Network


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

Hotspot


Dive into the research topics where Hualiang Xiao is active.

Publication


Featured researches published by Hualiang Xiao.


Journal of Immunology | 2012

Tumor-Associated Microglia/Macrophages Enhance the Invasion of Glioma Stem-like Cells via TGF-β1 Signaling Pathway

Xian-zong Ye; Sen-lin Xu; Yan-hong Xin; Shi-cang Yu; Yi-Fang Ping; Lu Chen; Hualiang Xiao; Bin Wang; Liang Yi; Qing-liang Wang; Xue-feng Jiang; Lang Yang; Peng Zhang; Cheng Qian; You-Hong Cui; Xia Zhang; Xiu-wu Bian

The invasion of malignant glioma cells into the surrounding normal brain tissues is crucial for causing the poor outcome of this tumor type. Recent studies suggest that glioma stem-like cells (GSLCs) mediate tumor invasion. However, it is not clear whether microenvironment factors, such as tumor-associated microglia/macrophages (TAM/Ms), also play important roles in promoting GSLC invasion. In this study, we found that in primary human gliomas and orthotopical transplanted syngeneic glioma, the number of TAM/Ms at the invasive front was correlated with the presence of CD133+ GSLCs, and these TAM/Ms produced high levels of TGF-β1. CD133+ GSLCs isolated from murine transplanted gliomas exhibited higher invasive potential after being cocultured with TAM/Ms, and the invasiveness was inhibited by neutralization of TGF-β1. We also found that human glioma-derived CD133+ GSLCs became more invasive upon treatment with TGF-β1. In addition, compared with CD133− committed tumor cells, CD133+ GSLCs expressed higher levels of type II TGF-β receptor (TGFBR2) mRNA and protein, and downregulation of TGFBR2 with short hairpin RNA inhibited the invasiveness of GSLCs. Mechanism studies revealed that TGF-β1 released by TAM/Ms promoted the expression of MMP-9 by GSLCs, and TGFBR2 knockdown reduced the invasiveness of these cells in vivo. These results demonstrate that TAM/Ms enhance the invasiveness of CD133+ GSLCs via the release of TGF-β1, which increases the production of MMP-9 by GSLCs. Therefore, the TGF-β1 signaling pathway is a potential therapeutic target for limiting the invasiveness of GSLCs.


Journal of Neuroimmunology | 2011

Glioma-initiating cells: A predominant role in microglia/macrophages tropism to glioma

Liang Yi; Hualiang Xiao; Minhui Xu; Xian-zong Ye; Jun Hu; Fei Li; Mei Li; Chunxia Luo; Shi-cang Yu; Xiu-wu Bian; Hua Feng

The relationship between cancer-initiating cells and cancer-related inflammation is unclear. Exploring the interaction between glioma-initiating cells (GICs) and tumor-associated microglia/macrophages (TAM/Ms) may offer us an opportunity to further understand the inflammatory response in glioma and the cellular/molecular features of the GIC niche. Here,we reported a positive correlation between the infiltration of TAM/Ms and the density of GICs. The capacity of GICs to recruit TAM/Ms was stronger than that of adhesive glioma cells (AGCs) in vitro. In vivo experiments suggested that implantations formed by GICs had a higher level of TAM/M infiltration than those formed by AGCs. Our studies indicate a predominant role of GICs in microglia/macrophages tropism to glioma and a close positive correlation between the distribution of GICs and TAM/Ms. As an important part of cancer-related inflammation, TAM/Ms may participate in the architecture of the GIC niche.


Stem Cells | 2012

Connexin 43 Reverses Malignant Phenotypes of Glioma Stem Cells by Modulating E-Cadherin†‡§

Shi-cang Yu; Hualiang Xiao; Xue-feng Jiang; Qing-liang Wang; Yan Li; Xiao-jun Yang; Yi-Fang Ping; Jiang Jie Duan; Jian-yong Jiang; Xian-zong Ye; Sen-lin Xu; Yang‐Hong Xin; Xiao-hong Yao; Jian-hong Chen; Wei‐Hua Chu; Wei Sun; Bing Wang; Ji Ming Wang; Xia Zhang; Xiu-wu Bian

Malfunctioned gap junctional intercellular communication (GJIC) has been thought associated with malignant transformation of normal cells. However, the role of GJIC‐related proteins such as connexins in sustaining the malignant behavior of cancer stem cells remains unclear. In this study, we obtained tumorspheres formed by glioma stem cells (GSCs) and adherent GSCs and then examined their GJIC. All GSCs showed reduced GJIC, and differentiated glioma cells had more gap junction‐like structures than GSCs. GSCs expressed very low level of connexins, Cx43 in particular, which are key components of gap junction. We observed hypermethylation in the promoter of gap junction protein α1, which encodes Cx43 in GSCs. Reconstitution of Cx43 in GSCs inhibited their capacity of self‐renewal, invasiveness, and tumorigenicity via influencing E‐cadherin and its coding protein, which leads to changes in the expression of Wnt/β‐catenin targeting genes. Our results suggest that GSCs require the low expression of Cx43 for maintaining their malignant phenotype, and upregulation of Cx43 might be a potential strategy for treatment of malignant glioma. STEM CELLS 2012; 30:108‐120.


Biochemical and Biophysical Research Communications | 2013

High expression of lysine-specific demethylase 1 correlates with poor prognosis of patients with esophageal squamous cell carcinoma

Yanyan Yu; Bin Wang; Kun Zhang; Zengjie Lei; Yan Guo; Hualiang Xiao; Jun Wang; Lilin Fan; Chunhui Lan; Yanling Wei; Qiang Ma; Li Lin; Chengyi Mao; Xin Yang; Xiaodi Chen; Yan Li; Yun Bai; Dongfeng Chen

Recent studies have elucidated the role of lysine-specific demethylase 1 (LSD1), a member of the histone demethylases, in epigenetic regulation of tumor suppressing/promoting genes and neoplastic growth. However, the expression of LSD1 in patients with esophageal squamous cell carcinoma (ESCC) is still unknown. Here, we reported that LSD1 expression was elevated in cancerous tissue and correlated with lymph node metastasis and poorer overall survival in patients with ESCC. Compared to EC109 cells, LSD1 expression was unregulated in aggressive cancer cell lines KYSE450 and KYSE150. Knockdown of LSD1 using lentivirus delivery of LSD1-specific shRNA abrogated the migration and invasion of ESCC cells in vitro. Further, a LSD1 inhibitor, tranylcypromine, suppressed H3K4me2 demethylation and attenuated cellular motility and invasiveness in a dose-dependent manner. Taken together, these data suggested that LSD1 was a potential prognostic maker and may be a molecular target for inhibiting invasion and metastasis in ESCC.


Modern Pathology | 2014

ALDH1A1 defines invasive cancer stem-like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma

Lang Yang; Yong Ren; Xi Yu; Feng Qian; Bai-shi-jiao Bian; Hualiang Xiao; Wei-Guang Wang; Sen-lin Xu; Jing Yang; Qiang Liu; Zhe Wang; Wei Guo; Gang Xiong; Kang Yang; Cheng Qian; Xia Zhang; Peng Zhang; You-Hong Cui; Xiu-wu Bian

Invasion and metastasis are the major cause of deaths in patients with esophageal cancer. In this study, we isolated cancer stem-like cells from an esophageal squamous cell carcinoma cell line EC109 based on aldehyde dehydrogenase 1A1 (ALDH1A1), and found that ALDH1A1high cells possessed the capacities of self-renewal, differentiation and tumor initiation, indications of stem cell properties. To support their stemness, ALDH1A1high cells exhibited increased potential of invasion and metastasis as compared with ALDH1A1low cells. ALDH1A1high esophageal squamous cell carcinoma cells expressed increased levels of mRNA for vimentin, matrix metalloproteinase 2, 7 and 9 (MMP2, MMP7 and MMP9), but decreased the level of E-cadherin mRNA, suggesting that epithelial–mesenchymal transition and secretary MMPs may be attributed to the high invasive and metastatic capabilities of ALDH1A1high cells. Furthermore, we examined esophageal squamous cell carcinoma specimens from 165 patients and found that ALDH1A1high cells were associated with esophageal squamous dysplasia and the grades, differentiation and invasion depth, lymph node metastasis and UICC stage of esophageal squamous cell carcinoma, as well as poor prognosis of patients. Our results provide the strong evidence that ALDH1A1high cancer stem-like cells contribute to the invasion, metastasis and poor outcome of human esophageal squamous cell carcinoma.


Oncotarget | 2015

miRNA-regulated delivery of lincRNA-p21 suppresses β-catenin signaling and tumorigenicity of colorectal cancer stem cells

Jun Wang; Zengjie Lei; Yan Guo; Tao Wang; Zhong-yi Qin; Hualiang Xiao; Lilin Fan; Dongfeng Chen; Xiu-wu Bian; Jia Liu; Bin Wang

Cancer stem cells (CSCs) are key cellular targets for effective cancer therapy, due to their critical roles in cancer progression and chemo/radio-resistance. Emerging evidence demonstrates that long non-coding RNAs (lncRNAs) are important players in the biology of cancers. However, it remains unknown whether lncRNAs could be exploited to target CSCs. We report that large intergenic non-coding RNA p21 (lincRNA-p21) is a potent suppressor of stem-like traits of CSCs purified from both primary colorectal cancer (CRC) tissues and cell lines. A novel lincRNA-p21-expressing adenoviral vector, which was armed with miRNA responsive element (MRE) of miR-451 (Ad-lnc-p21-MRE), was generated to eliminate CRC CSCs. Integration of miR-451 MREs into the adenovirus efficiently delivered lincRNA-p21 into CSCs that contained low levels of miR-451. Moreover, lincRNA-p21 inhibited the activity of β-catenin signaling, thereby attenuating the viability, self-renewal, and glycolysis of CSCs in vitro. By limiting dilution and serial tumor formation assay, we demonstrated that Ad-lnc-p21-MRE significantly suppressed the self-renewal potential and tumorigenicity of CSCs in nude mice. Importantly, application of miR-451 MREs appeared to protect normal liver cells from off-target expression of lincRNA-p21 in both tumor-bearing and naïve mice. Taken together, these findings suggest that lncRNAs may be promising therapeutic molecules to eradicate CSCs and MREs of tumor-suppressor miRNAs, such as miR-451, may be exploited to ensure the specificity of CSC-targeting strategies.


Clinical Cancer Research | 2015

miR-663 Suppresses Oncogenic Function of CXCR4 in Glioblastoma

Yu Shi; Cong Chen; Shi-Zhu Yu; Qing Liu; Jun Rao; Hua-rong Zhang; Hualiang Xiao; Ti-Wei Fu; Hua Long; Zhi-Cheng He; Kai Zhou; Xiao-hong Yao; You-Hong Cui; Xia Zhang; Yi-Fang Ping; Xiu-wu Bian

Purpose: To identify the miRNA regulators of C–X–C motif chemokine receptor 4 (CXCR4) and the underlying mechanism as well as the therapeutic and prognostic values in human glioblastoma (GBM). Experimental Design: miRNA profile analyses and bioinformatics predictions were used to identify the mediators of CXCR4, which were confirmed by luciferase reporter assay, Western blot assay and immunohistochemistry. The effects of miR-663 on CXCR4-mediated GBM malignancy were investigated by gain-of-function experiments. Orthotopic xenografts derived from constitutive or induced miR-663–expressing GBM cells were used to determine the antitumor effects of miR-663 and CXCR4-specific antagonist AMD3100. Bivariate correlation analyses were used to examine the correlation of miR-663 and CXCR4 levels in glioma. The prognostic values of miR-663 and CXCR4 were examined in 281 cases of astrocytic glioma from our hospital and 476 cases of GBM from The Cancer Genome Atlas database using the multivariate Cox regression analysis and Kaplan–Meier analysis. Results: miR-663 negatively regulated CXCR4 expression by targeting its coding sequence in GBM and compromised the proliferative and invasive capacities of GBM cells induced by CXCR4 overexpression. Constitutive or induced miR-663 overexpression combined with CXCR4 antagonist AMD3100 suppressed orthotopic GBM growth and prolonged tumor-bearing mice survival. Clinically, miR-663 and CXCR4 were inversely correlated in GBM and composed a valuable biomarker set in predicting the outcomes of GBM patients. Conclusions: miR-663 negatively regulated CXCR4 to inhibit its oncogenic effect. Combination of miR-663 and CXCR4 can serve as a valuable prognostic biomarker set as well as molecular targets for therapeutic intervention of GBM. Clin Cancer Res; 21(17); 4004–13. ©2015 AACR.


The Journal of Molecular Diagnostics | 2015

Droplet Digital PCR for Absolute Quantification of EML4-ALK Gene Rearrangement in Lung Adenocarcinoma

Qiushi Wang; Xin Yang; Yong He; Qiang Ma; Li Lin; Ping Fu; Hualiang Xiao

Crizotinib treatment significantly prolongs progression-free survival, increases response rates, and improves the quality of life in patients with ALK-positive non-small-cell lung cancer. Droplet Digital PCR (ddPCR), a recently developed technique with high sensitivity and specificity, was used in this study to evaluate the association between the abundance of ALK rearrangements and crizotinib effectiveness. FFPE tissues were obtained from 103 consecutive patients with lung adenocarcinoma. Fluorescent in situ hybridization (FISH) and ddPCR were performed. The results revealed that 14 (13.6%) of the 103 patients were positive by dual-color, break-apart FISH. Three variants (1, 2, and 3) of the EML4-ALK gene rearrangements were detected. Thirteen of 14 ALK-positive cases identified by FISH were confirmed by ddPCR (four with variant 1, two with variant 2, and seven with variant 3). The case missed by ddPCR was identified as KIF5B-ALK gene rearrangement by PCR-based direct sequencing. Sixteen patients were detected with low copy numbers of EML4-ALK gene rearrangement, which failed to meet the positive cutoff point of FISH. Two of them responded well to crizotinib after unsuccessful chemotherapy. Our study indicates that ddPCR can be used as a molecular analytical tool to accurately measure the EML4-ALK rearrangement copy numbers in FFPE samples of lung adenocarcinoma patients.


Molecular Cancer | 2015

Neurotensin promotes the progression of malignant glioma through NTSR1 and impacts the prognosis of glioma patients

Qing Ouyang; Xueyang Gong; Hualiang Xiao; Ji Zhou; Minhui Xu; Yun Dai; Lunshan Xu; Hua Feng; Hongjuan Cui; Liang Yi

BackgroundThe poor prognosis and minimally successful treatments of malignant glioma indicate a challenge to identify new therapeutic targets which impact glioma progression. Neurotensin (NTS) and its high affinity receptor (NTSR1) overexpression induces neoplastic growth and predicts the poor prognosis in various malignancies. Whether NTS can promote the glioma progression and its prognostic significance for glioma patients remains unclear.MethodsNTS precursor (ProNTS), NTS and NTSR1 expression levels in glioma were detected by immunobloting Elisa and immunohistochemistry assay. The prognostic analysis was conducted from internet by R2 microarray platform. Glioma cell proliferation was evaluated by CCK8 and BrdU incorporation assay. Wound healing model and Matrigel transwell assay were utilized to test cellular migration and invasion. The orthotopic glioma implantations were established to analyze the role of NTS and NTSR1 in glioma progression in vivo.ResultsPositive correlations were shown between the expression levels of NTS and NTSR1 with the pathological grade of gliomas. The high expression levels of NTS and NTSR1 indicate a worse prognosis in glioma patients. The proliferation and invasiveness of glioma cells could be enhanced by NTS stimulation and impaired by the inhibition of NTSR1. NTS stimulated Erk1/2 phosphorylation in glioma cells, which could be reversed by SR48692 or NTSR1-siRNA. In vivo experiments showed that SR48692 significantly prolonged the survival length of glioma-bearing mice and inhibited glioma cell invasiveness.ConclusionNTS promotes the proliferation and invasion of glioma via the activation of NTSR1. High expression levels of NTS and NTSR1 predict a poor prognosis in glioma patients.


Journal of Biological Chemistry | 2011

Glioma-derived T Cell Immunoglobulin- and Mucin Domain-containing Molecule-4 (TIM4) Contributes to Tumor Tolerance

Lunshan Xu; Hualiang Xiao; Minhui Xu; Chun Zhou; Liang Yi; Hong Liang

Background: We investigated the role of TIM4 in the induction of regulatory T cells (Tregs) in tumors. Results: High TIM4 levels were detected in glioma tissue and contributed to tumor-specific Treg development. Conclusion: TIM4 is important in inducing Tregs in gliomas. Significance: TIM4 may be a target in glioma treatment. Tumor tolerance plays a critical role in tumor growth and escape from immune surveillance. The mechanism of tumor tolerance development is not fully understood. Regulatory T cells (Tregs) play a critical role in tumor tolerance. TIM4 (T cell immunoglobulin- and mucin domain-containing molecule-4) is involved in immune regulation. We investigated the role of TIM4 in the induction of Tregs in tumors. Surgically removed glioma tissue and peripheral blood samples were obtained from 25 glioma patients. Immune cells were isolated from the tissue and blood samples. Confocal microscopy was employed to detect macrophages phagocytosing apoptotic T cells. The generation of tumor-specific Tregs and the immune suppression function of Tregs were observed in cell culture models. High levels of TIM4 were detected in glioma-derived macrophages. Phosphatidylserine (PS) was detected in glioma-derived T cells; naïve T cells expressed low levels of PS that could be up-regulated by hypoxia. Glioma-derived macrophages phagocytosed PS-expressing T cells, gaining the tolerogenic properties, which could induce tumor-specific Tregs; the latter could suppress tumor-specific CD8+ T cells. We conclude that macrophage-derived TIM4 plays an important role in the induction of Tregs in gliomas, which may play an important role in tumor tolerance.

Collaboration


Dive into the Hualiang Xiao's collaboration.

Top Co-Authors

Avatar

Xiu-wu Bian

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Li Lin

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiang Ma

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bin Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Dongfeng Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiushi Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Zeng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

You-Hong Cui

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jun Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Liang Yi

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge