Network


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

Hotspot


Dive into the research topics where Aiguo Liu is active.

Publication


Featured researches published by Aiguo Liu.


Cancer Research | 2011

STAT3 Is Necessary for Proliferation and Survival in Colon Cancer–Initiating Cells

Li Lin; Aiguo Liu; Zhengang Peng; Huey-Jen L Lin; Pui-Kai Li; Chenglong Li; Jiayuh Lin

STAT3 is constitutively activated in colon cancer but its contributions in cancer-initiating cells have not been explored. In this study, we characterized STAT3 in aldehyde dehydrogenase (ALDH)-positive (ALDH(+)) and CD133-positive (CD133(+)) subpopulations of human colon tumor cells that exhibited more potent tumor-initiating ability than ALDH(-)/CD133(-) cells in tumor xenograft assays in mice. We found that ALDH(+)/CD133(+) cells expressed higher levels of the active phosphorylated form of STAT3 than either ALDH(-)/CD133(-) or unfractionated colon cancer cells. STAT3 inhibition by RNA interference-mediated knockdown or small-molecule inhibitors LLL12 or Stattic blocked downstream target gene expression, cell viability, and tumorsphere-forming capacity in cancer-initiating cells. Similarly, treatment of mouse tumor xenografts with STAT3 short hairpin RNA (shRNA), interleukin 6 shRNA, or LLL12 inhibited tumor growth. Our results establish that STAT3 is constitutively activated in colon cancer-initiating cells and that these cells are sensitive to STAT3 inhibition. These findings establish a powerful rationale to develop STAT3 inhibitory strategies for treating advanced colorectal cancers.


Journal of Medicinal Chemistry | 2011

Fragment-Based Drug Design and Drug Repositioning Using Multiple Ligand Simultaneous Docking (MLSD): Identifying Celecoxib and Template Compounds as Novel Inhibitors of Signal Transducer and Activator of Transcription 3 (STAT3)

Huameng Li; Aiguo Liu; Zhenjiang Zhao; Yufang Xu; Jiayuh Lin; David Jou; Chenglong Li

We describe a novel method of drug discovery using MLSD and drug repositioning, with cancer target STAT3 being used as a test case. Multiple drug scaffolds were simultaneously docked into hot spots of STAT3 by MLSD, followed by tethering to generate virtual template compounds. Similarity search of virtual hits on drug database identified celecoxib as a novel inhibitor of STAT3. Furthermore, we designed two novel lead inhibitors based on one of the lead templates and celecoxib.


Cancer Prevention Research | 2011

Celecoxib Inhibits Interleukin-6/Interleukin-6 Receptor–Induced JAK2/STAT3 Phosphorylation in Human Hepatocellular Carcinoma Cells

Yan Liu; Aiguo Liu; Huameng Li; Chenglong Li; Jiayuh Lin

Growing evidence shows an association between chronic liver inflammation and hepatocellular carcinoma (HCC) development. STAT3, which is associated with inflammation and cellular transformation, is constitutively activated in human HCC tissues but not in normal human liver tissues. Although interleukin-6 (IL-6) is elevated in the serum of patients with HCC, it is not fully understood whether STAT3 constitutive activation is positively correlated with autocrine IL-6 secreted by HCC cells. Here, we reported that in HCC cells, the elevated levels of both IL-6 and IL-6 receptor (IL-6R, gp80), not IL-6 alone, correlated with STAT3 activation. We also explored whether the anticancer effects of celecoxib, an anti-inflammatory drug, may be due to the inhibition of the IL-6/STAT3 pathway in HCC cells. Our results showed that celecoxib decreased STAT3 phosphorylation by reducing Janus-activated kinase (JAK2) phosphorylation and caused apoptosis in HCC cells. Celecoxib could also block exogenous IL-6–induced STAT3 phosphorylation and nuclear translocation. Moreover, we observed more significant inhibition of cell viability when celecoxib was combined with doxorubicin or sorafenib. We conclude that the elevated levels of IL-6/IL-6R may be correlated with STAT3 activation in HCC cells. Celecoxib may be a candidate for HCC therapy through blocking IL-6/STAT3 pathway and can be combined with other anticancer drugs to reduce drug resistance caused by IL-6/STAT3 signals. Cancer Prev Res; 4(8); 1296–305. ©2011 AACR.


Investigational New Drugs | 2012

Small molecules, LLL12 and FLLL32, inhibit STAT3 and exhibit potent growth suppressive activity in osteosarcoma cells and tumor growth in mice

Grace-Ifeyinwa Onimoe; Aiguo Liu; Li Lin; Chang-Ching Wei; Eric B. Schwartz; Deepak Bhasin; Chenglong Li; James R. Fuchs; Pui-Kai Li; Peter J. Houghton; Amanda M. Termuhlen; Thomas G. Gross; Jiayuh Lin

SummaryConstitutive activation of Signal Transducers and Activators of Transcription 3 (STAT3) is frequently detected in osteosarcoma, and hence, may serve as a therapeutic target. In order to target STAT3, we tested two new STAT3 inhibitors, LLL12 and FLLL32. LLL12 and FLLL32 inhibit STAT3 phosphorylation and STAT3 downstream targets. LLL12 and FLLL32 also inhibit IL-6 induced STAT3 phosphorylation. The inhibition of STAT3 by LLL12 and FLLL32 resulted in the induction of apoptosis, reduction of plating efficiency, and migration in osteosarcoma cells. Furthermore, LLL12 and FLLL32 inhibited SJSA osteosarcoma cells and OS-33 tumor growth in murine xenografts. These results provide evidence that constitutive STAT3 signaling is required for osteosarcoma survival and migration in vitro and tumor growth in vivo. Blocking persistent STAT3 signaling by LLL12 and FLLL32 may be a novel therapeutic approach for osteosarcoma.


Apoptosis | 2011

XZH-5 inhibits STAT3 phosphorylation and causes apoptosis in human hepatocellular carcinoma cells

Yan Liu; Aiguo Liu; Zhenghu Xu; Wenying Yu; Hong Wang; Chenglong Li; Jiayuh Lin

Growing evidence has demonstrated that signal transducer and activator of transcription 3 (STAT3), which is constitutively activated in patients with hepatocellular carcinoma (HCC), plays an important role in HCC development, progression, and prognosis. Interleukin-6 (IL-6) is a multifunctional inflammatory cytokine, which may induce STAT3 activation in a variety of human cancers. In this study, we demonstrated that blocking STAT3 phosphorylation with STAT3 small molecule inhibitors SD-1029 and Stattic caused apoptosis in HCC cells. Then we characterized the inhibitory effects of a novel small molecule XZH-5 on STAT3 phosphorylation in HCC cell lines. XZH-5 reduced constitutive STAT3 phosphorylation at Tyr705 and the expression of STAT3 downstream genes. The inhibition of STAT3 in HCC cells resulted in the induction of apoptosis and reduction of colony forming ability. In addition, XZH-5 also inhibited IL-6-induced STAT3 phosphorylation, nuclear translocation and STAT3 DNA binding activity. In contrast, it had no effect on IFN-γ-induced STAT1 phosphorylation, indicating the more selective effects on STAT3. These results suggested that XZH-5 may serve as a lead compound for further development of STAT3 specific small molecule inhibitors for HCC therapy.


PLOS ONE | 2012

XZH-5 Inhibits STAT3 Phosphorylation and Enhances the Cytotoxicity of Chemotherapeutic Drugs in Human Breast and Pancreatic Cancer Cells

Aiguo Liu; Yan Liu; Zhigang Jin; Qun Hu; Li-li Lin; David Jou; Jing Yang; Zhenghu Xu; Hong Tian Wang; Chenglong Li; Jiayuh Lin

Constitutive activation of Signal Transducers and Activators of Transcription 3 (STAT3) signaling is frequently detected in breast and pancreatic cancer. Inhibiting constitutive STAT3 signaling represents a promising molecular target for therapeutic approach. Using structure-based design, we developed a non-peptide cell-permeable, small molecule, termed as XZH-5, which targeted STAT3 phosphorylation. XZH-5 was found to inhibit STAT3 phosphorylation (Tyr705) and induce apoptosis in human breast and pancreatic cancer cell lines expressing elevated levels of phosphorylated STAT3. XZH-5 could also inhibit interleukin-6-induced STAT3 phosphorylation in cancer cell lines expressing low phosphorylated STAT3. Inhibition of STAT3 signaling by XZH-5 was confirmed by the down-regulation of downstream targets of STAT3, such as Cyclin D1, Bcl-2, and Survivin at mRNA level. In addition, XZH-5 inhibited colony formation, cell migration, and enhanced the cytotoxicity of chemotherapeutic drugs when combined with Doxorubicin or Gemcitabine. Our results indicate that XZH-5 may be a potential therapeutic agent for breast and pancreatic cancers with constitutive STAT3 signaling.


Cancer Science | 2011

Novel small molecule, XZH-5, inhibits constitutive and interleukin-6-induced STAT3 phosphorylation in human rhabdomyosarcoma cells.

Aiguo Liu; Yan Liu; Zhenghu Xu; Wenying Yu; Hong Wang; Chenglong Li; Jiayuh Lin

Signal transducers and activators of transcription 3 (STAT3) signaling is constitutively activated in many types of human cancers and cancer cell lines and represents a promising target for cancer therapy. We previously reported that the STAT3 signaling pathway is constitutively activated in human rhabodomyosarcoma cell lines (RH28, RH30 and RD2). We also demonstrated that inhibition of the STAT3 pathway led to apoptosis in human rhabdomyosarcoma cells. In the present study, we investigated the inhibitory effects of a novel small molecule, XZH‐5, on the STAT3 signaling pathway in human rhabdomyosarcoma cells. XZH‐5 was designed based on STAT3 structure, and our idea was to design peptide mimics to bind to the phosphorylated Tyr705 site and the side pocket. We found that XZH‐5 downregulated STAT3 phosphorylation. The inhibition of STAT3 by XZH‐5 was confirmed by the inhibition of STAT3 DNA binding ability and the downregulation of STAT3 downstream genes, such as Bcl‐2, Bcl‐xL, Cyclin D1 and Survivin; we also demonstrated that blockade of STAT3 phosphorylation in human rhabdomyosarcoma cells with XZH‐5 caused apoptosis and suppressed colony‐forming ability and cell migration. In addition to reducing constitutive STAT3 phosphorylation, XZH‐5 also exhibited the potency to block interleukin‐6 (IL‐6)‐induced STAT3 phosphorylation and nuclear translocation but did not inhibit the stimulation of STAT1 phosphorylation by interferon (IFN)‐γ. Our findings indicate that XZH‐5 has the potential for targeting human rhabdomyosarcoma cells expressing constitutive STAT3. (Cancer Sci 2011; 102: 1381–1387)


Cellular Physiology and Biochemistry | 2013

Berberine Induces Apoptosis in p53-Null Leukemia Cells by Down-Regulating XIAP at the Post-Transcriptional Level

Jian Liu; Xiaoling Zhang; Aiguo Liu; Shuangyou Liu; Liuqing Zhang; Bei Wu; Qun Hu

Background: Berberine exerts anticancer activities both in vitro and in vivo through different mechanisms. However, the underlying molecular mechanisms of berberine induced p53-independent apoptosis remain unclear. Methods: The p53-null leukemia cell line EU-4 cells were exposed to berberine. Then the cell viability and apoptosis were determined. Western blot and PCR were employed to detect the expression of apoptosis related protein, XIAP and MDM2. Small interfering RNA (siRNA) was applied to knock down endogenous expression of MDM2 and XIAP. Results: Berberine induced p53-independent, XIAP-mediated apoptotic cell death in p53-null leukemia cells. Treatment with berberine resulted in suppression of XIAP protein in a dose- and time- dependent manner. Berberine induced down-regulation of XIAP protein involving inhibition of MDM2 expression and a proteasome-dependent pathway. Moreover, inhibition of XIAP by berberine or siRNA increased the sensitivity of leukemia cells to doxorubicin-induced apoptosis. Conclusion: Our findings characterize the molecular mechanisms of berberine-induced caspase activation and subsequent apoptosis, and berberine may be a novel candidate inducer of apoptosis in leukemia cells, which normally lack p53 expression.


Bioorganic & Medicinal Chemistry | 2015

Design, synthesis and evaluation of XZH-5 analogues as STAT3 inhibitors

Philias Daka; Aiguo Liu; Chamini V. Karunaratne; Erika Csatary; Cameron Williams; Hui Xiao; Jiayuh Lin; Zhenghu Xu; Richard C. Page; Hong Wang

Inhibition of the signaling pathways of signal transducer and activator of transcription 3 (STAT 3) has shown to be a promising strategy to combat cancer. In this paper we report the design, synthesis and evaluation of a novel class of small molecule inhibitors, that is, XZH-5 and its analogues, as promising leads for further development of STAT3 inhibitors. Preliminary SARs was established for XZH-5 and its derivatives; and the binding modes were predicted by molecular docking. Lead compounds with IC50 as low as 6.5μM in breast cancer cell lines and 7.6μM in pancreatic cancer cell lines were identified.


Molecular Cancer | 2017

MicroRNA-218 functions as a tumor suppressor in lung cancer by targeting IL-6/STAT3 and negatively correlates with poor prognosis

Yan Yang; Lili Ding; Qun Hu; Jia Xia; Junjie Sun; Xudong Wang; Hua Xiong; Deepak Gurbani; Lianbo Li; Yan Liu; Aiguo Liu

BackgroundAberrant expression of microRNAs in different human cancer types has been widely reported. MiR-218 acts as a tumor suppressor in diverse human cancer types impacting regulation of multiple genes in oncogenic pathways. Here, we evaluated the expression and function of miR-218 in human lung cancer and ALDH positive lung cancer cells to understand the potential mechanisms responsible for disease pathology. Also, the association between its host genes and the target genes could be useful towards the better understanding of prognosis in clinical settings.MethodsPublicly-available data from The Cancer Genome Atlas (TCGA) was mined to compare the levels of miR-218 and its host gene SLIT2/3 between lung cancer tissues and normal lung tissues. Transfection of miR-218 to investigate its function in lung cancer cells was done and in vivo effects were determined using miR-218 expressing lentiviruses. Aldefluor assay and Flow cytometry was used to quantify and enrich ALDH positive lung cancer cells. Levels of miR-218, IL-6R, JAK3 and phosphorylated STAT3 were compared in ALDH1A1 positive and ALDH1A1 negative cells. Overexpression of miR-218 in ALDH positive cells was carried to test the survival by tumorsphere culture. Finally, utilizing TCGA data we studied the association of target genes of miR-218 with the prognosis of lung cancer.ResultsWe observed that the expression of miR-218 was significantly down-regulated in lung cancer tissues compared to normal lung tissues. Overexpression of miR-218 decreased cell proliferation, invasion, colony formation, and tumor sphere formation in vitro and repressed tumor growth in vivo. We further found that miR-218 negatively regulated IL-6 receptor and JAK3 gene expression by directly targeting the 3′-UTR of their mRNAs. In addition, the levels of both miR-218 host genes and the components of IL-6/STAT3 pathway correlated with prognosis of lung cancer patients.ConclusionsMiR-218 acts as a tumor suppressor in lung cancer via IL-6/STAT3 signaling pathway regulation.

Collaboration


Dive into the Aiguo Liu's collaboration.

Top Co-Authors

Avatar

Jiayuh Lin

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yan Liu

Vanderbilt University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qun Hu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Li Lin

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Li Xie

Huazhong University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge