Network


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

Hotspot


Dive into the research topics where Gui-lian Xu is active.

Publication


Featured researches published by Gui-lian Xu.


PLOS ONE | 2011

Complement and the Alternative Pathway Play an Important Role in LPS/D-GalN-Induced Fulminant Hepatic Failure

Shihui Sun; Yan Guo; Guangyu Zhao; Xiaojun Zhou; Junfeng Li; Jingya Hu; Hong Yu; Yu Chen; Hongbin Song; Fei Qiao; Gui-lian Xu; Fei Yang; Yuzhang Wu; Stephen Tomlinson; Zhongping Duan; Yusen Zhou

Fulminant hepatic failure (FHF) is a clinically severe type of liver injury with an extremely high mortality rate. Although the pathological mechanisms of FHF are not well understood, evidence suggests that the complement system is involved in the pathogenesis of a variety of liver disorders. In the present study, to investigate the role of complement in FHF, we examined groups of mice following intraperitoneal injection of LPS/D-GalN: wild-type C57BL/6 mice, wild-type mice treated with a C3aR antagonist, C5aR monoclonal antibody (C5aRmAb) or CR2-Factor H (CR2-fH, an inhibitor of the alternative pathway), and C3 deficient mice (C3−/− mice). The animals were euthanized and samples analyzed at specific times after LPS/D-GalN injection. The results show that intraperitoneal administration of LPS/D-GalN activated the complement pathway, as evidenced by the hepatic deposition of C3 and C5b-9 and elevated serum levels of the complement activation product C3a, the level of which was associated with the severity of the liver damage. C3a receptor (C3aR) and C5a receptor (C5aR) expression was also upregulated. Compared with wild-type mice, C3−/− mice survived significantly longer and displayed reduced liver inflammation and attenuated pathological damage following LPS/D-GalN injection. Similar levels of protection were seen in mice treated with C3aR antagonist,C5aRmAb or CR2-fH. These data indicate an important role for the C3a and C5a generated by the alternative pathway in LPS/D-GalN-induced FHF. The data further suggest that complement inhibition may be an effective strategy for the adjunctive treatment of fulminant hepatic failure.


Hepatology | 2014

C5a/C5aR pathway is essential for the pathogenesis of murine viral fulminant hepatitis by way of potentiating Fgl2/fibroleukin expression.

Gui-lian Xu; Jian Chen; Fei Yang; Gui-qing Li; Li-xin Zheng; Yuzhang Wu

Viral fulminant hepatitis (FH) remains a serious clinical problem with very high mortality. Lacking understanding of FH pathogenesis has in essence hindered efficient clinical treatment. Inferring from a correlation observed between the genetic differences in the complement component 5 (C5) and the susceptibility of mouse strains to murine hepatitis virus strain‐3 (MHV‐3) infections, we propose that excessive complement activation plays a critical role in the development of FH. We show that MHV‐3 infection causes massive complement activation, along with a rapid increase in serum C5a levels and quick development of FH in susceptible strains. Mice deficient in the C5a receptor (C5aR) or the susceptible strains treated with C5aR antagonists (C5aRa) exhibit significant attenuation of the disease, accompanied by a remarkable reduction of hepatic fibrinogen‐like protein 2 (Fgl2), a hallmark protein that causes necrosis of infected livers. In accordance, biopsy of FH patients shows a dramatic increase of Fgl2 expression, which correlates with C5aR up‐regulation in the liver. In vitro C5a administration accelerates MHV‐3‐induced Fgl2 secretion by macrophages. Furthermore, inhibiting ERK1/2 and p38 efficiently blocks C5a‐mediated Fgl2 production during viral infections. Conclusion: These data provide evidence that mouse susceptibility to MHV‐3‐induced FH may rely on C5a/C5aR interactions, for which ERK1/2 and p38 pathways participate in up‐regulating Fgl2 expression. Inhibition of C5a/C5aR interactions is expected to be beneficial in the clinical treatment of FH patients. (Hepatology 2014;60:114–124)


Molecular Immunology | 2009

Mutual activation of CD4+ T cells and monocytes mediated by NKG2D-MIC interaction requires IFN-gamma production in systemic lupus erythematosus.

Di Yang; Huiming Wang; Bing Ni; Yani He; Jingyi Li; Yuyu Tang; Xiaolan Fu; Qinghong Wang; Gui-lian Xu; Kailong Li; Zhao Yang; Yuzhang Wu

The activating receptor NKG2D is mainly expressed by human CD8(+) T cells and NK cells but normally absent on CD4(+) T cells. However, a subset of autoreactive NKG2D(+)CD4(+) T cells has been found to exist in some autoimmune disease such as rheumatoid arthritis (RA) and to participate in the imbalance of immune response and inflammation. Up to date this observation has been extended to some autoimmune diseases such as RA and Crohns disease and the mechanism underlying the presence of this type of NKG2D(+)CD4(+) T cells has not been delineated yet. In this study, we found that a substantial proportion of CD4(+) T cells expressed NKG2D in the PBMC of SLE patients. We also found that monocytes in SLE aberrantly expressed the NKG2D ligand of MHC class I chain-related (MIC) molecules and membrane-bound IL-15 (mIL-15) at the cell surface. When cultured with the sera from SLE patients, the monocytes from healthy volunteers could be induced to express MIC and mIL-15. However, this induced expression of MIC and mIL-15 could be blocked with anti-IFN-gamma receptor (anti-IFN-gammaR) antibody. We further demonstrated that NKG2D could be induced on normal CD4(+) T cells either cocultured with monocytes from patients with SLE, or monocytes from healthy volunteers but pretreated with IFN-gamma. Moreover, Th1 cytokines were found to be produced by NKG2D(+)CD4(+) T cells in the coculture system. By transwell assay, we found that both NKG2D expression and Th1 cytokines production depended on the cell-cell contact. These results indicate that the elevated sera IFN-gamma may be responsible for MIC and mIL-15 induction on monocytes in SLE; mIL-15 on monocytes contribute to NKG2D receptor induction on a subset of CD4(+) T cells. Moreover, CD14(+) monocytes promote NKG2D(+)CD4(+) T cells activation through the NKG2D-MIC engagement in the pathogenesis of SLE.


PLOS ONE | 2015

STAT1-Mediated Down-Regulation of Bcl-2 Expression Is Involved in IFN-γ/TNF-α–Induced Apoptosis in NIT-1 Cells

Zhao-hui Cao; Quan-you Zheng; Gui-qing Li; Xiao-bo Hu; Shao-long Feng; Gui-lian Xu; Keqin Zhang

Tumor necrosis factor (TNF)-α and interferon (IFN)-γ are the major pro-inflammatory cytokines involved in beta-cell destruction. The fate of islet beta-cells in the cytokine-induced intrinsic mitochondrial apoptotic pathway is determined by the interaction between members of the Bcl-2 family. However, the mechanism through which beta-cell apoptosis is regulated remains unclear. In this study, we treated the murine beta-cell line NIT-1 with TNF-α and IFN-γ and then investigated the regulation of signal transducer and activator of transcription-1 (STAT-1) and expression of the members of the Bcl-2 family in this apoptotic pathway. Results showed that TNF-α and IFN-γ synergistically reduced NIT-1 cell viability. In addition, the decrease in cell growth was due to apoptosis as shown by apoptotic body formation, detected by confocal laser microscope, and a significant increase in Annexin-Vup+ cell percentage, detected by flow cytometry. Combination treatment with TNF-α and IFN-γ caused a remarkable increase in the release of cytochrome c, and in the activation of caspase-9 and caspase-3, as well as, an obvious enhancement in STAT-1 phosphorylation; the treatment, however, resulted in the down-regulation in Bcl-2 expression. The enhancement in STAT-1 activity and a down-regulation in Bcl-2 expression was also observed in MIN6 cells, another murine beta-cell derived line, after cells exposure to the combination of TNF-α and IFN-γ treatment. Knockdown of STAT-1 gene expression by siRNA or inhibition of STAT-1 activation with fludarabine reversed Bcl-2 down-expression and led to a significant decrease in apoptosis in TNF-α- and IFN-γ-treated NIT-1 cells. Taken together, our results suggest that STAT1-mediated down-regulation of Bcl-2 is involved in NIT-1 cell apoptosis induced by combination treatment with TNF-α and IFN-γ.


Journal of Immunology | 2013

An essential role for C5aR signaling in the optimal induction of a malaria-specific CD4+ T cell response by a whole-killed blood-stage vaccine.

Taiping Liu; Gui-lian Xu; Bo Guo; Yong Fu; Yuan Qiu; Yan Ding; Hong Zheng; Xiaolan Fu; Yuzhang Wu; Wenyue Xu

The protective immunity induced by the whole-killed parasite vaccine against malarial blood-stage infection is dependent on the CD4+ T cell response. However, the mechanism underlying this robust CD4+ T cell response elicited by the whole-killed parasite vaccine is still largely unknown. In this study, we observe that immunization with Plasmodium yoelii–parasitized RBC lysate activates complement C5 and generates C5a. However, the protective efficacy against P. yoelii 17XL challenge is considerably reduced, and the malaria-specific CD4+ T cell activation and memory T cell differentiation are largely suppressed in the C5aR-deficient (C5aR−/−) mice. An adoptive transfer assay demonstrates that the reduced protection of C5aR−/− mice is closely associated with the severely impaired CD4+ T cell response. This is further confirmed by the fact that administration of C5aR antagonist significantly reduces the protective efficacy of the immunized B cell–deficient mice. Further study indicates that the defective CD4+ T cell response in C5aR−/− mice is unlikely involved in the expansion of CD4+CD25+Foxp3+ T cells, but strongly linked to a defect in dendritic cell (DC) maturation and the ability to allostimulate CD4+ T cells. These results demonstrate that C5aR signaling is essential for the optimal induction of the malaria-specific CD4+ T cell response by the whole-killed parasite vaccine through modulation of DCs function, which provides us with new clues to design an effective blood-stage subunit vaccine and helps us to understand the mechanism by which the T cell response is regulated by the complement system.


Vaccine | 2010

Induction of protective and therapeutic antitumor immunity by a DNA vaccine with C3d as a molecular adjuvant

Gui-lian Xu; Ke-qin Zhang; Bo Guo; Ting-ting Zhao; Fei Yang; Man Jiang; Qinghong Wang; Yu-hang Shang; Yuzhang Wu

Although the critical role of complement component C3d as a molecular adjuvant in preventing virus infection is well established, its role in cancer therapies is unclear. In this study, we have engineered a DNA vaccine that expresses extracellular region of murine VEGFR-2 (FLK1(265-2493)) and 3 copies of C3d (C3d3), a component of complement as a molecular adjuvant, designed to increase antitumor immunity. VEGFR-2 has a more restricted expression on endothelial cells and is upregulated once these cells proliferate during angiogenesis in the tumor vasculature. Immunization of mice with vector encoding FLK1(265-2493) alone generated only background levels of anti-VEGFR-2 antibodies and slight inhibitory effect on tumor growth. However, the addition of C3d3 to the vaccine construct significantly augmented the anti-VEGFR-2 humoral immune response and inhibited the tumor growth. The antitumor activity induced by vaccination with vector encoding FLK1(265-2493)-C3d3 fusion protein was also demonstrated via growth inhibition of established tumors following passive transfer of immune serum from vaccinated mice. Our results suggest that vaccination with vector encoding FLK1(265-2493) with C3d3 as a molecular adjuvant induces adaptive humoral activity, which is directed against the murine VEGFR-2 and can significantly inhibit tumor growth, and that administration of C3d as a molecular adjuvant to increase antibodies levels to VEGFR-2 may provide an alternative treatment modality for cancer therapies.


Clinical Immunology | 2008

Mapping of binding epitopes of a human decay-accelerating factor monoclonal antibody capable of enhancing rituximab-mediated complement-dependent cytotoxicity

Bo Guo; Zhengwei Ma; Hua Li; Gui-lian Xu; Ping Zheng; Bo Zhu; Yuzhang Wu; Qiang Zou

Complement-dependent cytotoxicity (CDC) is thought to be one of the most important mechanisms of action of therapeutic monoclonal antibodies (mAbs). The decay-accelerating factor (DAF) overexpressed in certain tumors limits the CDC effect of the therapeutic anticancer antibodies. The use of DAF blocking antibodies targeted specifically at cancer cells in combination with immunotherapeutic mAbs of cancer may improve the therapeutic effect in cancer patients. In this study, the lysis of Raji cells mediated by CDC was determined after blocking DAF function by anti-DAF polyclonal antibody and 3 mAbs (DG3, DG9, DA11) prepared in our laboratory, respectively, in the presence of the anti-CD20 chimeric mAb rituximab. The binding domains of the three anti-DAF mAbs were identified using yeast surface display technique, and the mimic epitopes of mAb DG3 were screened from a random phage-display nonapeptide library. The results showed that blocking DAF function by anti-DAF polyclonal antibody enhanced complement-mediated killing of Raji cells. Among the 3 mAbs against DAF, only DG3 was found to be able to remarkably enhance the CDC effect of the therapeutic mAb rituximab. DG3 bound to the third short consensus repeat (SCR) of DAF. Binding of DG3 to immobilized DAF was inhibited by mimic epitope peptides screened from the peptide library. Our results suggest that a higher level of DAF expressed by certain tumor cells is significant to abolish the CDC effect of therapeutic anticancer antibodies, and mAbs binding to SCR3 can enhance the complement-mediated killing of Raji cells. It is of significance to identify the DAF epitopes required in inhibiting CDC not only for better understanding of the relationship between the structure and function of DAF, but also for designing and developing anti-DAF mAbs capable of enhancing CDC.


Journal of Cellular and Molecular Medicine | 2016

LIGHT/IFN‐γ triggers β cells apoptosis via NF‐κB/Bcl2‐dependent mitochondrial pathway

Quan-you Zheng; Zhao-hui Cao; Xiao-bo Hu; Gui-qing Li; Shi-Fang Dong; Gui-lian Xu; Keqin Zhang

LIGHT recruits and activates naive T cells in the islets at the onset of diabetes. IFN‐γ secreted by activated T lymphocytes is involved in beta cell apoptosis. However, whether LIGHT sensitizes IFNγ‐induced beta cells destruction remains unclear. In this study, we used the murine beta cell line MIN6 and primary islet cells as models for investigating the underlying cellular mechanisms involved in LIGHT/IFNγ – induced pancreatic beta cell destruction. LIGHT and IFN‐γ synergistically reduced MIN6 and primary islet cells viability; decreased cell viability was due to apoptosis, as demonstrated by a significant increase in Annexin V+ cell percentage, detected by flow cytometry. In addition to marked increases in cytochrome c release and NF‐κB activation, the combination of LIGHT and IFN‐γ caused an obvious decrease in expression of the anti‐apoptotic proteins Bcl‐2 and Bcl‐xL, but an increase in expression of the pro‐apoptotic proteins Bak and Bax in MIN6 cells. Accordingly, LIGHT deficiency led to a decrease in NF‐κB activation and Bak expression, and peri‐insulitis in non‐obese diabetes mice. Inhibition of NF‐κB activation with the specific NF‐κB inhibitor, PDTC (pyrrolidine dithiocarbamate), reversed Bcl‐xL down‐regulation and Bax up‐regulation, and led to a significant increase in LIGHT‐ and IFN‐γ‐treated cell viability. Moreover, cleaved caspase‐9, ‐3, and PARP (poly (ADP‐ribose) polymerase) were observed after LIGHT and IFN‐γ treatment. Pretreatment with caspase inhibitors remarkably attenuated LIGHT‐ and IFNγ‐induced cell apoptosis. Taken together, our results indicate that LIGHT signalling pathway combined with IFN‐γ induces beta cells apoptosis via an NF‐κB/Bcl2‐dependent mitochondrial pathway.


Scientific Reports | 2017

Increased expression of Fibrinogen-Like Protein 2 is associated with poor prognosis in patients with clear cell renal cell carcinoma

Ming Tang; Xu Cao; Peng Li; Kun Zhang; You Li; Quan-you Zheng; Gui-qing Li; Jian Chen; Gui-lian Xu; Keqin Zhang

Fibrinogen-like protein 2 (FGL2) is highly expressed in various tumour tissues and plays a vital role in tumour initiation and progression. This study evaluated the clinical significance of FGL2 in patients with clear cell renal cell carcinoma (ccRCC). FGL2 expression in fresh and 170 archived paraffin-embedded ccRCC tissues was measured by quantitative RT-PCR, western blotting, and immunohistochemitry. FGL2 expression was significantly upregulated in ccRCC. Statistical analyses by using Kaplan–Meier method showed that high FGL2 expression was associated with poor overall survival (OS) and recurrence-free survival (RFS) of patients with ccRCC. Multivariate analyses indicated that FGL2 was as an independent prognostic factor of survivaland that tumoural FGL2 levels could significantly predict the prognosis of patients with early-stage ccRCC. Nomogram systems, which integrated FGL2 expression and other clinical parameters, were established and were found to be better than TNM staging in predicting the OS and RFS of patients with ccRCC. FGL2 silencing led to a significant reduction in cells viability and increase in cells apoptosis, accompanied with a reduced ERK1/2 and p38 MAPK activation, in ccRCC cells. Thus, our results suggest that high FGL2 expression is a novel, independent, and an adverse prognostic factor of clinical outcomes in patients with ccRCC.


International Immunopharmacology | 2017

C5a/C5aR pathway accelerates renal ischemia-reperfusion injury by downregulating PGRN expression

Kun Zhang; Gui-qing Li; Qian-hui He; You Li; Ming Tang; Quan-you Zheng; Gui-lian Xu; Keqin Zhang

Abstract Recent reports indicate that the complement C5a/C5aR pathway and progranulin (PGRN) deficiency both contribute to ischemia‐reperfusion (IR)‐induced acute kidney injury. However, the underlying relationship between the C5a/C5aR signaling pathway and PGRN expression during acute kidney injury is poorly understood. In this study, we showed that C5aR expression was significantly upregulated after renal IR, and that C5aR deficiency led to a marked increase in PGRN expression and a significant reduction in tubular damage and production of inflammatory cytokines. In accordance with these results, recombinant C5a caused downregulation of PGRN protein and mRNA levels in renal tubular epithelial cells (HK‐2 cells), which could be negated by disruption of C5a/C5aR signaling by the C5aR antagonist, as confirmed by immunofluorescence, western blotting, and quantitative real‐time PCR. Moreover, C5aR deficiency resulted in attenuated NF‐&kgr;B expression 24 h after IR, and recombinant C5a potentiated TNF&agr;‐induced NF‐&kgr;B activation in HK‐2 cells. Inhibition of NF‐&kgr;B activation reversed C5a‐induced downregulation of PGRN expression. Our results show for the first time that the complement C5a/C5aR pathway aggravates IR‐induced acute kidney injury by suppressing PGRN expression and confirm that suppression of PGRN expression is associated with increased NF‐&kgr;B activation induced by C5a. HighlightsComplement C5a/C5aR pathway aggravates IR‐induced acute kidney injury.C5a/C5aR pathway down‐regulates PGRN expression after renal IR.C5a/C5aR suppresses PGRN expression via the NF‐&kgr;B‐dependent manner.

Collaboration


Dive into the Gui-lian Xu's collaboration.

Top Co-Authors

Avatar

Yuzhang Wu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Gui-qing Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bo Guo

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Keqin Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Quan-you Zheng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ming Tang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Kun Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaolan Fu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

You Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Fei Yang

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge