Network


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

Hotspot


Dive into the research topics where Xiaonan Zhu is active.

Publication


Featured researches published by Xiaonan Zhu.


Cell Biochemistry and Biophysics | 2012

Clinical and Imaging Characteristics of Cerebral Schistosomiasis

Liquan Wu; Mingcan Wu; Daofeng Tian; Shijie Chen; Baohui Liu; Qianxue Chen; Junmin Wang; Qiang Cai; Baowei Ji; Long Wang; Shenqi Zhang; Dong Ruan; Xiaonan Zhu; Zhentao Guo

In recent years, there has been a trend for increased incidence of cerebral schistosomiasis. It is often misdiagnosed because of the diversity of clinical symptoms. We wished to explore clinical characteristics and imaging findings in cerebral schistosomiasis. We retrospectively analyzed clinical data, laboratory tests, CT, and MRI results in 11 patients with cerebral schistosomiasis. All patients had chronic cerebral schistosomiasis (five with epilepsy type, five with brain tumor type, and one patient with stroke type). All patients with brain tumor type were misdiagnosed as having gliomas. There were typical findings on CT and MRI. In conclusion, clinical manifestations of cerebral schistosomiasis are variable, and the rate of misdiagnosis is high. For more precise diagnosis, a combination of laboratory and imaging data is required.


Brain Research | 2013

BMP4 reverses multidrug resistance through modulation of BCL-2 and GDNF in glioblastoma.

Baohui Liu; Qianxue Chen; Daofeng Tian; Liquan Wu; Huimin Dong; Junmin Wang; Baowei Ji; Xiaonan Zhu; Qiang Cai; Long Wang; Shenqi Zhang

Patients with glioblastoma are commonly treated with chemotherapy. But a significant proportion of patients develop disease progression after an initial response to chemotherapy. Presently, there is no standard of care for such patients. The bone morphogenetic protein 4 (BMP4) has been reported to play a tumor-suppressing role in glioblastoma, but its role in glioblastoma multidrug resistance (MDR) is not clear. We reported that BMP4 can reverse MDR of glioblastoma through the inhibition of B-cell lymphoma 2(BCL-2) and glial cell derived neurotrophic factor (GDNF). We showed that the expression level of BMP4 was lower in glioblastoma compared to normal brain tissue, and also showed that BMP4 expression decreased in multidrug resistance cell line U251/TMZ compared to U251 cells. Our research demonstrated that over-expression of BMP4 can reverse the multidrug resistance. BCL-2 and GDNF were inhibited when BMP4 was over-expressed, and this data were consistent with the negative relationship in human samples; analysis of 40 patients glioblastoma and brain samples revealed a significant negative correlation between BMP4 and BCL-2, GDNF. When BCL-2 and GDNF were knocked down, the effect of BMP4 in regulating MDR was partially lost. This novel result showed, for the first time, that BMP4 can reverse MDR in glioblastoma, which involved negative inhibition of BCL-2 and GDNF.


Cancer Medicine | 2015

Downregulation of RND3/RhoE in glioblastoma patients promotes tumorigenesis through augmentation of notch transcriptional complex activity

Baohui Liu; Xi Lin; Xiangsheng Yang; Huimin Dong; Xiaojing Yue; Kelsey C. Andrade; Zhentao Guo; Ji-An Yang; Liquan Wu; Xiaonan Zhu; Shenqi Zhang; Daofeng Tian; Junmin Wang; Qiang Cai; Qizuan Chen; Shanping Mao; Qianxue Chen; Jiang Chang

Activation of Notch signaling contributes to glioblastoma multiform (GBM) tumorigenesis. However, the molecular mechanism that promotes the Notch signaling augmentation during GBM genesis remains largely unknown. Identification of new factors that regulate Notch signaling is critical for tumor treatment. The expression levels of RND3 and its clinical implication were analyzed in GBM patients. Identification of RND3 as a novel factor in GBM genesis was demonstrated in vitro by cell experiments and in vivo by a GBM xenograft model. We found that RND3 expression was significantly decreased in human glioblastoma. The levels of RND3 expression were inversely correlated with Notch activity, tumor size, and tumor cell proliferation, and positively correlated with patient survival time. We demonstrated that RND3 functioned as an endogenous repressor of the Notch transcriptional complex. RND3 physically interacted with NICD, CSL, and MAML1, the Notch transcriptional complex factors, promoted NICD ubiquitination, and facilitated the degradation of these cofactor proteins. We further revealed that RND3 facilitated the binding of NICD to FBW7, a ubiquitin ligase, and consequently enhanced NICD protein degradation. Therefore, Notch transcriptional activity was inhibited. Forced expression of RND3 repressed Notch signaling, which led to the inhibition of glioblastoma cell proliferation in vitro and tumor growth in the xenograft mice in vivo. Downregulation of RND3, however, enhanced Notch signaling activity, and subsequently promoted glioma cell proliferation. Inhibition of Notch activity abolished RND3 deficiency‐mediated GBM cell proliferation. We conclude that downregulation of RND3 is responsible for the enhancement of Notch activity that promotes glioblastoma genesis.


Brain Research | 2015

LRIG1, human EGFR inhibitor, reverses multidrug resistance through modulation of ABCB1 and ABCG2

Baohui Liu; Zhentao Guo; Huimin Dong; Tian Daofeng; Qiang Cai; Baowei Ji; Shenqi Zhang; Liquan Wu; Junmin Wang; Long Wang; Xiaonan Zhu; Yue Liu; Qianxue Chen

In our previous study, we have found that leucine-rich repeats and immunoglobulin-like domains 1(LRIG1) can improve the chemosensitivity in U251 cells whereas the role of LRIG1 in multidrug resistance (MDR) remains unknown. Here, we reported that LRIG1 can reverse MDR by inhibiting epidermal growth factor (EGF) receptor (EGFR) and secondary inhibiting ATP-binding cassette, sub-family B member 1(ABCB1) and ATP-binding cassette, sub-family G (WHITE), member 2 (ABCG2). Our data showed that the expression of LRIG1 was significantly higher in O6-methylguanine DNA methyltransferase (MGMT) Promoter Methylation positive glioblastoma tissues compared to MGMT Promoter Methylation negative glioblastoma tissues. In addition, we found that LRIG1 expression was significantly decreased in MDR cells U251/TMZ compared to U251cells. Our results demonstrated that over-expression of LRIG1 can reverse the MDR. The expression of ABCB1 and ABCG2 were markedly suppressed when LRIG1 was over-expressed, supporting the negative relationship between LRIG1 level and ABCB1 and ABCG2 level in human specimen. Furthermore, we found that LRIG1 downregulated ABCB1 and ABCG2 through suppressing EGFR expression. In case of EGFR knockdown, the effect of LRIG1 on regulating MDR, ABCB1 and ABCG2 was partially compromised. Our results, for the first time, showed that LRIG1 can reverse MDR in glioblastoma, by negatively regulating EGFR and secondary suppressing the levels of ABCB1 and ABCG2.


PLOS ONE | 2016

Expression of Ferritin Light Chain (FTL) Is Elevated in Glioblastoma, and FTL Silencing Inhibits Glioblastoma Cell Proliferation via the GADD45/JNK Pathway.

Tingfeng Wu; Yuntao Li; Baohui Liu; Shenqi Zhang; Liquan Wu; Xiaonan Zhu; Qianxue Chen

Accumulating evidence suggests that iron-associated proteins contribute to tumor initiation and development. Ferritin light chain (FTL), a key protein in iron metabolism, is associated with the survival of glioblastoma multiforme (GBM) patients; however, the molecular mechanisms underlying this association remain largely unclear. Therefore, in the present study, we investigated the role of FTL in the pathogenesis of GBM. By using quantitative real-time RT-PCR, we found that expression of FTL was higher in patients with GBM than in those with low-grade glioma. Immunofluorescence showed that FTL was mainly localized in the nucleus of GBM cells and was closely associated with mitotic spindles. Knockdown of FTL resulted in inhibition of cell growth and activation of the GADD45A/JNK pathway in GBM cells. Immunoblotting revealed that levels of GADD45A protein decreased in GBM cells when FTL expression increased. Furthermore, transfection of GADD45A in GBM cells significantly decreased cell viability, and this effect was impeded by co-transfection of FTL. Moreover, FTL was found to localize with GADD45A in GBM cells, and a coimmunoprecipitation experiment showed that the two proteins physically interacted. Taken together, these results demonstrate a novel mechanism by which FTL regulates the growth of GBM cells via the GADD45/JNK pathway.


Oncotarget | 2016

RND3 promotes Snail 1 protein degradation and inhibits glioblastoma cell migration and invasion

Baohui Liu; Huimin Dong; Xi Lin; Xiangsheng Yang; Xiaojing Yue; Ji-An Yang; Yuntao Li; Liquan Wu; Xiaonan Zhu; Shenqi Zhang; Daofeng Tian; Junmin Wang; Qiang Cai; Shanping Mao; Qianxue Chen; Jiang Chang

Activation of Snail1 signaling promotes the migration and invasion of multiple tumors, including glioblastoma multiforme (GBM). However, the molecular mechanism that augments Snail1 signaling during GBM cell migration and invasion remains largely unknown. Identification of the factors that regulate Snail1 signaling is critical to block tumor cell migration and invasion. By screening human GBM specimens, we found that the expression levels of small GTPase RND3 positively correlated with the expression levels of E-cadherin and claudin, the glioblastoma migration biomarkers negatively regulated by Snail1. Downregulation of E-cadherin and claudin has been associated with the migration and invasion of GBM cells. We demonstrated that RND3 functioned as an endogenous inhibitor of the Snail-directed transcriptional regulation. RND3 physically interacted with Snail1 protein, enhanced Snail1 ubiquitination, and facilitated the protein degradation. Forced expression of RND3 inhibited Snail1 activity, which in turn blocked glioblastoma cell migration and invasion in vitro in cell culture and in vivo in GBM xenograft mice. In contrast, downregulation of RND3 augmented Snail1 activity, and subsequently decreased E-cadherin expression, eventually promoted glioblastoma cell migration and invasion. The pro-migration induced by RND3 downregulation was attenuated by Snail1 knockdown. The findings partially explain why Snail1 activity is augmented in GBM, and defines a new function of RND3 in GBM cell migration and invasion.


Cell Biochemistry and Biophysics | 2015

LRIG1 Improves Chemosensitivity Through Inhibition of BCL-2 and MnSOD in Glioblastoma

Jianjun Ding; Baohui Liu; Yi He; Xianhou Yuan; Daofeng Tian; Baowei Ji; Long Wang; Liquan Wu; Huimin Dong; Junmin Wang; Xiaonan Zhu; Qiang Cai; Shenqi Zhang; Qianxue Chen

We have reported that Leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) can improve the chemosensitivity in U251 cells. However, the underlying mechanisms how LRIG1 improves the chemosensitivity remain unknown. In this study, we reported that LRIG1 can improve the chemosensitivity through the inhibition of B cell lymphoma 2 (BCL-2) and manganese superoxide dismutase (MnSOD). In addition, we showed that the expression level of LRIG1 was significantly negatively correlated with BCL-2 and MnSOD expression in glioma. Our research demonstrated that overexpression of LRIG1 can enhance the chemosensitivity. BCL-2 and MnSOD were inhibited in LRIG1 overexpressing cells. On the other hand, when BCL-2 and MnSOD were knocked down, the chemosensitivity of U251 cells decreased, and the effect of LRIG1 in regulating chemosensitivity was compromised. For the first time, our results showed that LRIG1 can enhance chemosensitivity in glioblastoma by inhibition of BCL-2 and MnSOD.


Cell Biochemistry and Biophysics | 2015

Down-Regulation of MRP1 Expression in C6/VP16 Cells by Chitosan-MRP1-siRNA Nanoparticles

Haitao Xu; Xiaoqi Nie; Liquan Wu; Xiaonan Zhu; Wei Yi; Shulan Huang

High expression of multidrug resistance-associated protein 1 (MRP1) in tumor cells reduces effectiveness of chemotherapy drugs. In this study, we screened MRP1 interfering RNA (MRP1-siRNA) molecules that are able to reverse etoposide (VP16) resistance in multidrug resistance rat glioma cell line C6/VP16, and identified one siRNA molecule that is able to effectively deplete the expression of MRP1 gene and reverse tumor cells resistance to etoposide. Since siRNA instability limits its application in treatment of diseases, we next tested silencing effect of chitosan-MRP1-siRNA nanoparticles and found that the nanoparticles with N:P ratio 175 are able to effectively inhibit MRP1 mRNA and protein expression. Our data demonstrate that chitosan can be used as siRNA carrier for high efficient gene silencing in tumor cells.


Journal of Huazhong University of Science and Technology-medical Sciences | 2015

Photodynamic therapy mediated by 5-aminolevulinic acid suppresses gliomas growth by decreasing the microvessels

Wei Yi; Haitao Xu; Daofeng Tian; Liquan Wu; Shenqi Zhang; Long Wang; Baowei Ji; Xiaonan Zhu; Humphrey Okechi; Gang Liu; Qianxue Chen

SummaryAlthough 5-aminolevulinic acid (5-ALA)-mediated photodynamic therapy (PDT) has been demonstrated to be a novel and effective therapeutic modality for some human malignancies, its effect and mechanism on glioma are still controversial. Previous studies have reported that 5-ALA-PDT induced necrosis of C6 rat glioma cells in vitro. The aim of this study was to further investigate the effect and mechanism of 5-ALA-PDT on C6 gliomas implanted in rats in vivo. Twenty-four rats bearing similar size of subcutaneously implanted C6 rat glioma were randomly divided into 3 groups: receiving 5-ALA-PDT (group A), laser irradiation (group B), and mock procedures but without any treatment (group C), respectively. The growth, histology, microvessel density (MVD), and apoptosis of the grafts in each group were determined after the treatments. As compared with groups B and C, the volume of tumor grafts was significantly reduced (P<0.05), MVD was significantly decreased (P<0.001), and the cellular necrosis was obviously increased in group A. There was no significant difference in apoptosis among the three groups. The in vivo studies confirmed that 5-ALA-PDT may be an effective treatment for gliomas by inhibiting the tumor growth. The mechanism underlying may involve increasing the cellular necrosis but not inducing the cellular apoptosis, which may result from the destruction of the tumor microvessels.


Experimental and Therapeutic Medicine | 2018

FOXK1 promotes glioblastoma proliferation and metastasis through activation of Snail transcription

Haitao Xu; Shulan Huang; Xiaonan Zhu; Wangcheng Zhang; Xiangyang Zhang

Forkhead box K1 (FOXK1) has been identified to have a crucial function in development and oncogenesis. However, its role in glioblastoma has remained largely elusive and was therefore assessed in the present study. In human glioblastoma multiforme (GBM) tissue samples, FOXK1 was determined to be highly expressed compared with adjacent normal tissue samples. In addition, high levels of FOXK1 were detected in the T98G and LN18 GBM cell lines as compare with those in normal human astrocytes. Of note, high expression of FOXK1 was revealed to be associated with metastasis and tumor size. Loss- and gain-of-function experiments were then performed to determine whether FOXK1 regulates epithelial to mesenchymal transition (EMT) and cell proliferation. Knockdown of FOXK1 significantly suppressed EMT and metastasis of GBM cells, while ectopic expression of FOXK1 promoted them. A luciferase reporter assay and a chromatin immunoprecipitation assay revealed that FOXK1 activated the transcription of Snail. In addition, as the results indicated that FOXK1 promotes GBM cell proliferation, the potential effect of FOXK1 on the cell cycle and apoptosis were further assessed. While FOXK1 had no effect on apoptosis, it promoted cell proliferation via enhancing the S-phase population. In brief, the present study indicated that FOXK1 acts as an oncogene with a key function in glioblastoma cell proliferation and EMT.

Collaboration


Dive into the Xiaonan Zhu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge