Network


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

Hotspot


Dive into the research topics where Shiguang Zhao is active.

Publication


Featured researches published by Shiguang Zhao.


Journal of Molecular Medicine | 2011

MiR-106a inhibits glioma cell growth by targeting E2F1 independent of p53 status

Guang Yang; Ruyou Zhang; Xiaofeng Chen; Yulong Mu; Jing Ai; Chen Shi; Yaohua Liu; Changbin Shi; Lihua Sun; Nikolai G. Rainov; Hulun Li; Baofeng Yang; Shiguang Zhao

MicroRNAs are single-stranded small non-coding RNA molecules which regulate mammalian cell growth, differentiation, and apoptosis by altering the expression of other genes and play a role in tumor genesis and progression. MiR-106a is upregulated in several types of malignancies and provides a pro-tumorigenic effect. However, its role in glioma is largely unknown. Our findings demonstrate that the low expression of miR-106a in human glioma specimens is significantly correlated with high levels of E2F1 protein and high-grade glioma. Here, we present the first evidence that miR-106a provides a tumor-suppressive effect via suppressing proliferation of and inducing apoptosis in human glioma cells. We further show that E2F1 is a direct functional target of miR-106a, suggesting that the effect of miR-106a on the glioma suppressive effect may result from inhibition of E2F1 via post-transcriptional regulation. In addition, our results reveal that miR-106a can increase p53 expression via E2F1 inhibition, whereas the effect of miR-106a on the proliferation of glioma cells is independent of p53 status. Further investigations will focus on the therapeutic use of miR-106a-mediated antitumor effects in glioma.


Cancer Letters | 2013

miR-143 inhibits glycolysis and depletes stemness of glioblastoma stem-like cells

Shiguang Zhao; Huailei Liu; Yaohua Liu; Jianing Wu; Chunlei Wang; Xu Hou; Xiaofeng Chen; Guang Yang; Ling Zhao; Hui Che; Yunke Bi; Hongyu Wang; Fei Peng; Jing Ai

Glioblastomas rely mainly on aerobic glycolysis to sustain proliferation and growth; however, little is known about the regulatory mechanisms of metabolism in glioblastoma stem cells. We show that miR-143 is significantly down-regulated in glioma tissues and glioblastoma stem-like cells (GSLCs), while miR-143 over-expression inhibits glycolysis by directly targeting hexokinase 2, and promotes differentiation of GSLCs. Moreover, miR-143 inhibits proliferation of GSLCs under hypoxic conditions and decreases tumor formation capacity of GSLCs in vivo. We also show that a combination of miR-143 and 2-DG, a widely used glycolysis inhibitor, has synergistic effects against GSLCs. miR-143 is a potential therapeutic target for glioblastoma treatment.


Molecular Biology Reports | 2008

Arsenic trioxide induces different gene expression profiles of genes related to growth and apoptosis in glioma cells dependent on the p53 status

Shiguang Zhao; Jian Zhang; Xu Zhang; Xuesong Dong; Xueying Sun

We have previously reported that As2O3 affected cell cycle progression and cyclins D1 and B1 expression in two glioma cell lines differing in p53 status (U87MG-wt; T98G-mutated). In the present study, we further demonstrated that As2O3 affected proliferation, viability and apoptosis of the two cell lines in a dose- and time-dependent manner, and T98G cells were more sensitive than U87MG cells to As2O3 -induced apoptosis and inhibition of proliferation and viability. We further investigated the expression profiles of genes related with apoptosis and cell cycle in the two cell lines with a human cDNA-microarray (SuperArray) spotted with 267 genes of apoptosis and cell cycle. Thirty five genes were upregulated and 15 genes downregulated at least 2-fold by As2O3 in U87-MG cells; whereas, 38 genes were upregulated and 21 genes downregulated at least 2-fold in T98G cells by As2O3. After As2O3 treatment, p53 expression was upregulated 56.5-fold in T98G cells, but only 6.0-fold in U87MG cells. The results indicate that As2O3 suppresses the growth of U87MG cells mainly by regulating expression of genes of cell cycle arrest, stress and toxicity; whereas As2O3 affects T98G cells mainly by regulating expression of genes belonging to Bcl-2, tumor necrotic factor receptor and ligand families. The data may be helpful for optimizing As2O3 as an anti-cancer drug in the treatment of gliomas.


Toxicology Letters | 2013

Arsenic trioxide depletes cancer stem-like cells and inhibits repopulation of neurosphere derived from glioblastoma by downregulation of Notch pathway.

Jianing Wu; Zhiyong Ji; Huailei Liu; Yaohua Liu; Dayong Han; Chen Shi; Changbin Shi; Chunlei Wang; Guang Yang; Xiaofeng Chen; Chen Shen; Huadong Li; Yunke Bi; Dongzhi Zhang; Shiguang Zhao

Notch signaling has been demonstrated to have a central role in cancer stem-like cells (CSLCs) in glioblastoma multiforme (GBM). We have recently demonstrated the inhibitory effect of arsenic trioxide (ATO) on CSLCs in glioblastoma cell lines. In this study we used neurosphere recovery assay that measured neurosphere formation at three time points to assess the capacity of the culture to repopulate after ATO treatment. Our results provided strong evidence that ATO depleted CSLCs in GBM, and inhibited neurosphere recovery and secondary neurosphere formation. ATO inhibited the phosphorylation and activation of AKT and STAT3 through Notch signaling blockade. These data show that the ATO is a promising new approach to decrease glioblastoma proliferation and recurrence by downregulation of Notch pathway.


Journal of Neurology, Neurosurgery, and Psychiatry | 2011

A predictive value of hyponatremia for poor outcome and cerebral infarction in high-grade aneurysmal subarachnoid haemorrhage patients

Bingjie Zheng; Y Qiu; H Jin; Lu Wang; Xiaofeng Chen; Changbin Shi; Shiguang Zhao

Background The clinical significance of hyponatremia has not been investigated in high-grade aneurysmal subarachnoid haemorrhage (aSAH) patients. Thus, we assessed the predictive value of hyponatremia for poor outcome or cerebral infarction in high grade patients (the World Federation of Neurological Surgeons Scale (WFNS) grade 4 or 5) after aSAH. Methods Patients with WFNS grade 4 or 5 after aSAH were selected into this study between January 2005 and January 2008. In the same period, patients with WFNS grade 1, 2 or 3 after aSAH (low grade) were also chosen into this study. Hyponatremia was determined with serum sodium measurements obtained within 9 days after aSAH. Prognosis of patients was estimated with Glasgow Outcome Scale at 3 months. The relationship between hyponatremia and poor outcome and association of hyponatremia and cerebral infarction were analysed, respectively. Results A total of 124 high-grade patients were included in this study. Of those, 78 patients developed hyponatremia. Hyponatremia developed in 32.3% of cases between days 1 and 3 after aSAH, and 30.6% developed hyponatremia after 3 days post-aSAH. Multivariable analysis revealed that hyponatremia was not correlated with poor outcome in high-grade aSAH patients. Furthermore, only late-onset hyponatremia was correlated with cerebral infarction in these patients. Meanwhile, there was no significant correlation between hyponatremia and poor outcome or cerebral infarction in 259 low-grade aSAH patients. Conclusions Hyponatremia does not predict poor outcome in all-grade aSAH patients. However, late-onset hyponatremia in high-grade aSAH patients is associated with cerebral infarction. Therefore, the appropriate management of hyponatremia could be beneficial in those patients.


Journal of Neuro-oncology | 2011

Inhibition of heme oxygenase-1 enhances anti-cancer effects of arsenic trioxide on glioma cells

Yaohua Liu; Yuan Liang; Tianhu Zheng; Guang Yang; Xu Zhang; Zhe Sun; Changbin Shi; Shiguang Zhao

We have previously reported that arsenic trioxide (ATO) could inhibit glioma growth both in vitro and in vivo, and demonstrated its potent therapeutic effects on gliomas. In this study we showed that ATO induced cell damage and heme oxygenase-1 (HO-1) expression in glioma cells via ROS generation. HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. In addition, knockdown of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) strongly inhibited HO-1 expression induced by ATO, and significantly enhanced ATO-induced oxidative damage. Our results demonstrated, for the first time, that HO-1 inhibition or Nrf2 knockdown significantly potentiated ATO’s effects on glioma cells. Considering that HO-1 is highly expressed in glioma tissues, administration of ATO in combination with either HO-1 inhibitor or Nrf2 knockdown may act as a new approach to the treatment of glioma.


Clinical Cancer Research | 2006

2-methoxyestradiol, an endogenous mammalian metabolite, radiosensitizes colon carcinoma cells through c-Jun NH2-terminal kinase activation.

Huichao Zou; Masaaki Adachi; Kohzoh Imai; Masato Hareyama; Katsuji Yoshioka; Shiguang Zhao; Yasuhisa Shinomura

Purpose: 2-Methoxyestradiol (2ME), an estrogen metabolite, induces apoptosis in various cell types. We investigated whether 2ME pretreatment can radiosensitize colon adenocarcinoma cells. Experimental Design: Radiosensitizing effects of 2ME were evaluated by cell death, clonogenic assay, nuclear fragmentation, and tumor progression of xenografts. Ionizing radiation–induced DNA damage was evaluated by histone H2AX phosphorylation and its foci. The c-Jun NH2-terminal kinase (JNK) activation was evaluated by anti-phosphorylated JNK antibody and inhibited by the JNK-specific inhibitor SP600125 or dominant-negative SEK1 expression. Results: Clonogenic assays revealed that 2ME, but not estradiol, radiosensitized three colon carcinoma cells, DLD-1, HCT-8, and HCT-15, and strongly suppressed tumor progression of DLD-1 xenografts. Gene transfer–mediated Bcl-xL overexpression largely abolished both augmented apoptosis and reduced survival fractions. Pretreatment with 2ME enhanced H2AX phosphorylation, its foci, and phosphorylation of ATM kinase and delayed re-entry of cell cycle progression after ionizing radiation. Augmentation of both radiosensitivity and H2AX phosphorylation was substantially reduced by SP600125 or overexpression of a dominant-negative mutant SEK1. Conclusion: 2ME radiosensitized colon carcinoma cells through enhanced DNA damage via JNK activation, thereby representing a novel radiosensitizing therapy against colon cancer.


Journal of Chemotherapy | 2008

Intravenous Administration of Arsenic Trioxide Encapsulated in Liposomes Inhibits the Growth of C6 Gliomas in Rat Brains

Shiguang Zhao; Xu Zhang; Jian Zhang; Jianhua Zhang; Huichao Zou; Yaohua Liu; Xuesong Dong; Xueying Sun

Abstract Arsenic trioxide (ATO) is a potent anti-tumor agent used to treat acute promyelocytic leukemia (APL), and more recently solid tumors including gliomas. However, the dose of ATO required to suppress gliomas is markedly higher than that used to treat APL, which leads to toxicity and undesirable side-effects, even though the local concentration of ATO in brains is relatively low after systemic administration. In an attempt to minimize the toxicity, enhance the penetrating activity across the bloodbrain barrier, and reduce enzyme degradation, we prepared ATO encapsulated in liposomes, and investigated its therapeutic effect on C6 gliomas established in rat brains. The prepared ATO liposomes were stable at room temperature for 3 days and the latency rate was over 90% within 72 h. Intravenous injection of ATO liposomes led to a much higher concentration of ATO (5- fold, compared with ATO solution) in rat brains, resulting in inhibition of C6 gliomas in brains and prolonging the survival of rats bearing brain gliomas. ATO-liposome therapy resulted in fewer side effects, compared with free ATO solution. ATO-liposome therapy inhibited tumor angiogenesis by downregulating the expression of vascular endothelial growth factor (VEGF), and inducing cell apoptosis. The results warrant future investigation of the use of ATO encapsulated in liposomes to treat gliomas.


Neuropathology and Applied Neurobiology | 2011

Targeting X box‐binding protein‐1 (XBP1) enhances sensitivity of glioma cells to oxidative stress

Yaohua Liu; X. Zhang; Y. Liang; H. Yu; Xiaofeng Chen; Tianhu Zheng; Bingjie Zheng; Ligang Wang; L. Zhao; Changbin Shi; Shiguang Zhao

Y. Liu, X. Zhang, Y. Liang, H. Yu, X. Chen, T. Zheng, B. Zheng, L. Wang, L. Zhao, C. Shi and S. Zhao (2011) Neuropathology and Applied Neurobiology37, 395–405 
Targeting X box‐binding protein‐1 (XBP1) enhances sensitivity of glioma cells to oxidative stress


Molecular Biology Reports | 2012

Heat stress activates ER stress signals which suppress the heat shock response, an effect occurring preferentially in the cortex in rats

Yaohua Liu; Hiroaki Sakamoto; Masaaki Adachi; Shiguang Zhao; Wataru Ukai; Eri Hashimoto; Masato Hareyama; Tadao Ishida; Kohzoh Imai; Yasuhisa Shinomura

Although heat stress induces a variety of illnesses, there have been few studies designed to uncover the molecular mechanisms underlining the illnesses. We here demonstrate that heat activates ER stress, which inhibits heat shock responses (HSR) via translational block. In heat-stressed rats, ER stress responses, as represented by eIF2α phosphorylation and XBP1 splicing, occurred mainly in the cortex, where the HSR was substantially inhibited. Heat exposure also activated ER stress signals in primary cortical neurons. Since HSF1 knockdown enhanced heat-induced ER stress and subsequent cell death, HSR inhibition in turn augments ER stress, implying a vicious spiral of both stresses. Taken together, heat-induced ER stress impairs the HSR and enhances cell damage, thereby manifesting its unique effect on heat stress.

Collaboration


Dive into the Shiguang Zhao's collaboration.

Top Co-Authors

Avatar

Yaohua Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaofeng Chen

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guang Yang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Chunlei Wang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Huailei Liu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianing Wu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Tianhu Zheng

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xu Zhang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Bingjie Zheng

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge