Network


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

Hotspot


Dive into the research topics where Zhijie Xu is active.

Publication


Featured researches published by Zhijie Xu.


International Journal of Nanomedicine | 2013

Delivery system for DNAzymes using arginine-modified hydroxyapatite nanoparticles for therapeutic application in a nasopharyngeal carcinoma model.

Yan Chen; Lifang Yang; Suping Huang; Zhi Li; Lu Zhang; Jiang He; Zhijie Xu; Liyu Liu; Ya Cao; Lunquan Sun

DNAzymes are synthetic, single-stranded, catalytic nucleic acids that bind and cleave target mRNA in a sequence-specific manner, and have been explored for genotherapeutics. One bottleneck restricting their application is the lack of an efficient delivery system. As an inorganic nanomaterial with potentially wide application, nano-hydroxyapatite particles (nHAP) have attracted increasing attention as new candidates for nonviral vectors. In this study, we developed an nHAP-based delivery system and explored its cellular uptake mechanisms, intracellular localization, and biological effects. Absorption of arginine-modified nanohydroxyapatite particles (Arg-nHAP) and DZ1 (latent membrane protein 1 [LMP1]-targeted) reached nearly 100% efficiency under in vitro conditions. Using specific inhibitors, cellular uptake of the Arg-nHAP/DZ1 complex was shown to be mediated by the energy-dependent endocytosis pathway. Further, effective intracellular delivery and nuclear localization of the complex was confirmed by confocal microscopy. Biologically, the complex successfully downregulated the expression of LMP1 in nasopharyngeal carcinoma cells. In a mouse tumor xenograft model, the complex was shown to be delivered efficiently to tumor tissue, downregulating expression of LMP1 and suppressing tumor growth. These results suggest that Arg-nHAP may be an efficient vector for nucleic acid-based drugs with potential clinical application.


Acta Biochimica et Biophysica Sinica | 2015

The receptor proteins: pivotal roles in selective autophagy

Zhijie Xu; Lifang Yang; San Xu; Zhibao Zhang; Ya Cao

Autophagy is a highly regulated and multistep biological process whereby cells under metabolic, proteotoxic, or other stresses remove dysfunctional organelles and/or misfolded/polyubiquitinated proteins by shuttling them via specialized structures called autophagosomes to the lysosome for degradation. Although autophagy is generally considered to be a non-selective process, accumulating evidence suggests that it can also selectively degrade specific target cargoes. These selective targets include proteins, mitochondria, and even invading bacteria. The discovery and characterization of autophagic adapters, such as p62/Sequestosome 1 (SQSTM1) and Neighbor of BRCA1 gene 1 (NBR1), have provided mechanistic insights into selective autophagy. These receptors are all able to act as cargo receptors for the degradation of ubiquitinated substrates. This review mainly summarizes the most up-to-date findings regarding the key receptor proteins that play important roles in regulating selective autophagy.


Cancer Biology & Therapy | 2014

Targeting EBV-LMP1 DNAzyme enhances radiosensitivity of nasopharyngeal carcinoma cells by inhibiting telomerase activity

Lifang Yang; Zhijie Xu; Liyu Liu; Xiangjian Luo; Jingchen Lu; Lunquan Sun; Ya Cao

The latent membrane protein 1 (LMP1), which is encoded by the Epstein–Barr virus (EBV), has been suggested to be one of the major oncogenic factors in nasopharyngeal carcinoma (NPC). In previous studies, we experimentally demonstrated that downregulation of LMP1 expression by targeting EBV-LMP1 DNAzyme (Dz1) could increase the radiosensitivity of NPC. However, how Dz1 contributes to the radiosensitivity in NPC is still not clear. In the present study, we confirmed that Dz1 decreases LMP1 expression and downregulates the expression of the catalytic subunit of telomerase (hTERT), both at the protein and mRNA levels (P < 0.01), and therefore, consequently inhibits telomerase activity (P < 0.05) in LMP1-positive NPC cells. We also observed that LMP1 mediated Akt phosphorylation is involved in the regulation of hTERT expression and phosphorylation. After LMP1 and hTERT expression was silenced by Dz1 and hTERT-targeted siRNA, respectively, the radiosensitivity increased in CNE1-LMP1 cells (P < 0.05). The inhibition was more significant after Dz1 treatment was combined with siRNA (P < 0.01). We concluded that hTERT expression and phosphorylation could be regulated by LMP1 through the Akt pathway, and Dz1 enhances radiosensitivity of LMP1-positive NPC cells by inhibiting telomerase activity.


Cancer Letters | 2016

EBV-LMP1 suppresses the DNA damage response through DNA-PK/AMPK signaling to promote radioresistance in nasopharyngeal carcinoma.

Jingchen Lu; Min Tang; Hongde Li; Zhijie Xu; Xinxian Weng; Jiangjiang Li; Xinfang Yu; Luqing Zhao; Hongwei Liu; Yongbin Hu; Zheqiong Tan; Lifang Yang; Meizuo Zhong; Jian Zhou; Jia Fan; Ann M. Bode; Wei Yi; Jinghe Gao; Lunquan Sun; Ya Cao

We conducted this research to explore the role of latent membrane protein 1 (LMP1) encoded by the Epstein-Barr virus (EBV) in modulating the DNA damage response (DDR) and its regulatory mechanisms in radioresistance. Our results revealed that LMP1 repressed the repair of DNA double strand breaks (DSBs) by inhibiting DNA-dependent protein kinase (DNA-PK) phosphorylation and activity. Moreover, LMP1 reduced the phosphorylation of AMP-activated protein kinase (AMPK) and changed its subcellular location after irradiation, which appeared to occur through a disruption of the physical interaction between AMPK and DNA-PK. The decrease in AMPK activity was associated with LMP1-mediated glycolysis and resistance to apoptosis induced by irradiation. The reactivation of AMPK significantly promoted radiosensitivity both in vivo and in vitro. The AMPKα (Thr172) reduction was associated with a poorer clinical outcome of radiation therapy in NPC patients. Our data revealed a new mechanism of LMP1-mediated radioresistance and provided a mechanistic rationale in support of the use of AMPK activators for facilitating NPC radiotherapy.


Molecular Medicine | 2013

Antiangiogenic and antitumoral effects mediated by a vascular endothelial growth factor receptor 1 (VEGFR-1)-targeted DNAzyme.

Liangfang Shen; Qin Zhou; Ying Wang; Weihua Liao; Yan Chen; Zhijie Xu; Lifang Yang; Lunquan Sun

Antiangiogenesis is a promising antitumor strategy that inhibits tumor vascular formation to suppress tumor growth. DNAzymes are synthetic single-strand deoxyribonucleic acid (DNA) molecules that can cleave ribonucleic acids (RNAs). Here, we conducted a comprehensive in vitroselection of active DNAzymes for their activity to cleave the vascular endothelial growth factor receptor (VEGFR-1) mRNA and screened for their biological activity in a matrigel tube-formation assay. Among the selected DNAzymes, DT18 was defined as a lead molecule that was further investigated in several model systems. In a rat corneal vascularization model, DT18 demonstrated significant and specific antiangiogenic activity, as evidenced by the reduced area and vessel number in VEGF-induced corneal angiogenesis. In a mouse melanoma model, DT18 was shown to inhibit B16 tumor growth, whereas it did not affect B16 cell proliferation. We further assessed the DT18 effect in mice with established human nasopharyngeal carcinoma (NPC). A significant inhibition of tumor growth was observed, which accompanied downregulation of VEGFR-1 expression in NPC tumor tissues. To evaluate DT18 effect on vasculature, we performed dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) on the human NPC xenograft mice treated with DT18 and showed a reduction of the parameter of Ktrans (volume constant for transfer of contrast agent), which reflects the condition of tumor microvascular permeability. When examining the safety and tolerability of DT18, intravenous administration of Dz18 to healthy mice caused no substantial toxicities, as shown by parameters such as body weight, liver/kidney function, and histological and biochemical analyses. Taken together, our data suggest that the anti-VEGFR-1DNAzyme may be used as a therapeutic agent for the treatment of cancer, such as NPC.


International Journal of Oncology | 2013

EBV-LMP1-targeted DNAzyme induces DNA damage and causes cell cycle arrest in LMP1-positive nasopharyngeal carcinoma cells.

Xiaoqian Ma; Zhijie Xu; Lifang Yang; Lanbo Xiao; Min Tang; Jingchen Lu; San Xu; Yiping Tang; Xinxian Wen; Xingming Deng; Lunquan Sun; Ya Cao

This study aimed to determine the molecular mechanisms underlying the effect of the LMP1-targeted DNAzyme 1 (DZ1) on cell cycle progression in nasopharyngeal carcinoma (NPC) cells. We showed that the active DZ1 inhibited the expression of latent membrane protein 1 (LMP1) and induced a G1 phase arrest. In addition, this cell cycle deregulation was shown to be accompanied by upregulation of the DNA damage marker γ-H2AX, downregulation of the DNA damage response factor p-p53-Ser15 and cell proliferation inhibition. To investigate what affected the cell cycle progression, we examined the expression of two checkpoint-related cyclins and cyclin-dependent kinases (CDKs). We found a decrease of cyclin D1 and cyclin E protein levels at 24 h from the DZ1 treatment. Moreover, we observed inhibition of CDK4 activity and decreased cyclin D1 expression in the complexes immunoprecipitated with CDK4 antibody. We also found a reduction in cdc2 phosphorylation at Thr161 which partially stands for the cdc2 kinase activity in DZ1-treated CNE1-LMP1 cells, although the downregulation of LMP1 expression had no effect on the cyclin B1 and cdc2 expression. Further, we analyzed changes in cdc2 kinase activity induced by DZ1 and found that the downregulation of the LMP1 expression resulted in a 5-fold reduction in cdc2 kinase activity in CNE1-LMP1. The data suggest that the downregulation of the LMP1 expression by DZ1 was able to induce DNA damage, which then further inhibited the cell proliferation and resulted in malfunction of cell cycle checkpoints that led to G1 phase arrest and the decrease in number of cells in G2/M phase.


Oncotarget | 2016

Grifolin inhibits tumor cells adhesion and migration via suppressing interplay between PGC1α and Fra-1/LSF-MMP2/CD44 axes

Xiangjian Luo; Namei Li; Juanfang Zhong; Zheqiong Tan; Ying Liu; Xin Dong; Can Cheng; Zhijie Xu; Hongde Li; Lifang Yang; Min Tang; Xinxian Weng; Wei Yi; Ji-Kai Liu; Ya Cao

Grifolin, a farnesyl phenolic compound isolated from the fresh fruiting bodies of the mushroom Albatrellus confluens, exhibits effective antitumor bioactivity in previous study of our group and other lab. In this study, we observed that grifolin inhibited tumor cells adhesion and migration. Moreover, grifolin reduced reactive oxygen species (ROS) production and caused cellular ATP depletion in high-metastatic tumor cells. PGC1α (Peroxisome proliferator-activated receptor γ, coactivator 1α) encodes a transcriptional co-activator involved in mitochondrial biogenesis and respiration and play a critical role in the maintenance of energy homeostasis. Interestingly, grifolin suppressed the mRNA as well as protein level of PGC1α. We further identified that MMP2 and CD44 expressions were PGC1α inducible. PGC1α can bind with metastatic-associated transcription factors: Fra-1 and LSF and the protein-protein interaction was attenuated by grifolin treatment. Overall, these findings suggest that grifolin decreased ROS generation and intracellular ATP to suppress tumor cell adhesion/migration via impeding the interplay between PGC1α and Fra-1 /LSF-MMP2/CD44 axes. Grifolin may develop as a promising lead compound for antitumor therapies by targeting energy metabolism regulator PGC1α signaling.


The International Journal of Biochemistry & Cell Biology | 2013

WITHDRAWN: miR-504 affects the radio-resistance in nasopharyngeal carcinoma by down-regulating the expression of nuclear respiratory factor 1

Luqing Zhao; Zheyu Hu; Min Tang; Bin Yan; Jingchen Lu; Lanbo Xiao; Zhijie Xu; Ya Cao

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.


Oncotarget | 2015

EBV-LMP1 targeted DNAzyme enhances radiosensitivity by inhibiting tumor angiogenesis via the JNKs/HIF-1 pathway in nasopharyngeal carcinoma

Lifang Yang; Liyu Liu; Zhijie Xu; Weihua Liao; Deyun Feng; Xin Dong; San Xu; Lanbo Xiao; Jingchen Lu; Xiangjian Luo; Min Tang; Ann M. Bode; Zigang Dong; Lunquan Sun; Ya Cao


Chinese Science Bulletin | 2012

Use of DNAzymes for cancer research and therapy

Zhijie Xu; Lifang Yang; Lunquan Sun; Ya Cao

Collaboration


Dive into the Zhijie Xu's collaboration.

Top Co-Authors

Avatar

Lifang Yang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Ya Cao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Lunquan Sun

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jingchen Lu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Min Tang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Liyu Liu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Xiangjian Luo

Central South University

View shared research outputs
Top Co-Authors

Avatar

Hongde Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Lanbo Xiao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Luqing Zhao

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge