of MKK3 and MKK6 mediates the anti-glioblastoma effect of ß-elemene. MKK3 and MKK6 might be two putative targets for molecular therapy against glioblastoma.


Journal of Neuro-oncology | 2012

β-Elemene inhibits Hsp90/Raf-1 molecular complex inducing apoptosis of glioblastoma cells

Yongshun Zhao; Ting-Zhun Zhu; Yanwei Chen; Yiqun Yao; Chunming Wu; Zhenqing Wei; Wei Wang; Yinghui Xu

Abstractβ-Elemene, an active component of herb medicine Curcuma wenyujin, has been shown to antagonize glioblastoma cells by inducing apoptosis. However, how β-elemene induces apoptosis of these cells remains unclear. In this study, we report that β-elemene disrupted the formation of the Hsp90/Raf-1 complex, a key step in maintaining the conformation stability of Raf-1, and caused deactivation of Raf-1 and inhibition of the ERK pathway, thereby leading to apoptosis of glioblastoma cells. Specifically, treatment of glioblastoma cell lines with β-elemene attenuated phosphorylation of multiple members of the kinase families in the Ras/Raf/MEK/ERK cascade, including Raf-1 and ERK as well as downstream signaling targets such as Bcl-2. These results suggest that the Hsp90/Raf-1 complex could be a promising molecular target for new drug development for the treatment of glioblastoma.


Cancer Biotherapy and Radiopharmaceuticals | 2012

Down-Regulation of Survivin and Hypoxia-Inducible Factor-1α by β-elemene Enhances the Radiosensitivity of Lung Adenocarcinoma Xenograft

Guoquan Li; Bingbing Xie; Xiaolong Li; Yinghai Chen; Qi Wang; Yinghui Xu; Meng Xu-Welliver; Lijuan Zou

Elemene (1-methyl-1-vinyl-2,4-diisopropenyl-cyclohexane) is a naturally occurring compound that can be isolated from the traditional Chinese medicinal herb Curcuma wenyujin. β-elemene, its active component, has recently been demonstrated to enhance the radiosensitivity of human cancer cell lines in vitro and of one animal tumor in vivo. The underlying mechanism, however, is still unclear. In this study, we demonstrated for the first time that β-elemene significantly improves the radiosensitivity of A549 lung adenocarcinoma xenograft in vivo as measured by tumor regrowth delay experiments. Our results showed that β-elemene, at 45 mg/kg, significantly inhibited radiation-induced expression of survivin and hypoxia-inducible factor (HIF)-1 α proteins. Because HIF-1 α is known to regulate survivin transcription and acts as upstream regulator of survivin, it is possible that β-elemene regulates the transcription of survivin through HIF-1 α. Our study suggests that β-elemene is a promising drug to enhance tumor radioresponse, and survivin and HIF-1 α are novel targets of β-elemene.


Journal of Cell Science | 2015

Crucial role of TRPC6 in maintaining the stability of HIF-1α in glioma cells under hypoxia

Shanshan Li; Jinkui Wang; Yi Wei; Yongjian Liu; Xia Ding; Bin Dong; Yinghui Xu; Yizheng Wang

ABSTRACT Hypoxia-inducible factor-1 (HIF-1) is a key transcription factor responsible for the expression of a broad range of genes that facilitate acclimatization to hypoxia. Its stability is predominantly controlled by rapid hydroxylation of two proline residues in its α-subunit. However, how the rapid hydroxylation of HIF-1α is regulated is not fully understood. Here, we report that transient receptor potential canonical (TRPC) 6 channels control hydroxylation and stability of HIF-1α in human glioma cells under hypoxia. TRPC6 was rapidly activated by IGF-1R–PLCγ–IP3R pathway upon hypoxia. Inhibition of TRPC6 enhanced the levels of α-ketoglutarate and promoted hydroxylation of HIF-1α to suppress HIF-1α accumulation without affecting its transcription or translation. Dimethyloxalylglycine N-(methoxyoxoacetyl)-glycine methyl ester (DMOG), an analog of α-ketoglutarate, reversed the inhibition of HIF-1α accumulation. Moreover, TRPC6 regulated GLUT1 (also known as SLC2A1) expression in a manner that was dependent on HIF-1α accumulation to affect glucose uptake during hypoxia. Our results suggest that TRPC6 regulates metabolism to affect HIF-1α stability and consequent glucose metabolism in human glioma cells under hypoxia. Summary: TRPC6 regulates metabolism to affect HIF-1α stability and consequent glucose metabolism in human glioma cells under hypoxia.


Cellular Physiology and Biochemistry | 2015

DNA Damage-Induced NF-κB Activation in Human Glioblastoma Cells Promotes miR-181b Expression and Cell Proliferation

Ruixue Xu; Rong-Yao Liu; Chunming Wu; Yongshun Zhao; Yang Li; Yiqun Yao; Yinghui Xu

Background: Glioblastoma (GBM) is the most common and most aggressive form of brain cancer. After surgery, radiotherapy is the mainstay of treatment for GBM patients. Unfortunately, the vast majority of GBM patients fail responding to radiotherapy because GBM cells remain highly resistant to radiation. Radiotherapy-induced DNA damage response may correlate with therapeutic resistance. Methods: Ionizing radiation (IR) was used to induce DNA damage. Cell proliferation and migration were detected by wound-healing, MTT and apoptosis assays. Dual-luciferase assays and Western blot analysis were performed to evaluate NF-κB activation and validate microRNA targets. Real-time PCR was used to study mRNA and microRNA levels. Results: IR-induced DNA damage activated NF-κB in GBM cells which promoted expression of IL-6, IL-8 and Bcl-xL, thereby contributing to cell survival and invasion. Knockdown SENP2 expression enhanced NF-κB essential modulator (NEMO) SUMOylation and NF-κB activity following IR exposure. miR-181b targets SENP2 and positively regulated NF-κB activity. Conclusion: NF-κB activation by DNA damage in GBM cells confers resistance to radiation-induced death.


Oncology Reports | 2015

Downregulation of peroxiredoxin-1 by β-elemene enhances the radiosensitivity of lung adenocarcinoma xenografts.

Guoquan Li; Bingbing Xie; Xiaolong Li; Yinghai Chen; Yinghui Xu; Meng Xu-Welliver; Lijuan Zou

β-elemene, the active component of elemene (1-methyl-1-vinyl-2,4-diisopropenyl-cyclohexane), is a naturally occurring compound isolated from the traditional Chinese medicinal herb Curcuma wenyujin. Studies have confirmed that β-elemene enhances the radiosensitivity of lung cancer cell lines such as A549, by multiple pathways; however, their underlying mechanisms and pathways are yet to be elucidated. In the present study, two-dimensional differential in-gel electrophoresis and matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometry were used to profile the different proteins in A549 cell xenograft models of both treatment groups. The protein/mRNA expression was assessed by reverse transcription-polymerase chain reaction and western blotting techniques in tumor samples from all treatment groups. As a critical player in redox regulation of cancer cells, inhibition of peroxiredoxin-1 (Prx-1) may be an effective option for enhancing the tumor response to radiation. We further verified Prx-1 expression at the transcription and translation levels. β-elemene at a dose of 45 mg/kg had little effect on the Prx-1 protein expression, which was correlated with a moderate antitumor effect. However, a 45 mg/kg dose of β-elemene significantly inhibited the Prx-1 mRNA expression, thereby suggesting a possible influence on the transcriptional process, and radiation significantly increased the Prx-1 mRNA/protein expression compared to the control group (p<0.01). Notably, Prx-1 mRNA/protein expression was significantly lower in the β-elemene/radiation co-treatment group compared to the baseline levels in the control group (p<0.01). These results suggest that radiation-induced Prx-1 expression is directly or indirectly suppressed by β-elemene, thus suggesting a new pathway by which to reverse radioresistance.


Molecular Medicine Reports | 2015

Naringin ameliorates cognitive deficits via oxidative stress, proinflammatory factors and the PPARγ signaling pathway in a type 2 diabetic rat model

Zhonghua Qi; Yinghui Xu; Zhanhua Liang; Sheng Li; Jie Wang; Yi Wei; Bin Dong

Naringenin is a flavonoid polyphenolic compound, which facilitates the removal of free radicals, oxidative stress and inflammation. The present study aimed to obtain a better understanding of the effects of curcumin on the regulation of diabetes‑associated cognitive decline, and its underlying mechanisms. An experimental diabetes mellitus (DM) rat model was induced by streptozoticin (50 mg/kg). Following treatment with naringin (100 and 200 mg/kg) for 16 weeks, the body weight and blood glucose levels of the DM rats were measured. A morris water maze test was used to analyze the effects of naringin on the cognitive deficit of the DM rats. The levels of oxidative stress, proinflammatory factors, caspase‑3 and caspase‑9, and the protein expression of peroxisome proliferator‑activated receptor γ (PPARγ) were quantified in the DM rats using a commercially‑available kit and western blot assay, respectively. In addition, a GW9662 PPARγ inhibitor (0.3 mg/kg) was administered to the DM rats to determine whether PPARγ affected the effects of naringin on the cognitive deficit of the DM rats. The results demonstrated that naringin increased the body weight, blood glucose levels, and cognitive deficits of the DM rats. The levels of oxidative stress and proinflammatory factors in the naringin‑treated rats were significantly lower, compared with those of the DM rats. In addition, naringin activated the protein expression of PPARγ, and administration of the PPARγ inhibitor decreased the protein expression of PPARγ, and attenuated the effects of naringin on cognitive deficit. The results also demonstrated that naringin decreased the expression levels of caspase‑3 and caspase‑9 in the DM rats. These results suggested that naringin ameliorated cognitive deficits via oxidative stress, proinflammatory factors and the PPARγ signaling pathway in the type 2 diabetic rat model. Furthermore, oxidative stress, proinflammatory factors and PPARγ signaling may be involved in mediating these effects.


Scientific Reports | 2016

RNF135, RING finger protein, promotes the proliferation of human glioblastoma cells in vivo and in vitro via the ERK pathway

Yongjian Liu; Feng Wang; Yongsheng Liu; Yiqun Yao; Xiupeng Lv; Bin Dong; Jun Li; Siyang Ren; Yiwen Yao; Yinghui Xu

Ring finger protein 135 (RNF135), located on chromosome 17q11.2, is a RING finger domain-containing E3 ubiquitin ligase that was identified as a bio-marker and therapy target of glioblastoma. In our study, we confirmed that RNF135 was up-regulated in glioblastoma tissues compared with normal brain (NB) tissues, and that RNF135 knockdown inhibited proliferation and migration and led to cell cycle arrest in the G0/G1 phase in vivo. By lowering RNF135 expression, phosphorylated Erk and cell cycle protein CDK4 were down-regulated, while p27Kip1 and p21Waf1/Cip1 were up-regulated in U87 and U251 cells in vitro. In addition, using the immunofluorescence double labelling method, we found that RNF135 and P-Erk were co-localized in the cytoplasm and were highly expressed in glioblastoma samples compared with NB tissues. Moreover, the growth of U87 cell-transplanted tumours in nude mice was inhibited while transduced with Lv-shRNF135. Taken together, our findings demonstrate the biological effects of RNF135 in glioblastoma cell proliferation, migration and cell cycle, and its role in the progression of glioblastoma may be associated with the ERK signal transduction pathway.

Collaboration


Dive into the Yinghui Xu's collaboration.

Top Co-Authors

Avatar

Bin Dong

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Yiqun Yao

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Yongjian Liu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiupeng Lv

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jie Wang

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi Wei

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhenqing Wei

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Chunming Wu

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun Li

Dalian Medical University

View shared research outputs
Top Co-Authors

Avatar

Siyang Ren

Dalian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge