Network


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

Hotspot


Dive into the research topics where Zhi-Yuan Xiao is active.

Publication


Featured researches published by Zhi-Yuan Xiao.


Journal of Experimental & Clinical Cancer Research | 2016

MicroRNA-224 sustains Wnt/β-catenin signaling and promotes aggressive phenotype of colorectal cancer

Tingting Li; Qiuhua Lai; Shu-Yang Wang; Juan-Juan Cai; Zhi-Yuan Xiao; Danling Deng; Liu-Qing He; Hong-Li Jiao; Ya-Ping Ye; Li Liang; Yanqing Ding; Wen-Ting Liao

BackgroundGrowing evidence suggests that Wnt/β-catenin pathway plays an important role in CRC development, progression and metastasis. Aberrant miR-224 expression has been reported in CRC. However, the mechanism of miR-224 promotes both proliferation and metastatic ability largely remains unclear.MethodsReal-time PCR was used to quantify miR-224 expression. Luciferase reporter assays were conducted to confirm the activity of Wnt/β-catenin pathway and target gene associations, and immunofluorescence staining assay was performed to observe the nuclear translocation of β-catenin. Bioinformatics analysis combined with in vivo and vitro functional assays showed the potential target genes, GSK3β and SFRP2, of miR-224. Specimens from forty patients with CRC were analyzed for the expression of miR-224 and the relationship with GSK3β/SFRP2 by real-time PCR and western blot.ResultsBioinformatics and cell luciferase function studies verified the direct regulation of miR-224 on the 3’-UTR of the GSK3β and SFRP2 genes, which leads to the activation of Wnt/β-catenin signaling and the nuclear translocation of β-catenin. In addition, knockdown of miR-224 significantly recovered the expression of GSK3β and SFRP2 and attenuated Wnt/β-catenin-mediated cell metastasis and proliferation. The ectopic upregulation of miR-224 dramatically inhibited the expression of GSK3β/SFRP2 and enhanced CRC proliferation and invasion.ConclusionOur research showed mechanistic links between miR-224 and Wnt/β-catenin in the pathogenesis of CRC through modulation of GSK3β and SFRP2.


Oncotarget | 2016

MiR-384 inhibits human colorectal cancer metastasis by targeting KRAS and CDC42

Yong-Xia Wang; Yan-Ru Chen; Shanshan Liu; Ya-Ping Ye; Hong-Li Jiao; Shu-Yang Wang; Zhi-Yuan Xiao; Wen-Ting Wei; Jun-Feng Qiu; Li Liang; Wen-Ting Liao; Yanqing Ding

Colorectal cancer (CRC) is the third most common cancer worldwide. Metastatic progression is a primary factor contributing to lethality of CRC patients. However, the molecular mechanisms forming early local invasion and distant metastatic colonies are still unclear and the present therapeutic approaches for CRC are unsatisfactory. Therefore, novel therapies targeting metastatic invasion that could prevent tumor spreading and recurrence are urgently needed. Our study showed that the decrease of miR-384 was found in 83.0% (83/100) CRC patients. And low-leveled expression of miR-384 was closely correlated with the invasive depth, lymph node and distant metastasis of CRC. Overexpression of miR-384 could inhibit the invasive and migrating abilities of CRC cells in vitro and the metastatic potential in vivo. Luciferase assays showed that miR-384 repressed the expression of Kirsten Ras (KRAS) and Cell division cycle 42 (CDC42) by directly targeting their 3’-untranslated regions. There is functional and mechanistic relationship between miRNA-384 and KRAS, CDC42 in the invasion and metastasis of CRC. And our findings suggest that miR-384could be a potent therapeutic target for CRC. Restoration of miR-384 expression might provide novel therapeutic approach to the reduction of CRC metastasis.


Oncotarget | 2016

TLE4 promotes colorectal cancer progression through activation of JNK/c-Jun signaling pathway

Shu-Yang Wang; Ke Gao; Danling Deng; Juan-Juan Cai; Zhi-Yuan Xiao; Liu-Qing He; Hong-Li Jiao; Ya-Ping Ye; Run-Wei Yang; Tingting Li; Li Liang; Wen-Ting Liao; Yanqing Ding

The Groucho transcriptional co-repressor TLE4 protein has been shown to be a tumor suppressor in a subset of acute myeloid leukemia. However, little is known about its role in development and progression of solid tumor. In this study, we found that the expression of TLE4 in colorectal cancer (CRC) tissues was significantly higher than that in their matched adjacent intestine epithelial tissues. In addition, high expression of TLE4 was significantly correlated with advanced Dukes stage, lymph node metastasis and poor prognosis of CRC. Moreover, enforced expression of TLE4 in CRC cell lines significantly enhanced proliferation, invasion and tumor growth. On the contrary, knock down of TLE4 repressed cell proliferation, invasion and tumor growth. Furthermore, our study exhibited that the TLE4 promoted cell proliferation and invasion partially via activation of JNK-c-Jun pathway and subsequently increased cyclinD1 and decreased P27Kip1 expression. In conclusion, these results suggested that TLE4, a potential prognostic biomarker for CRC, plays an important role in the development and progression of human CRC.


Cancer Letters | 2015

The tumor-suppressor gene LZTS1 suppresses colorectal cancer proliferation through inhibition of the AKT-mTOR signaling pathway.

Wei Zhou; Mei-Rong He; Hong-Li Jiao; Liu-Qing He; Danling Deng; Juan-Juan Cai; Zhi-Yuan Xiao; Ya-Ping Ye; Yanqing Ding; Wen-Ting Liao; Si-De Liu

The Leucine zipper tumor suppressor gene 1 (LZTS1/FEZ1) gene was originally identified as a potential tumor suppressor. However, the expression pattern and the role of LZTS1 in the progression of colorectal cancer (CRC) have not been well characterized. Herein, we reported that LZTS1 was markedly reduced in CRC tissues compared with matched adjacent normal intestine epithelial tissues. In analysis of 160 CRC specimens, we revealed that decreased expression of LZTS1 was correlated to aggressive characteristics and poor survival of patients with CRC. Moreover, we found that expression of LZTS1 in CRC cells significantly inhibited cell proliferation in vitro and prohibited tumor growth in vitro. On the contrary, silence of LZTS1 promoted cell proliferation and tumor growth in CRC cells. Furthermore, we demonstrated that LZTS1 inhibited cell proliferation and tumor growth in CRC in part via suppression of AMT-mTOR, subsequently down-regulating p27Kip and up-regulating cyclin D1. These findings suggest that LZTS1 plays a potential tumor suppressor role in CRC progression and represents a valuable clinical prognostic marker of this disease.


Oncotarget | 2016

miR-450b-5p induced by oncogenic KRAS is required for colorectal cancer progression

Ya-Ping Ye; Ping Wu; Chun-cai Gu; Danling Deng; Hong-Li Jiao; Tingting Li; Shu-Yang Wang; Yong-Xia Wang; Zhi-Yuan Xiao; Wen-Ting Wei; Yan-Ru Chen; Jun-Feng Qiu; Run-Wei Yang; Jie Lin; Li Liang; Wen-Ting Liao; Yanqing Ding

The development and progression of CRC are regarded as a complicated network and progressive event including genetic and/or epigenetic alterations. Recent researches revealed that MicroRNAs are biomarkers and regulators of CRC progression. Analyses of published microarray datasets revealed that miR-450b-5p was highly up-regulated in CRC tissues. In addition, high expression of miR-450b-5p was significantly associated with KRAS mutation. However, the role of miR-450b-5p in the progression of CRC remains unknown. Here, we sought to validate the expression of miR-450b-5p in CRC tissues and investigate the role and underlying mechanism of miR-450b-5p in the progression of CRC. The results revealed that miR-450b-5p was up-regulated in CRC tissues, high expression level of miR-450b-5p was positively associated with poor differentiation, advanced TNM classification and poor prognosis. Moreover, miR-450b-5p was especially high in KRAS-mutated cell lines and could be up-regulated by KRAS/AP-1 signaling. Functional validation revealed that overexpression of miR-450b-5p promoted cell proliferation and tumor growth while inhibited apoptosis of CRC cells. Furthermore, we demonstrated that miR-450b-5p directly bound the 3′-UTRs of SFRP2 and SIAH1, and activated Wnt/β-Catenin signaling. In conclusion, miR-450b-5p induced by oncogenic KRAS is required for colorectal cancer progression. Collectively, our work helped to understand the precise role of miR-450b-5p in the progression of CRC, and might promote the development of new therapeutic strategies against CRC.


Cell Death and Disease | 2018

STX2 promotes colorectal cancer metastasis through a positive feedback loop that activates the NF-κB pathway

Yong-Xia Wang; Honghai Xu; Hong-Li Jiao; Shu-Yang Wang; Zhi-Yuan Xiao; Yali Zhao; Jiaxin Bi; Wen-Ting Wei; Shanshan Liu; Jun-Feng Qiu; Tingting Li; Li Liang; Ya-Ping Ye; Wen-Ting Liao; Yanqing Ding

Metastatic progression is the main contributor to the poor prognosis of colorectal cancer (CRC). Thus, identifying the determinants of CRC metastasis will be of great significance. Based on our previous bioinformatics analysis, Syntaxin2 (STX2) may be upregulated and correlated with the poor prognosis of CRC patients. In this study, we found that STX2 expression was associated with CRC invasion and metastasis and poor patient survival. Gain- and loss-of-function analyses demonstrated that STX2 functioned as a key oncogene by promoting CRC invasion and metastasis. Mechanistically, STX2 selectively interacted with tumor necrosis factor receptor-associated factor 6 (TRAF6) and activated the nuclear transcription factor-κB (NF-κB) signaling pathway. Furthermore, chromatin immunoprecipitation (ChIP) analysis revealed that NF-κB directly bound to the STX2 promoter and drove STX2 transcription. Therefore, STX2 activated the NF-κB pathway, and in turn, NF-κB increased STX2 expression, forming a positive signaling loop that eventually promoted CRC metastasis. Collectively, our results reveal STX2 as a crucial modulator of the aggressive CRC phenotype and highlight STX2 as a potential prognostic biomarker and therapeutic target for combating CRC metastasis.


Clinical Cancer Research | 2017

Downregulation of SAFB Sustains the NF-κB Pathway by Targeting TAK1 during the Progression of Colorectal Cancer

Hong-Li Jiao; Ya-Ping Ye; Run-Wei Yang; Hui-Ying Sun; Shu-Yang Wang; Yong-Xia Wang; Zhi-Yuan Xiao; Liu-Qing He; Juan-Juan Cai; Wen-Ting Wei; Yan-Ru Chen; Chun-cai Gu; Yue-Long Cai; Yun-Teng Hu; Qiuhua Lai; Jun-Feng Qiu; Li Liang; Guangwen Cao; Wen-Ting Liao; Yanqing Ding

Purpose: To investigate the role and the underlying mechanism of scaffold attachment factor B (SAFB) in the progression of colorectal cancer (CRC). Experimental Design: SAFB expression was analyzed in the Cancer Outlier Profile Analysis of Oncomine and in 175 paraffin-embedded archived CRC tissues. Gene Ontology analyses were performed to explore the mechanism of SAFB in CRC progression. Western blot, RT-PCR, luciferase assay, and chromatin immunoprecipitation (ChIP) were used to detect the regulation of transforming growth factor-β–activated kinase 1 (TAK1) and NF-κB signaling by SAFB. The role of SAFB in invasion, metastasis, and angiogenesis was investigated using in vitro and in vivo assays. The relationship between SAFB and TAK1 was analyzed in CRC tissues. Results: SAFB was downregulated in CRC tissues, and low expression of SAFB was significantly associated with an aggressive phenotype and poorer survival of CRC patients. The downregulation of SAFB activated NF-κB signaling by targeting the TAK1 promoter. Ectopic expression of SAFB inhibited the development of aggressive features and metastasis of CRC cells both in vitro and in vivo. The overexpression of TAK1 could rescue the aggressive features in SAFB-overexpressed cells. Furthermore, the expression of SAFB in CRC tissues was negatively correlated with the expression of TAK1- and NF-κB–related genes. Conclusions: Our results show that SAFB regulated the activity of NF-κB signaling in CRC by targeting TAK1. This novel mechanism provides a comprehensive understanding of both SAFB and the NF-κB signaling pathway in the progression of CRC and indicates that the SAFB–TAK1–NF-κB axis is a potential target for early therapeutic intervention in CRC progression. Clin Cancer Res; 23(22); 7108–18. ©2017 AACR.


Cancer Cell International | 2017

miR-422a inhibits cell proliferation in colorectal cancer by targeting AKT1 and MAPK1

Wen-Ting Wei; Xin-Xin Nian; Shu-Yang Wang; Hong-Li Jiao; Yong-Xia Wang; Zhi-Yuan Xiao; Run-Wei Yang; Yanqing Ding; Ya-Ping Ye; Wen-Ting Liao

BackgroundmiRNAs are regarded as molecular biomarkers and therapeutic targets for colorectal cancer (CRC), a series of miRNAs have been proven to involve into CRC carcinogenesis, invasion and metastasis. Aberrant miR-422a expression and its roles have been reported in some cancers. However, the function and underlying mechanism of miR-422a in the progression of CRC remain largely unknown.MethodsReal-time PCR were used to quantify miR-422a expression in CRC tissues. Both vivo and vitro functional assays showed miR-422a inhibits CRC cell proliferation. Target prediction program (miRBase) and luciferase reporter assays were conducted to confirm the target genes AKT1 and MAPK1 of miR-422a. Specimens from 50 patients with CRC were analyzed for the correlation between the expression of miR-422a and the expression of the target genes AKT1 and MAPK1 by real-time PCR.ResultsMiR-422a was down‑regulated in CRC tissues and cell lines. Ectopic expression of miR-422a inhibited cell proliferation and tumor growth ability; inhibition of endogenous miR-422a, by contrast, promoted cell proliferation and tumor growth ability of CRC cells. MiR-422a directly targets 3′-UTR of the AKT1 and MAPK1, down-regulation of miR-422a led to the activation of Raf/MEK/ERK and PI3K/AKT signaling pathways to promote cell proliferation in CRC. In addition, miR-422a expression was negatively correlated with the expressions of AKT1 and MAPK1 in CRC tissues.ConclusionmiR-422a inhibits cell proliferation in colorectal cancer by targeting AKT1 and MAPK1.


Neoplasia | 2018

FOXF1 Induces Epithelial-Mesenchymal Transition in Colorectal Cancer Metastasis by Transcriptionally Activating SNAI1

Shu-Yang Wang; Shanshan Yan; Shaowei Zhu; Yali Zhao; Junyu Yan; Zhi-Yuan Xiao; Jiaxin Bi; Jun-Feng Qiu; Dan Zhang; Zexuan Hong; Lingjie Zhang; Chengmei Huang; Tingting Li; Li Liang; Wen-Ting Liao; Hong-Li Jiao; Yanqing Ding; Ya-Ping Ye

Forkhead Box F1 (FOXF1) has been recently implicated in cancer progression and metastasis of lung cancer and breast cancer. However, the biological functions and underlying mechanisms of FOXF1 in the regulation of the progression of colorectal cancer (CRC) are largely unknown. We showed that FOXF1 was up-regulated in 93 paraffin-embedded archived human CRC tissue, and both high expression and nuclear location of FOXF1 were significantly associated with the aggressive characteristics and poorer survival of CRC patients. The GSEA analysis showed that the higher level of FOXF1 was positively associated with an enrichment of EMT gene signatures, and exogenous overexpression of FOXF1 induced EMT by transcriptionally activating SNAI1. Exogenous overexpression FOXF1 functionally promoted invasion and metastasis features of CRC cells, and inhibition of SNAI1 attenuates the invasive phenotype and metastatic potential of FOXF1-overexpressing CRC cells. Furthermore, the results of the tissue chip showed that the expression of FOXF1 was positively correlated with SNAI1 in CRC tissues chip. These results suggested that FOXF1 plays a critical role in CRC metastasis by inducing EMT via transcriptional activation of SNAI1, highlighting a potential new therapeutic strategy for CRC.


Cancer Letters | 2018

FOXF1 promotes angiogenesis and accelerates bevacizumab resistance in colorectal cancer by transcriptionally activating VEGFA

Shu-Yang Wang; Zhi-Yuan Xiao; Zexuan Hong; Hong-Li Jiao; Shaowei Zhu; Yali Zhao; Jiaxin Bi; Jun-Feng Qiu; Dan Zhang; Junyu Yan; Lingjie Zhang; Chengmei Huang; Tingting Li; Li Liang; Wen-Ting Liao; Ya-Ping Ye; Yanqing Ding

Forkhead box F1 (FOXF1) has been recently implicated in the progression and metastasis of lung cancer and breast cancer. However, the biological functions and underlying mechanisms by which FOXF1 regulates the progression of colorectal cancer (CRC) are largely unknown. As shown in our previous study, FOXF1 is upregulated in 182 CRC tissues, and elevated FOXF1 expression is significantly associated with microvessel density and advanced TNM (T = primary tumour; N = regional lymph nodes; M = distant metastasis) stages. In this study, 43 CRC tissues collected from patients who underwent treatment with first-line standard chemotherapeutic regimens in combination with bevacizumab were used to explore the correlation between FOXF1 expression and resistance to bevacizumab. In addition, FOXF1 regulated angiogenesis by inducing the transcription of vascular endothelial growth factor A1 (VEGFA) in vitro and in vivo. Furthermore, upregulation of FOXF1 enhanced bevacizumab resistance in CRC, and inhibition of VEGFA attenuated angiogenesis and bevacizumab resistance in FOXF1-overexpressing CRC cells. These results suggest that FOXF1 plays critical roles in CRC angiogenesis and bevacizumab resistance by inducing VEGFA transcription and that FOXF1 represents a potentially new therapeutic strategy and biomarker for anti-angiogenic therapy against CRC.

Collaboration


Dive into the Zhi-Yuan Xiao's collaboration.

Top Co-Authors

Avatar

Hong-Li Jiao

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Wen-Ting Liao

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Ya-Ping Ye

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Yanqing Ding

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Shu-Yang Wang

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Liang

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Tingting Li

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun-Feng Qiu

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Danling Deng

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Juan-Juan Cai

Southern Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge