Network


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

Hotspot


Dive into the research topics where Bingzheng Lu is active.

Publication


Featured researches published by Bingzheng Lu.


Molecular Therapy | 2016

Activation of Cyclic Adenosine Monophosphate Pathway Increases the Sensitivity of Cancer Cells to the Oncolytic Virus M1.

Kai Li; Haipeng Zhang; Jianguang Qiu; Yuan Lin; Jiankai Liang; Xiao Xiao; Liwu Fu; Fang Wang; Jing Cai; Yaqian Tan; Wenbo Zhu; Wei Yin; Bingzheng Lu; Fan Xing; Lipeng Tang; Min Yan; Jialuo Mai; Yuan Li; Wenli Chen; Pengxin Qiu; Xingwen Su; Guangping Gao; Phillip W.L. Tai; Jun Hu; Guangmei Yan

Oncolytic virotherapy is a novel and emerging treatment modality that uses replication-competent viruses to destroy cancer cells. Although diverse cancer cell types are sensitive to oncolytic viruses, one of the major challenges of oncolytic virotherapy is that the sensitivity to oncolysis ranges among different cancer cell types. Furthermore, the underlying mechanism of action is not fully understood. Here, we report that activation of cyclic adenosine monophosphate (cAMP) signaling significantly sensitizes refractory cancer cells to alphavirus M1 in vitro, in vivo, and ex vivo. We find that activation of the cAMP signaling pathway inhibits M1-induced expression of antiviral factors in refractory cancer cells, leading to prolonged and severe endoplasmic reticulum (ER) stress, and cell apoptosis. We also demonstrate that M1-mediated oncolysis, which is enhanced by cAMP signaling, involves the factor, exchange protein directly activated by cAMP 1 (Epac1), but not the classical cAMP-dependent protein kinase A (PKA). Taken together, cAMP/Epac1 signaling pathway activation inhibits antiviral factors and improves responsiveness of refractory cancer cells to M1-mediated virotherapy.


Human Gene Therapy | 2016

Naturally Existing Oncolytic Virus M1 Is Nonpathogenic for the Nonhuman Primates After Multiple Rounds of Repeated Intravenous Injections

Haipeng Zhang; Yuan Lin; Kai Li; Jiankai Liang; Xiao Xiao; Jing Cai; Yaqian Tan; Fan Xing; Jialuo Mai; Yuan Li; Wenli Chen; Longxiang Sheng; Jiayu Gu; Wenbo Zhu; Wei Yin; Pengxin Qiu; Xingwen Su; Bingzheng Lu; Xuyan Tian; Jinhui Liu; Wanjun Lu; Yunling Dou; Yijun Huang; Zhuang Kang; Guangping Gao; Zixu Mao; Shi Yuan Cheng; Ling Lu; Xuetao Bai; Guangmei Yan

Cancers figure among the leading causes of morbidity and mortality worldwide. The number of new cases is expected to rise by about 70% over the next 2 decades. Development of novel therapeutic agents is urgently needed for clinical cancer therapy. Alphavirus M1 is a Getah-like virus isolated from China with a genome of positive single-strand RNA. We have previously identified that alphavirus M1 is a naturally existing oncolytic virus with significant anticancer activity against different kinds of cancer (e.g., liver cancer, bladder cancer, and colon cancer). To support the incoming clinical trial of intravenous administration of alphavirus M1 to cancer patients, we assessed the safety of M1 in adult nonhuman primates. We previously presented the genome sequencing data of the cynomolgus macaques (Macaca fascicularis), which was demonstrated as an ideal animal species for virus infection study. Therefore, we chose cynomolgus macaques of either sex for the present safety study of oncolytic virus M1. In the first round of administration, five experimental macaques were intravenously injected with six times of oncolytic virus M1 (1 × 10(9) pfu/dose) in 1 week, compared with five vehicle-injected control animals. The last two rounds of injections were further completed in the following months in the same way as the first round. Body weight, temperature, complete blood count, clinical biochemistries, cytokine profiles, lymphocytes subsets, neutralizing antibody, and clinical symptoms were closely monitored at different time points. Magnetic resonance imaging was also performed to assess the possibility of encephalitis or arthritis. As a result, no clinical, biochemical, immunological, or medical imaging or other pathological evidence of toxicity was found during the whole process of the study. Our results in cynomolgus macaques suggested the safety of intravenous administration of oncolytic virus M1 in cancer patients in the future.


Cell Reports | 2017

The Anti-Warburg Effect Elicited by the cAMP-PGC1α Pathway Drives Differentiation of Glioblastoma Cells into Astrocytes

Fan Xing; Yizhao Luan; Jing Cai; Sihan Wu; Jialuo Mai; Jiayu Gu; Haipeng Zhang; Kai Li; Yuan Lin; Xiao Xiao; Jiankai Liang; Yuan Li; Wenli Chen; Yaqian Tan; Longxiang Sheng; Bingzheng Lu; Wanjun Lu; Mingshi Gao; Pengxin Qiu; Xingwen Su; Wei Yin; Jun Hu; Zhongping Chen; Ke Sai; Jing Wang; Furong Chen; Yinsheng Chen; Shida Zhu; Dongbing Liu; Shi Yuan Cheng

SUMMARY Glioblastoma multiforme (GBM) is among the most aggressive of human cancers. Although differentiation therapy has been proposed as a potential approach to treat GBM, the mechanisms of induced differentiation remain poorly defined. Here, we established an induced differentiation model of GBM using cAMP activators that specifically directed GBM differentiation into astroglia. Transcriptomic and proteomic analyses revealed that oxidative phosphorylation and mitochondrial biogenesis are involved in induced differentiation of GBM. Dibutyryl cyclic AMP (dbcAMP) reverses the Warburg effect, as evidenced by increased oxygen consumption and reduced lactate production. Mitochondrial biogenesis induced by activation of the CREB-PGC1α pathway triggers metabolic shift and differentiation. Blocking mitochondrial biogenesis using mdivi1 or by silencing PGC1α abrogates differentiation; conversely, overexpression of PGC1α elicits differentiation. In GBM xenograft models and patient-derived GBM samples, cAMP activators also induce tumor growth inhibition and differentiation. Our data show that mitochondrial biogenesis and metabolic switch to oxidative phosphorylation drive the differentiation of tumor cells.


Science Translational Medicine | 2017

Targeting VCP enhances anticancer activity of oncolytic virus M1 in hepatocellular carcinoma

Haipeng Zhang; Kai Li; Yuan Lin; Fan Xing; Xiao Xiao; Jing Cai; Wenbo Zhu; Jiankai Liang; Yaqian Tan; Liwu Fu; Fang Wang; Wei Yin; Bingzheng Lu; Pengxin Qiu; Xingwen Su; Xuetao Bai; Jun Hu; Guangmei Yan

Inhibition of VCP sensitizes hepatocellular carcinoma cells to oncolytic virus M1–induced apoptosis. A virus and its reinforcement Oncolytic viruses can be effective against a variety of cancers, including hepatocellular carcinoma, where a viral treatment is showing evidence of efficacy in people. Zhang et al. performed a high-throughput drug screen to search for compounds to pair with an oncolytic virus called M1 to further increase its effectiveness against hepatocellular carcinoma. Through this screen, they identified inhibitors of valosin-containing protein, then used them together with M1, and demonstrated the efficacy of this regimen in mouse models of cancer. In addition, the combination was well tolerated in primates, suggesting that the drug and virus combination may translate to human patients. Oncolytic virotherapy is rapidly progressing through clinical evaluation. However, the therapeutic efficacy of oncolytic viruses in humans has been less than expected from preclinical studies. We describe an anticancer drug screen for compounds that enhance M1 oncolytic virus activity in hepatocellular carcinoma (HCC). An inhibitor of the valosin-containing protein (VCP) was identified as the top sensitizer, selectively increasing potency of the oncolytic virus up to 3600-fold. Further investigation revealed that VCP inhibitors cooperated with M1 virus–suppressed inositol-requiring enzyme 1α (IRE1α)–X-box binding protein 1 (XBP1) pathway and triggered irresolvable endoplasmic reticulum (ER) stress, subsequently promoting robust apoptosis in HCC. We show that VCP inhibitor improved the oncolytic efficacy of M1 virus in several mouse models of HCC and primary HCC tissues. Finally, this combinatorial therapeutic strategy was well tolerated in nonhuman primates. Our study identifies combined VCP inhibition and oncolytic virus as a potential treatment for HCC and demonstrates promising therapeutic potential.


Molecular Medicine Reports | 2016

Effect of glutamate on lysosomal membrane permeabilization in primary cultured cortical neurons.

Min Yan; Wenbo Zhu; Xiaoke Zheng; Yuan Li; Lipeng Tang; Bingzheng Lu; Wenli Chen; Pengxin Qiu; Tiandong Leng; Suizhen Lin; Guangmei Yan; Wei Yin

Glutamate is the principal neurotransmitter in the central nervous system. Glutamate-mediated excitotoxicity is the predominant cause of cerebral damage. Recent studies have shown that lysosomal membrane permeabilization (LMP) is involved in ischemia-associated neuronal death in non-human primates. This study was designed to investigate the effect of glutamate on lysosomal stability in primary cultured cortical neurons. Glutamate treatment for 30 min induced the permeabilization of lysosomal membranes as assessed by acridine orange redistribution and immunofluorescence of cathepsin B in the cytoplasm. Inhibition of glutamate excitotoxicity by the NMDA receptor antagonist MK-801 and the calcium chelator ethylene glycolbis (2-aminoethylether)-N, N, N′, N′-tetraacetic acid, rescued lysosomes from permeabilization. The role of calpain and reactive oxygen species (ROS) in inducing LMP was also investigated. Ca2+ overload following glutamate treatment induced the activation of calpain and the production of ROS, which are two major contributors to neuronal death. It has been reported that lysosomal-associated membrane protein 2 (LAMP2) and heat shock protein (HSP)70 are two calpain substrates that promote LMP in cancer cells; however, it was found that calpains were activated by glutamate, but only LAMP2 was subsequently degraded. Furthermore, LMP was not alleviated by treatment with the calpain inhibitors calpeptin and SJA6017, which blocked the cleavage of the calpain substrate α-fodrin. It was demonstrated that LMP was significantly alleviated by treatment with the antioxidant N-Acetyl-L-cysteine, indicating that LMP involvement in early glutamate excitotoxicity may be mediated partly by ROS rather than calpain activation. Overall, these data shed light on the role of ROS-mediated LMP in early glutamate excitotoxicity.


Steroids | 2015

Cholesterol metabolite cholestane-3β,5α,6β-triol suppresses epileptic seizures by negative modulation of voltage-gated sodium channels.

Lipeng Tang; Youqiong Wang; Tiandong Leng; Huanhuan Sun; Yuehan Zhou; Wenbo Zhu; Pengxin Qiu; Jingxia Zhang; Bingzheng Lu; Min Yan; Wenli Chen; Xinwen Su; Wei Yin; Yijun Huang; Haiyan Hu; Guangmei Yan

Imbalance of excitation and inhibition in neurons is implicated in the pathogenesis of epilepsy. Voltage-gated sodium channels, which play a vital role in regulating neuronal excitability, are one of the major targets for developing anti-epileptic drugs. Here we provide evidence that cholestane-3β,5α,6β-triol (triol), a major metabolic oxysterol of cholesterol, is an effective state-dependent negative sodium channels modulator. Triol reduced Na(+) current density in a concentration-dependent manner. 10 μM triol shifted steady-state/fast/slow inactivation curves of sodium channels toward the hyperpolarizing direction. Additionally, triol reduced voltage-gated sodium currents in a voltage- and frequency-dependent manner. In a kainic acid-induced seizures mouse model, triol (25 mg/kg) significantly increased the latency of seizure onset and attenuated seizure severity. Our findings provide novel insights for understanding the modulatory role of a small molecular oxysterol on voltage-gated sodium channels and suggest triol may represent a novel and promising candidate for epilepsy intervention.


Neuro-oncology | 2015

Transcriptional upregulation of microtubule-associated protein 2 is involved in the protein kinase A-induced decrease in the invasiveness of glioma cells

Yuxi Zhou; Sihan Wu; Chaofeng Liang; Yuan Lin; Yan Zou; Kai Li; Bingzheng Lu; Minfeng Shu; Yijun Huang; Wenbo Zhu; Zhuang Kang; Dong Xu; Jun Hu; Guangmei Yan

BACKGROUND Malignant glioma is the most lethal primary tumor of the central nervous system, with notable cell invasion causing significant recurrence. Suppression of glioma invasion is very important for improving clinical outcomes. Drugs that directly disrupt the cytoskeleton have been developed for this purpose; however, drug resistance and unsatisfactory selectivity have limited their clinical use. Previously, we reported that protein kinase A (PKA, also known as cyclic-AMP dependent protein kinase) activation induced the differentiation of glioma cells. METHODS We used several small molecular inhibitors and RNA interference, combined with wound healing assays, Matrigel transwell assay, and microscopic observation, to determine whether activation of the PKA pathway could inhibit the invasion of human glioma cells. RESULTS Activation of PKA decreased the invasion of glioma cells. The mechanism operated via transcriptional upregulation of microtubule-associated protein 2 (MAP2), which was activated by the PKA pathway and led to ossification of microtubule dynamics via polymerization of tubulin. This resulted in morphological changes and a reduction in glioma cell invasion. Furthermore, chromosome immunoprecipitation and quantitative real-time polymerase chain reaction showed that signal transducer and activator of transcription 3 (STAT3) is involved in the transcriptional upregulation of MAP2. CONCLUSION Our findings suggested that PKA may represent a potential target for anti-invasion glioma therapy and that the downstream modulators (eg, STAT3/MAP2) partially mediate the effects of PKA.


Journal of Neurochemistry | 2018

Hexokinase 2-dependent hyperglycolysis driving microglial activation contributes to ischemic brain injury

Yuan Li; Bingzheng Lu; Longxiang Sheng; Zhu Zhu; Hongjiaqi Sun; Yuwei Zhou; Yang Yang; Dongdong Xue; Wenli Chen; Xuyan Tian; Yun Du; Min Yan; Wenbo Zhu; Fan Xing; Kai Li; Suizhen Lin; Pengxin Qiu; Xingwen Su; Yijun Huang; Guangmei Yan; Wei Yin

Hyperglycolysis, observed within the penumbra zone during brain ischemia, was shown to be detrimental for tissue survival because of lactate accumulation and reactive oxygen species overproduction in clinical and experimental settings. Recently, mounting evidence suggests that glycolytic reprogramming and induced metabolic enzymes can fuel the activation of peripheral immune cells. However, the possible roles and details regarding hyperglycolysis in neuroinflammation during ischemia are relatively poorly understood. Here, we investigated whether overactivated glycolysis could activate microglia and identified the crucial regulators of neuroinflammatory responses in vitro and in vivo. Using BV 2 and primary microglial cultures, we found hyperglycolysis and induction of the key glycolytic enzyme hexokinase 2 (HK2) were essential for microglia‐mediated neuroinflammation under hypoxia. Mechanistically, HK2 up‐regulation led to accumulated acetyl‐coenzyme A, which accounted for the subsequent histone acetylation and transcriptional activation of interleukin (IL)‐1β. The inhibition and selective knockdown of HK2 in vivo significantly protected against ischemic brain injury by suppressing microglial activation and IL‐1β production in male Sprague–Dawley rats subjected to transient middle cerebral artery occlusion (MCAo) surgery. We provide novel insights for HK2 specifically serving as a neuroinflammatory determinant, thus explaining the neurotoxic effect of hyperglycolysis and indicating the possibility of selectively targeting HK2 as a therapeutic strategy in acute ischemic stroke.


Acta Pharmaceutica Sinica B | 2014

Synthesis of taurine-fluorescein conjugate and evaluation of its retina-targeted efficiency in vitro.

Meihong Huang; Jiaqi Song; Bingzheng Lu; Huizhi Huang; Yizhen Chen; Wei Yin; Wenbo Zhu; Xinwen Su; Chuanbin Wu; Haiyan Hu

In this work, retinal penetration of fluorescein was achieved in vitro by covalent attachment of taurine to fluorescein, yielding the F–Tau conjugate. Nuclear magnetic resonance (NMR) and high resolution mass spectrometry (HRMS) were used to confirm the successful synthesis of F–Tau. The cellular uptake of F–Tau in adult retinal pigment epithelial cells (ARPE-19) and human retinal microvascular endothelial cells (hRMECs) was visualized via confocal scanning microscopy. The results indicated an improvement of solubility and a reduction of logP of F–Tau compared with fluorescein. As compared with fluorescein, F–Tau showed little toxicity, and was retained longer by cells in uptake experiments. F–Tau also displayed higher transepithelial permeabilities than fluorescein in ARPE-19 and hRMECs monolayer cells (P<0.05). These results showed that taurine may be a useful ligand for targeting small-molecule hydrophobic pharmaceuticals into the retina.


PLOS ONE | 2017

Mycoplasma-associated multidrug resistance of hepatocarcinoma cells requires the interaction of P37 and Annexin A2

Danyang Liu; Yang Hu; Ying Guo; Zhu Zhu; Bingzheng Lu; Xuelan Wang; Yijun Huang

Mycoplasma infection has been reported to be associated with cancer migration, invasion, epithelial-mesenchymal transition as well as the resistance to nucleoside analogues chemotherapeutic drugs. In this study, we found that the sensitivity of hepatocarcinoma cells to Cisplatin, Gemcitabine and Mitoxantrone was increased by mycoplasma elimination. Similar to the effect of anti-mycoplasma agent, interrupting the interaction between Mycoplasma hyorhinis membrane protein P37 and Annexin A2 of host cells using the N-terminal of ANXA2 polypeptide enhanced the sensitivity of HCC97L cells to Gemcitabine and Mitoxantrone. Meanwhile, we did not observe any changes in expression or distribution of multidrug resistance associated transporters, ATP-Binding Cassette protein B1, C1 and G2, on the removal of mycoplasma. These results suggest that mycoplasma induces a resistance to multiple drugs in hepatocarcinoma cells which required the interaction of P37 and Annexin A2. The pathway downstream this interaction needs to be explored.

Collaboration


Dive into the Bingzheng Lu's collaboration.

Top Co-Authors

Avatar

Wei Yin

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenbo Zhu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Pengxin Qiu

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Fan Xing

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Kai Li

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Wenli Chen

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Xingwen Su

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yuan Li

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yuan Lin

Sun Yat-sen University

View shared research outputs
Researchain Logo
Decentralizing Knowledge