Network


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

Hotspot


Dive into the research topics where Zhengkai Liao is active.

Publication


Featured researches published by Zhengkai Liao.


Proceedings of the National Academy of Sciences of the United States of America | 2014

RIG-I–like receptor LGP2 protects tumor cells from ionizing radiation

Ryan C. Widau; A. Parekh; M.C. Ranck; Daniel W. Golden; Kiran A. Kumar; Ravi F. Sood; Sean P. Pitroda; Zhengkai Liao; Xiaona Huang; Thomas E. Darga; David Xu; Lei Huang; Jorge Andrade; Bernard Roizman; Ralph R. Weichselbaum; Nikolai N. Khodarev

Significance An undesirable outcome of radiotherapy (ionizing radiation, IR) of cancer is the emergence of radioresistant cells. We report that Laboratory of Genetics and Physiology 2 (LGP2), a resident RIG-I (retinoic acid inducible gene I)–like receptor protein, can induce radioresistance. IR induces interferon and stimulates accumulation of LGP2. In turn, LGP2 shuts off the synthesis of interferon and blocks its cytotoxic effects. Ectopic expression of LGP2 enhances resistance to IR, whereas depletion enhances cytotoxic effects of IR. Here we show that LGP2 is associated with radioresistance in numerous diverse cancer cell lines. Examination of available databases links expression of LGP2 with poor prognosis in cancer patients. An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)–like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNβ. Specifically (i) suppression of LGP2 leads to enhanced IFNβ, (ii) cytotoxic effects following IR correlated with expression of IFNβ inasmuch as inhibition of IFNβ by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNβ and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNβ.


Cancer | 2012

Novel, chimeric, cancer-specific, and radiation-inducible gene promoters for suicide gene therapy of cancer†

Jie Xiong; Wenjie Sun; Wei‐feng Wang; Zhengkai Liao; Fuxiang Zhou; Hai‐yan Kong; Yu Xu; Conghua Xie; Yunfeng Zhou

Although the promoter of the human telomerase reverse transcriptase (hTERT) gene has been widely used in gene therapy for targeted cancer cells, it has some limitations for clinical use because of its low activity and potential toxicity to certain normal cells. To overcome these defects, the authors generated novel chimeric hTERT promoters that contained the radiation‐inducible sequence CC(A/T)6GG (known as CArG elements).


Journal of Translational Medicine | 2011

Gene therapy with tumor-specific promoter mediated suicide gene plus IL-12 gene enhanced tumor inhibition and prolonged host survival in a murine model of Lewis lung carcinoma

Yu Xu; Jin-Xuan Hou; Zhengchun Liu; Haijun Yu; Wenjie Sun; Jie Xiong; Zhengkai Liao; Fuxiang Zhou; Conghua Xie; Yunfeng Zhou

BackgroundGene therapy is a promising therapeutic approach for cancer. Targeted expression of desired therapeutic proteins within the tumor is the best approach to reduce toxicity and improve survival. This study is to establish a more effective and less toxic gene therapy of cancer.MethodsCombined gene therapy strategy with recombinant adenovirus expressing horseradish peroxidase (HRP) mediated by human telomerase reverse transcriptase (hTERT) promoter (AdhTERTHRP) and murine interleukin-12 (mIL-12) under the control of Cytomegalovirus (CMV) promoter (AdCMVmIL-12) was developed and evaluated against Lewis lung carcinoma (LLC) both in vivo and in vitro. The mechanism of action and systemic toxicities were also investigated.ResultsThe combination of AdhTERTHRP/indole-3-acetic acid (IAA) treatment and AdCMVmIL-12 resulted in significant tumor growth inhibition and survival improvement compared with AdhTERTHRP/IAA alone (tumor volume, 427.4 ± 48.7 mm3vs 581.9 ± 46.9 mm3, p = 0.005 on day 15; median overall survival (OS), 51 d vs 33 d) or AdCMVmIL-12 alone (tumor volume, 362.2 ± 33.8 mm3vs 494.4 ± 70.2 mm3, p = 0.046 on day 12; median OS, 51 d vs 36 d). The combination treatment stimulated more CD4+ and CD8+ T lymphocyte infiltration in tumors, compared with either AdCMVmIL-12 alone (1.3-fold increase for CD4+ T cells and 1.2-fold increase for CD8+ T cells, P < 0.01) or AdhTERTHRP alone (2.1-fold increase for CD4+ T cells and 2.2-fold increase for CD8+ T cells, P < 0.01). The apoptotic cells in combination group were significantly increased in comparison with AdCMVmIL-12 alone group (2.8-fold increase, P < 0.01) or AdhTERTHRP alone group (1.6-fold increase, P < 0.01). No significant systematic toxicities were observed.ConclusionsCombination gene therapy with AdhTERTHRP/IAA and AdCMVmIL-12 could significantly inhibit tumor growth and improve host survival in LLC model, without significant systemic adverse effects.


Cancer Letters | 2009

Radiation enhances suicide gene therapy in radioresistant laryngeal squamous cell carcinoma via activation of a tumor-specific promoter

Zhengkai Liao; Chenghu Huang; Fuxiang Zhou; Jie Xiong; Jie Bao; Hongyan Zhang; Wenjie Sun; Conghua Xie; Yunfeng Zhou

Radioresistant cells have been shown to correlate with poor outcome after radiotherapy. Here, we found that human telomerase reverse transcriptase promoter (hTERTp) had lower activity in laryngeal squamous carcinomas cells than in radioresistant variant cells. Combining radiotherapy with plasmid phTERTp-HRP, in which expression of enzyme horseradish peroxidase (HRP) controlled by hTERTp, resulted in increased apoptosis and necrosis of tumor cells after prodrug indole-3-acetic acid (IAA; converted by HRP into a cytotoxin) incubation. Volume and wet weight of xenograft tumor were reduced more in the combination groups. These data suggest that hTERTp has potential use in targeted cancer gene therapy, especially for radioresistant tumors. Combining radiotherapy with hTERTp-HRP/IAA may overcome radioresistance in laryngeal squamous carcinomas cells and amplify the killing effect in targeted cancer cells.


PLOS ONE | 2013

Telomere-Binding Protein TPP1 Modulates Telomere Homeostasis and Confers Radioresistance to Human Colorectal Cancer Cells

Lei Yang; Wenbo Wang; Liu Hu; Xiaoxi Yang; Juan Zhong; Zheng Li; Hui Yang; Han Lei; Haijun Yu; Zhengkai Liao; Fuxiang Zhou; Conghua Xie; Yunfeng Zhou

Background Radiotherapy is one of the major therapeutic strategies in cancer treatment. The telomere-binding protein TPP1 is an important component of the shelterin complex at mammalian telomeres. Our previous reports showed that TPP1 expression was elevated in radioresistant cells, but the exact effects and mechanisms of TPP1 on radiosensitivity is unclear. Principal Findings In this study, we found that elevated TPP1 expression significantly correlated with radioresistance and longer telomere length in human colorectal cancer cell lines. Moreover, TPP1 overexpression showed lengthened telomere length and a significant decrease of radiosensitivity to X-rays. TPP1 mediated radioresistance was correlated with a decreased apoptosis rate after IR exposure. Furthermore, TPP1 overexpression showed prolonged G2/M arrest mediated by ATM/ATR-Chk1 signal pathway after IR exposure. Moreover, TPP1 overexpression accelerated the repair kinetics of total DNA damage and telomere dysfunction induced by ionizing radiation. Conclusions We demonstrated that elevated expressions of TPP1 in human colorectal cancer cells could protect telomere from DNA damage and confer radioresistance. These results suggested that TPP1 may be a potential target in the radiotherapy of colorectal cancer.


Radiation Research | 2010

cDNA expression analysis of a human radiosensitive-radioresistant cell line model identifies telomere function as a hallmark of radioresistance.

Fuxiang Zhou; Jie Xiong; Zhi-Guo Luo; Jing Dai; Haijun Yu; Zhengkai Liao; Han Lei; Conghua Xie; Yunfeng Zhou

Abstract To achieve a more complete understanding of the molecular mechanisms underlying tumor radioresistance, we established a radioresistant cell line from the human larynx squamous cell carcinoma cell line Hep-2 after long-term radiation induction. The biological features of the resulting cell lines were characterized. cDNA microarray technology was used to measure the alterations of gene expression in the radioresistant cells. We found that certain genes associated with DNA repair, cell cycle, apoptosis, etc. were significantly changed. In particular, genes related to telomeres, such as POT1, were significantly altered. Radioresistant cells had higher telomerase activity and longer telomeres than their parental cells. Our research suggests that telomere function is a novel hallmark of cellular radiosensitivity, and the mechanistic link between telomere maintenance and radiosensitivity may involve the genes and pathways we implicated in this study.


Journal of Cancer Research and Clinical Oncology | 2015

Knockdown of telomeric repeat binding factor 2 enhances tumor radiosensitivity regardless of telomerase status

Xiaoxi Yang; Zheng Li; Lei Yang; Han Lei; Haijun Yu; Zhengkai Liao; Fuxiang Zhou; Conghua Xie; Yunfeng Zhou

PurposeTo investigate the effects of TRF2 depletion on radiosensitivity in both the telomerase-positive cell lines (A549) and alternative lengthening of telomere (ALT) cell lines (U2OS).MethodsX-ray irradiation was used to establish two radioresistant cancer models (A549R and U2OSR) from A549 and U2OS. Colony formation assay was applied to examine the radiosensitivity of radioresistant A549R and U2OSR cells and TRF2 low-expression cells. Real-time PCR and TeloTAGGG Telomerase PCR ELISA Kit were performed to examine telomere length and telomerase activity separately. γ-H2AX was detected by immunofluorescence to assess the radiation-induced DSBs.ResultsRadioresistant cancer models were established, in which TRF2 was significantly over-expressed. Low expression of TRF2 protein could enhance the radiosensitivity and induce telomere length of A549 and U2OS cell shortening. In A549 cells with TRF2 down-regulated, the telomerase activity was inhibited, too. TRF2 deficiency increases γ-H2AX foci and fails to protect telomere from radiation.ConclusionThe data suggest that TRF2 is a radioresistant protein in A549 and U2OS cells, and could potentially be a target for radiosensitization of both telomerase-positive and ALT cells in radiotherapy.


PLOS ONE | 2013

Inhibition of UBE2D3 Expression Attenuates Radiosensitivity of MCF-7 Human Breast Cancer Cells by Increasing hTERT Expression and Activity

Wenbo Wang; Lei Yang; Liu Hu; Fen Li; Li Ren; Haijun Yu; Yu Liu; Ling Xia; Han Lei; Zhengkai Liao; Fuxiang Zhou; Conghua Xie; Yunfeng Zhou

The known functions of telomerase in tumor cells include replenishing telomeric DNA and maintaining cell immortality. We have previously shown the existence of a negative correlation between human telomerase reverse transcriptase (hTERT) and radiosensitivity in tumor cells. Here we set out to elucidate the molecular mechanisms underlying regulation by telomerase of radiosensitivity in MCF-7 cells. Toward this aim, yeast two-hybrid (Y2H) screening of a human laryngeal squamous cell carcinoma radioresistant (Hep2R) cDNA library was first performed to search for potential hTERT interacting proteins. We identified ubiquitin-conjugating enzyme E2D3 (UBE2D3) as a principle hTERT-interacting protein and validated this association biochemically. ShRNA-mediated inhibition of UBE2D3 expression attenuated MCF-7 radiosensitivity, and induced the accumulation of hTERT and cyclin D1 in these cells. Moreover, down-regulation of UBE2D3 increased hTERT activity and cell proliferation, accelerating G1 to S phase transition in MCF-7 cells. Collectively these findings suggest that UBE2D3 participates in the process of hTERT-mediated radiosensitivity in human breast cancer MCF-7 cells by regulating hTERT and cyclin D1.


Biochemical and Biophysical Research Communications | 2011

Co-expression of interleukin 12 enhances antitumor effects of a novel chimeric promoter-mediated suicide gene therapy in an immunocompetent mouse model

Yu Xu; Zhengchun Liu; Hai‐yan Kong; Wenjie Sun; Zhengkai Liao; Fuxiang Zhou; Conghua Xie; Yunfeng Zhou

The human telomerase reverse transcriptase (hTERT) promoter has been widely used in target gene therapy of cancer. However, low transcriptional activity limited its clinical application. Here, we designed a novel dual radiation-inducible and tumor-specific promoter system consisting of CArG elements and the hTERT promoter, resulting in increased expression of reporter genes after gamma-irradiation. Therapeutic and side effects of adenovirus-mediated horseradish peroxidase (HRP)/indole-3-acetic (IAA) system downstream of the chimeric promoter were evaluated in mice bearing Lewis lung carcinoma, combining with or without adenovirus-mediated interleukin 12 (IL12) gene driven by the cytomegalovirus promoter. The combination treatment showed more effective suppression of tumor growth than those with single agent alone, being associated with pronounced intratumoral T-lymphocyte infiltration and minor side effects. Our results suggest that the combination treatment with HRP/IAA system driven by the novel chimeric promoter and the co-expression of IL12 might be an effective and safe target gene therapy strategy of cancer.


Journal of Clinical Oncology | 2011

Gene therapy with tumor-specific promoter mediated suicide gene plus interleukin-12 gene enhanced tumor inhibition and prolonged host survival in a murine model of Lewis lung carcinoma.

Yunfeng Zhou; Yu Xu; Haijun Yu; Wenjie Sun; Zhengkai Liao; Fuxiang Zhou; Conghua Xie

e13086 Background: Gene therapy is a promising therapeutic approach for cancer. Targeted expression of desired therapeutic proteins within the tumor is the best approach to reduce toxicity and improve survival. This study was to establish a more effective and less toxic gene therapy of cancer. METHODS Combined gene therapy strategy with recombinant adenovirus expressing horseradish peroxidase (HRP) mediated by human telomerase reverse transcriptase (hTERT) promoter (AdhTERTHRP) and murine interleukin-12 (mIL-12) under the control of Cytomegalovirus (CMV) promoter (AdCMVmIL-12) was developed and evaluated against Lewis lung carcinoma (LLC) both in vivo and in vitro. The mechanism of action and systemic toxicities were also investigated. RESULTS HRP and mIL-12 proteins were detected in LLC cells and subcutaneous tumors. The combination of AdhTERTHRP/IAA treatment and AdCMVmIL-12 resulted in significant tumor growth inhibition and survival improvement compared with AdhTERTHRP/IAA alone (mean tumor volume, 427.4 ± 48.7 mm3 vs 581.9 ± 46.9 mm3, p = 0.005 on day 15; median overall survival (OS), 51 d vs 33 d) or AdCMVmIL-12 alone (mean tumor volume, 362.2 ± 33.8 mm3 vs 494.4 ± 70.2 mm3, p = 0.046 on day 12; median OS, 51 d vs 36 d). The expressed mIL-12 in tumor in combination group stimulated more CD4+ T helper cells and CD8+ cytotoxic T cells, compared with either AdCMVmIL-12 alone (1.3-fold increase for CD4+ T cells and 1.2-fold increase for CD8+ T cells, P<0.01) or AdhTERTHRP alone (2.1-fold increase for CD4+ T cells and 2.2-fold increase for CD8+ T cells, P <0.01). The apoptotic cells in combination group were significantly increased compared with AdCMVmIL-12 alone group (2.8-fold increase, P<0.01) or AdhTERTHRP alone group (1.6-fold increase, P<0.01). No significant systematic toxicities were observed. CONCLUSIONS Combination gene therapy with AdhTERTHRP/IAA and AdCMVmIL-12 could significantly inhibit tumor growth and improve host survival in LLC model, without significant systemic adverse effects.

Collaboration


Dive into the Zhengkai Liao'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