Network


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

Hotspot


Dive into the research topics where Xiangling Feng is active.

Publication


Featured researches published by Xiangling Feng.


Oncology | 2007

Epigenetic and Genetic Alterations of the EDNRB Gene in Nasopharyngeal Carcinoma

Liang Zhou; Xiangling Feng; Wenjiao Shan; Wen Zhou; Weidong Liu; Lei Wang; Bin Zhu; Hongmei Yi; Kaitai Yao; Caiping Ren

Background: Loss of heterozygosity (LOH) at 13q22 is a common event in nasopharyngeal carcinoma (NPC). EDNRB gene located at 13q22 has been demonstrated to be hypermethylated in some kinds of tumors. In the current study, we focused on the epigenetic and genetic alterations of EDNRB in NPC. Methods: The mRNA expression of EDNRB was detected by semiquantitative RT-PCR and real-time quantitative PCR in 49 NPC and 12 chronic nasopharyngitis biopsies. The methylation and LOH status of EDNRB were examined by methylation-specific polymerase chain reaction, microsatellite PCR and sequencing. We also examined the mRNA expression of EDNRB in four NPC cell lines after 5-aza-2′-deoxycytidine treatment. Results:EDNRB was downregulated in primary NPC tissues and NPC cell lines, and a relatively higher methylation level of EDNRB was found in NPC biopsies (84%) compared to that in chronic nasopharyngitis biopsies (42%). Treatment of NPC cell lines with 5-aza-2′-deoxycytidine activated EDNRB expression. LOH of EDNRB gene was also found at two microsatellite sites with ratios of 6.25 and 16.67% in NPC. Conclusion: Our results suggested that EDNRB expression may be affected by aberrant promoter methylation and gene deletion and may play a role in the development of NPC.


Medical Oncology | 2015

DLC-1 induces mitochondrial apoptosis and epithelial mesenchymal transition arrest in nasopharyngeal carcinoma by targeting EGFR/Akt/NF-κB pathway.

Wei Huang; Jie Liu; Xiangling Feng; Huan Chen; Liang Zeng; Guoling Huang; Weidong Liu; Lei Wang; Wei Jia; Jiawen Chen; Caiping Ren

Loss of deleted in liver cancer-1 (DLC-1) can induce apoptosis and inhibit the mobility, migration and metastasis in several cancers. Previously, we revealed that ectopic expression of DLC-1 can suppress proliferation, mobility, migration and tumorigenesis in nasopharyngeal carcinoma (NPC). However, the molecular mechanisms accounting for the roles of DLC-1 in NPC are still obscure. In the present work, we attempted to study and uncover the mechanisms underlying the functions of DLC-1 in NPC. The apoptosis of 5-8F-DLC-1 cells, established previously, was analyzed by mitochondrial membrane potentials assay and flow cytometer analysis. And the antibodies involving pathways such as mitochondrial-associated apoptosis, epithelial mesenchymal transition and metastasis were applied to detect and compare the expression level of targeted proteins. The obvious apoptosis of 5-8F-DLC-1 cells was observed reflected by mitochondrial depolarization and lower ratio in cell viability. Subsequently, the activation of mitochondrial apoptosis was verified by the increased expressions of Bax, Apaf1, cleave-caspases and cleave-PARP, etc, and the decreased expressions of Bcl-2, Bcl-xL, Mcl-1, Survivin, etc, in 5-8F-DLC-1 cells. Then, the inhibited epithelial mesenchymal transition of 5-8F-DLC-1 cells was validated by upregulated expression of E-cadherin and downregulated expression of N-cadherin, Snail, Vimentin. Subsequently, downregulated expressions of proteins such as FAK, RhoA, ROCK1 and cdc25 related to cell adhesion and cytoskeleton organization were also observed. And expressions of MMPs were inhibited in 5-8F-DLC-1 cells. At last, the inhibited activity of EGFR/Akt/NF-κB axis was revealed by the decreased expressions of phosho-EGFR, phosho-Akt, phosho-p38MAPK, phosho-IKKα and phosho-p65. Here, we systematically explored the mechanisms underlying the negative roles of DLC-1 in NPC cells. For the first time, we confirmed that the ectopic expression DLC-1 can induce mitochondrial apoptosis, inhibit EMT and related processes by targeting the EGFR/Akt/NF-κB pathway, which, beyond all doubt, offered beneficial guidelines for other studies and laid a good foundation for its clinical applications ultimately.


International Journal of Oncology | 2018

High COX-2 expression contributes to a poor prognosis through the inhibition of chemotherapy-induced senescence in nasopharyngeal carcinoma

Chen Shi; Yongjun Guan; Liang Zeng; Guizhu Liu; Yinghong Zhu; He Xu; Yichen Lu; Jiabin Liu; Jiaojiao Guo; Xiangling Feng; Xinying Zhao; Weihong Jiang; Guancheng Li; Guiyuan Li; Yun Dai; Fengyan Jin; Wei Li; Wen Zhou

Resistance to radiotherapy and chemotherapy currently represents one of the major reasons for therapeutic failure in nasopharyngeal carcinoma (NPC). However, the mechanisms underlying resistance to chemotherapy in NPC remain unclear. In this study, cell counting assay, cell cycle assay and senescence associated β-galactosidase activity were performed to evaluate cell growth, proliferation and senescence, respectively. We found that the aberrant expression of cyclooxygenase-2 (COX-2) was associated with a poor outcome and recurrance in patients with NPC. In NPC cells, COX-2 overexpression increased cell proliferation, inhibited cellular senescence and resulted in chemoresistance, while the knockdown of COX-2 reduced cell proliferation, promoted cellular senescence and overcame chemoresistance. Furthermore, fibroblasts from COX-2 knockout mice exhibited cellular senescence, particularly when treated with chemotherapeutic agents. Mechanistically, COX-2 interacted with p53 protein and inhibited cellular senescence, which resulted in chemotherapeutic resistance. On the whole, these findings indicate that COX-2 may play a critical role in chemotherapeutic resistance in NPC via the inhibition of chemotherapy-induced senescence via the inactivation of p53. This study provides experimental evidence for the preclinical value of increasing chemotherapy-induced senescence by targeting COX-2 as an effective antitumor treatment in patients with recurrent NPC.


Nature Genetics | 2001

Cloning and analyzing 5′ flanking sequences of a tissue-specifically expressed gene derived from mouse nasopharynx

Ling Zhang; Kaitai Yao; Ke Lan; Hong Li; Xiangling Feng; Zhiwei He; Weinong Han

Cloning and analyzing 5′ flanking sequences of a tissue-specifically expressed gene derived from mouse nasopharynx


Journal of Cellular Biochemistry | 2018

High NEK2 confers to poor prognosis and contributes to cisplatin-based chemotherapy resistance in nasopharyngeal carcinoma: XU et al.

He Xu; Liang Zeng; Yongjun Guan; Xiangling Feng; Yinghong Zhu; Yichen Lu; Chen Shi; Shilian Chen; Jiliang Xia; Jiaojiao Guo; Chunmei Kuang; Wei Li; Fengyan Jin; Wen Zhou

Nasopharyngeal carcinoma (NPC) is a common malignant tumor in southern China and Southeast Asia, but the molecular mechanism of its pathogenesis is poorly understood. Our previous work demonstrated that NEK2 is overexpressed in multiple cancers. However, how NEK2 involves in NPC development remains to be elucidated. In this study, we firstly identified NEK2, located at +1q32‐q33, a late event in NPC pathogenesis, overexpressed in the stage III‐IV and paired sequential recurrent patients with NPC by immunohistochemistry. Furthermore, Kaplan‐Meier analysis indicated high NEK2 conferred an inferior overall survival in NPC. In addition, cisplatin experiments with cell counting kit‐8, colony formation, and a xenograft mice model of NPC demonstrated that NEK2 contributed to proliferation and cisplatin resistance in vitro and in vivo. On the contrary, downregulation of NEK2 by short hairpin RNA inhibited NPC cell growth and increased the sensitivity of cisplatin treatment in vitro. Thus, increased expression of NEK2 protein could not be predicted for poor survival but used as a novel biomarker for recurrence of NPC. Targeting NEK2 has the potential to eradicate the cisplatin‐based chemotherapy resistant NPC cells.


Acta Biochimica et Biophysica Sinica | 2009

Inactivation of LARS2, located at the commonly deleted region 3p21.3, by both epigenetic and genetic mechanisms in nasopharyngeal carcinoma

Wen Zhou; Xiangling Feng; Hong Li; Lei Wang; Bin Zhu; Weidong Liu; Ming Zhao; Kaitai Yao; Caiping Ren


Biochemical and Biophysical Research Communications | 2007

Transcriptional profiling of human embryonic stem cells and embryoid bodies identifies HESRG, a novel stem cell gene

Ming Zhao; Caiping Ren; Hong Yang; Xiangling Feng; Xingjun Jiang; Bin Zhu; Wen Zhou; Lei Wang; Ying Zeng; Kaitai Yao


Biochemical and Biophysical Research Communications | 2000

Characterization of the functionally related sites in the neural inducing gene noggin.

Weidong Liu; Caiping Ren; Jianling Shi; Xiangling Feng; Zhiwei He; Liangguo Xu; Ke Lan; Lu Xie; Ying Peng; Jing Fan; Hsiang-Fu Kung; Kaitai Yao; Ren-He Xu


Acta Biochimica et Biophysica Sinica | 2005

Critical Role of Cys168 in Noggin Protein's Biological Function

Weidong Liu; Xiangling Feng; Caiping Ren; Jianling Shi; Xu-Yu Yang; Ming Zhao; Liang Zhou; Ke Lan; Kaitai Yao


Acta Biochimica et Biophysica Sinica | 2003

[Construction of N-LMP1 transgenic mice with the specific regulation region in nasopharynx].

Zhang L; Ke Lan; Xiangling Feng; Qiao Gl; Shen Xm; Shi Ym; Hong Li; Kaitai Yao

Collaboration


Dive into the Xiangling Feng's collaboration.

Top Co-Authors

Avatar

Kaitai Yao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Caiping Ren

Central South University

View shared research outputs
Top Co-Authors

Avatar

Weidong Liu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Lei Wang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Hong Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Ke Lan

Central South University

View shared research outputs
Top Co-Authors

Avatar

Wen Zhou

Central South University

View shared research outputs
Top Co-Authors

Avatar

Bin Zhu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Liang Zeng

Central South University

View shared research outputs
Top Co-Authors

Avatar

Ming Zhao

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge