Network


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

Hotspot


Dive into the research topics where Liangliang Zhan is active.

Publication


Featured researches published by Liangliang Zhan.


Tumor Biology | 2014

MicroRNA-21 is a novel promising target in cancer radiation therapy

Jia Liu; Hongcheng Zhu; Xi Yang; Yangyang Ge; Chi Zhang; Qin Qin; Jing Lu; Liangliang Zhan; Hongyan Cheng; Xinchen Sun

MicroRNAs (miRNAs) represent an important nonprotein part of the human genome in tumor biology. Among the several types of miRNAs, microRNA-21 (miR-21) is dysregulated in several types of cancer and plays a key role in carcinogenesis, recurrence, and metastasis. Thus, it can be a potential target for cancer therapy including radiation therapy. In this review, we focus on miR-21, which has been identified in human cancer tissues, to suggest reasonable strategies for future research. miR-21 may have an influence on cell cycle, DNA damage repair, apoptosis, autophagy, and hypoxia of cancer during irradiation. We review the use of miR-21 in cancer radiation therapy and describe the known functions and possible underlying molecular mechanisms of miR-21 in radiosensitivity and radioresistance. Furthermore, the current and potential future applications of miR-21 in cancer radiation therapy are also discussed.


Tumor Biology | 2014

STAT3 inhibitor NSC74859 radiosensitizes esophageal cancer via the downregulation of HIF-1α

Chi Zhang; Xi Yang; Qu Zhang; Qing Guo; Jia He; Qin Qin; Hongcheng Zhu; Jia Liu; Liangliang Zhan; Jing Lu; Zheming Liu; Liping Xu; Jianxin Ma; Shengbin Dai; Hongyan Cheng; Xinchen Sun

Radiotherapy is the main therapy for inoperable and locally advanced esophageal squamous cell carcinoma (ESCC). However, radioresistance in ESCC remains a challenge. The aim of this study is to investigate the radiosensitizing effects of STAT3 inhibitor NSC74859 on ESCC and explore the underlying mechanisms. ECA109 and TE13 cells were exposed to hypoxia, and treated with NSC74859 or radiation, alone or in combination. Cell proliferation, survival, apoptosis, and double-stranded DNA breaks (DSBs) were examined. Nude mice model of ECA109 xenograft was treated with radiation and/or NSC74859. The levels of STAT3, p-STAT3, HIF-1α, and VEGF were detected by Western blot analysis. NSC74859 efficiently radiosensitized ESCC cells and xenografts in nude mice, and inhibited hypoxia-/radiation-induced activation of STAT3 and upregulation of HIF-1α and VEGF expression. NSC74859 confers radiosensitivity in ESCC via the inhibition of STAT3 activation and the downregulation of HIF-1α and VEGF expression. NSC74859 may become a promising radiosensitizer for ESCC radiotherapy.


Tumor Biology | 2014

Smac mimetic compound LCL161 sensitizes esophageal carcinoma cells to radiotherapy by inhibiting the expression of inhibitor of apoptosis protein

Qin Qin; Yun Zuo; Xi Yang; Jing Lu; Liangliang Zhan; Liping Xu; Chi Zhang; Hongcheng Zhu; Jia Liu; Zheming Liu; Guangzhou Tao; Shengbin Dai; Xizhi Zhang; Jianxin Ma; Jing Cai; Xinchen Sun

Currently, unresectable esophageal squamous cell carcinoma (ESCC) is primarily treated by chemoradiotherapy. However, the outcome has not improved significantly due to radioresistance of cancer cells. This study aimed to determine the radiosensitizing effect of LCL161, a novel second mitochondrial-derived activator of caspase (Smac) mimetic, in ESCC cells. ESCC cell lines were treated with LCL161 or radiation, alone or in combination. Cell proliferation was detected by MTT assay. Radiosensitization was evaluated by clonogenic survival assay. Cell apoptosis was detected by flow cytometry. The results showed that LCL161 potently sensitized ESCC cells to radiation with a sensitization enhancement ratio of 1.4–2.0. LCL161 increased radiation-induced DNA double-stranded breaks and promoted the apoptosis of ESCC cells, which could be abrogated by a pan-caspase inhibitor z-VAD-FMK. Furthermore, LCL161 decreased the level of cIAP1 in ESCC cells in a dose-dependent manner and synthesized with irradiation to promote the activation of caspase 8 and the upregulation of TNFα expression in ESCC cells. In conclusion, LCL161 acts as a strong radiosensitizer in human esophageal cancer cells by inhibiting the expression of cIAP1 and promoting the activation of caspase 8, leading to enhanced apoptosis.


PLOS ONE | 2013

Polymorphisms in XPD Gene Could Predict Clinical Outcome of Platinum-Based Chemotherapy for Non-Small Cell Lung Cancer Patients: A Meta-Analysis of 24 Studies

Qin Qin; Chi Zhang; Xi Yang; Hongcheng Zhu; Baixia Yang; Jing Cai; Hongyan Cheng; Jianxin Ma; Jing Lu; Liangliang Zhan; Jia Liu; Zheming Liu; Liping Xu; Xinchen Sun

Objective Xeroderma pigmentosum group D (XPD) is an essential gene involved in the nucleotide excision repair (NER) pathway. Two commonly studied single nucleotide polymorphisms (SNPs) of XPD (Lys751Gln, A>C, rs13181; Asp312Asn, G>A, rs1799793) are implicated in the modulation of DNA repair capacity, thus related to the responses to platinum-based chemotherapy. Here we performed a meta-analysis to better evaluate the association between the two XPD SNPs and clinical outcome of platinum-based chemotherapy in non-small cell lung cancer (NSCLC) patients. Methods A comprehensive search of PubMed database was conducted to identify relevant articles. Primary outcomes included objective response (i.e., complete response + partial response vs. stable disease + progressive disease), progression-free survival (PFS) and overall survival (OS). The pooled and 95% confidence intervals (CIs) of ORs (odds ratios) and HRs (hazard ratios) were estimated using the fixed or random effect model. Results Twenty-four studies were eligible according to the inclusion criteria. None of the XPD Lys751Gln/Asp312Asn polymorphisms was associated with objective response, PFS or OS in NSCLC patients treated with platinum drugs. However, in stratified analysis by ethnicity, the XPD Lys751Gln (A>C) polymorphism was not significantly associated with increased response in Caucasians (OR = 1.35, 95%CI = 1.0–1.83, P = 0.122 for heterogeneity) but was associated with decreased PFS in Asians (HR = 1.39, 95%CI = 1.07–1.81, P = 0.879 for heterogeneity). Furthermore, a statistically significant difference existed in the estimates of effect between the two ethnicities (P = 0.014 for TR; P<0.001 for PFS). Conclusions XPD Lys751Gln (A>C) may have inverse predictive and prognostic role in platinum-based treatment of NSCLC according to different ethnicities. Further studies are needed to validate our findings.


Journal of Hematology & Oncology | 2014

Small-molecule survivin inhibitor YM155 enhances radiosensitization in esophageal squamous cell carcinoma by the abrogation of G2 checkpoint and suppression of homologous recombination repair

Qin Qin; Hongyan Cheng; Jing Lu; Liangliang Zhan; Jianchao Zheng; Jing Cai; Xi Yang; Liping Xu; Hongcheng Zhu; Chi Zhang; Jia Liu; Jianxin Ma; Xizhi Zhang; Shengbin Dai; Xinchen Sun

BackgroundSurvivin is overexpressed in cancer cells and plays a crucial role in apoptosis evasion. YM155, a small-molecule inhibitor of survivin, could enhance the cytotoxicity of various DNA-damaging agents. Here, we evaluated the radiosensitizaion potential of YM155 in human esophageal squamous cell carcinoma (ESCC).MethodsCell viability was determined by CCK8 assay. The radiosensitization effect of YM155 was evaluated by clonogenic survival and progression of tumor xenograft. Cell cycle progression was determined by flow cytometric analysis. Radiation-induced DNA double strand break (DSB) and homologous recombination repair (HRR) were detected by the staining of γ-H2AX and RAD51, respectively. Expression of survivin and cell cycle regulators was detected by Western blot analysis.ResultsYM155 induced radiosensitization in ESCC cell lines Eca109 and TE13, associated with the abrogation of radiation induced G2/M checkpoint, impaired Rad51 focus formation, and the prolongation of γ-H2AX signaling. G2/M transition markers, including the activation of cyclinB1/Cdc2 kinase and the suppression of Cdc2 Thr14/Tyr15 phosphorylation were induced by YM155 in irradiated cells. The combination of YM155 plus irradiation delayed the growth of ESCC tumor xenografts to a greater extent compared with either treatment modality alone.ConclusionsOur findings suggest that the abrogation of G2 checkpoint and the inhibition of HRR contribute to radiosensitization by YM155 in ESCC cells.


Cell Stress & Chaperones | 2015

Eukaryotic elongation factor 2 kinase confers tolerance to stress conditions in cancer cells

Hongcheng Zhu; Xi Yang; Jia Liu; Lu Zhou; Chi Zhang; Liping Xu; Qin Qin; Liangliang Zhan; Jing Lu; Hongyan Cheng; Xinchen Sun

Eukaryotic elongation factor 2 (eEF2) is a member of the GTP-binding translation elongation factor family that is essential for protein synthesis. eEF2 kinase (eEF2K) is a structurally and functionally unique protein kinase in the calmodulin-mediated signaling pathway. eEF2K phosphorylates eEF2, thereby inhibiting eEF2 function under stressful conditions. eEF2K regulates numerous processes, such as protein synthesis, cell cycle progression, and induction of autophagy and apoptosis in cancer cells. This review will demonstrate the mechanisms underlying eEF2K activity in cancer cells under different stresses, such as nutrient deprivation, hypoxia, and DNA damage via eEF2 regulation. In vivo, in vitro, and clinical studies indicated that eEF2K may be a novel biomarker and therapeutic target for cancer.


PLOS ONE | 2014

PARP-1 Val762Ala polymorphism and risk of cancer: a meta-analysis based on 39 case-control studies.

Qin Qin; Jing Lu; Hongcheng Zhu; Liping Xu; Hongyan Cheng; Liangliang Zhan; Xi Yang; Chi Zhang; Xinchen Sun

Background Poly(ADP-ribose) polymerase-1 (PARP-1) is a nuclear chromatin-associated enzyme involved in several important cellular processes, particularly in the DNA repair system. PARP-1 rs1136410: C>T is among the most studied polymorphisms and likely involved in human carcinogenesis. However, results from previous studies are inconclusive. Thus, a meta-analysis was conducted to derive a more precise estimation of the effects of this enzyme. Methodology and Principal Findings A comprehensive search was conducted in the PubMed and EMBASE databases until December 9, 2013. A total of 39 studies with 16,783 cancer cases and 23,063 control subjects were included in the meta-analysis on the basis of the inclusion and exclusion criteria. No significant association between the PARP-1 Val762Ala polymorphism and cancer risk was found when all of the studies were pooled into the analysis (VA + AA vs. VV: OR = 1.03, 95% CI = 0.95–1.11). The subgroup analysis of cancer types revealed that the –762Ala allele was associated with increased risk of gastric, cervical, and lung cancers and a decreased risk of glioma. In addition, a significantly increased risk of cancer associated with the polymorphism was observed in Asian descendents (VA + AA vs. VV: OR = 1.17, 95% CI = 1.09–1.25; AA vs. VV: OR = 1.28, 95% CI = 1.08–1.51; VA vs. VV: OR = 1.12, 95% CI = 1.04–1.20; AA vs. VA + VV: OR = 1.09, 95% CI = 1.03–1.39). These results also indicated that a joint effect between PARP-1 Val762Ala and XRCC1 Arg399Gln could be involved in the risk of cancer development (OR = 3.53, 95% CI = 1.30–9.59). Conclusion The present meta-analysis provides evidence that the PARP-1 Val762Ala may be involved in cancer development at least in some ethnic groups (Asian) or some specific cancer types (gastric, cervical, and lung cancers, and glioma).


Scientific Reports | 2015

Recombinant human endostatin enhances the radioresponse in esophageal squamous cell carcinoma by normalizing tumor vasculature and reducing hypoxia.

Hongcheng Zhu; Xi Yang; Yuqiong Ding; Jia Liu; Jing Lu; Liangliang Zhan; Qin Qin; Hao Zhang; Xiaochen Chen; Yuehua Yang; Yan Yang; Zheming Liu; Meiling Yang; Xifa Zhou; Hongyan Cheng; Xinchen Sun

The aim of this study was to investigate the effect of recombinant human endostatin (rh-Endo) in combination with radiation therapy (RT) on esophageal squamous cell carcinoma (ESCC) and explore the potential mechanisms. ECA109-bearing nude mice were administered RT and/or rh-Endo treatment. Tumor volume, survival, hypoxia and vascular parameters were recorded during the treatment schedule and follow-up as measures of treatment response. ESCC cell lines (ECA109 and TE13) and human umbilical vein endothelial cells (HUVECs) were developed to investigate the outcomes and toxicities of rh-Endo and RT in vitro. Hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) were also evaluated. In vivo studies of ECA109-bearing xenografts showed that rh-Endo improved the radioresponse, with normalization of tumor vasculature and a reduction in hypoxia. In vitro studies showed that rh-Endo did not radiosensitize ESCC cell lines but did affect endothelial cells with a time- and dose-dependent manner. Studies of the molecular mechanism indicated that the improved radioresponse might be due to crosstalk between cancer cells and endothelial cells involving HIF and VEGF expression. Our data suggest that rh-Endo may be a potential anti-angiogenic agent in ESCC especially when combined with RT. The improved radioresponse arises from normalization of tumor vasculature and a reduction in hypoxia.


Tumor Biology | 2014

Melittin radiosensitizes esophageal squamous cell carcinoma with induction of apoptosis in vitro and in vivo

Hongcheng Zhu; Xi Yang; Jia Liu; Yangyang Ge; Qin Qin; Jing Lu; Liangliang Zhan; Zheming Liu; Hao Zhang; Xiaochen Chen; Chi Zhang; Liping Xu; Hongyan Cheng; Xinchen Sun

Currently, unresectable esophageal squamous cell carcinoma (ESCC) is primarily treated by chemoradiotherapy. However, the outcome has not improved significantly because of radioresistance of cancer cells. This study aimed to determine the radiosensitizing effect of melittin, a novel component of bee venom, in ESCC. ESCC cell lines were irradiated with or without melittin. Cell proliferation was detected by Cell Counting Kit 8 assay. Radiosensitization was evaluated by clonogenic survival assay. Cell apoptosis was detected by flow cytometry. Results show that melittin potently sensitized ESCC cells to radiation with a sensitization enhancement ratio of 1.15–1.42. Radiosensitization was accompanied with enhanced apoptosis and regulated by apoptosis proteins. The results were confirmed by in vivo studies on tumor-bearing xenografts. In summary, these results provide support that melittin may be a potentially promising radiosensitizer in ESCC radiation therapy.


Tumor Biology | 2014

Melittin enhances radiosensitivity of hypoxic head and neck squamous cell carcinoma by suppressing HIF-1α.

Xi Yang; Hongcheng Zhu; Yangyang Ge; Jia Liu; Jing Cai; Qin Qin; Liangliang Zhan; Chi Zhang; Liping Xu; Zheming Liu; Yan Yang; Yuehua Yang; Jianxin Ma; Hongyan Cheng; Xinchen Sun

Hypoxia is a widespread phenomenon present in many human solid tumors and is associated with a poor prognosis and therapy resistance. Here, we tested the feasibility of melittin, a major component of bee venom, on radiosensitization of hypoxic head and neck squamous cell carcinoma (HNSCC). CNE-2 and KB cells were treated with melittin and radiation response was determined. Cell viability, cytotoxicity and apoptosis induction were examined by CCK-8 assay, colony formation assay, and flow cytometry. Expression of hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) proteins were assessed using western blotting. Additionally, we also examined the effect of melittin on tumor growth and radiosensitivity in vivo using a xenograft model of HNSCC. Treatment with melittin resulted in cell growth inhibition, induction of cell apoptosis, and reduction of HIF-1α and VEGF expression, which has been linked to hypoxia cell radioresistance. In addition, intraperitoneal injection of melittin significantly reduced the growth of HNSCC tumors in CNE-2 tumor-bearing mice. These data suggest that melittin enhances radiosensitivity of HNSCC under hypoxia condition, and this is associated with the suppression of HIF-1α expression. Melittin appears to be a potential radiotherapy sensitization agent due to its significant antihypoxia activity.

Collaboration


Dive into the Liangliang Zhan's collaboration.

Top Co-Authors

Avatar

Qin Qin

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xi Yang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xinchen Sun

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongcheng Zhu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongyan Cheng

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Chi Zhang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Lu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jia Liu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Liping Xu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zheming Liu

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge