Network


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

Hotspot


Dive into the research topics where Zhuoya Li is active.

Publication


Featured researches published by Zhuoya Li.


Endocrinology | 2008

Blockade of Tumor Necrosis Factor (TNF) Receptor Type 1-Mediated TNF-α Signaling Protected Wistar Rats from Diet-Induced Obesity and Insulin Resistance

Huifang Liang; Bingjiao Yin; Hailong Zhang; Shu Zhang; Qingling Zeng; Jing Wang; Xiaodan Jiang; Li Yuan; Cong Yi Wang; Zhuoya Li

TNF-alpha plays an important role in the pathogenesis of obesity and insulin resistance in which the effect of TNF-alpha signaling via TNF receptor type 1 (TNFR1) largely remains controversial. To delineate the role of TNFR1-mediated TNF-alpha signaling in the pathogenesis of this disorder, a TNFR1 blocking peptide-Fc fusion protein (TNFR1BP-Fc) was used for the present study. Wistar rats were fed a high-fat/high-sucrose (HFS) diet for 16 wk until obesity and insulin resistance developed. In comparison with increased body weight and fat weight, enlarged adipocytes, and hypertriglyceridemia in the obese state, the subsequent 4-wk treatment with TNFR1BP-Fc resulted in significant weight loss characterized by decreased fat pad weight and adipocyte size and reduced plasma triglycerides. Furthermore, obesity-induced insulin resistance, including hyperinsulinemia, elevated C-peptide, higher degree of hyperglycemia after glucose challenge, and less hypoglycemic response to insulin, was markedly improved, and the compensatory hyperplasia and hypertrophy of pancreatic islets were reduced. Interestingly, treatment with TNFR1BP-Fc markedly suppressed systemic TNF-alpha release and its local expression in pancreatic islets and muscle and adipose tissues. In addition, blockage of TNFR1-mediated TNF-alpha signaling in obese rats significantly enhanced tyrosine phosphorylation of insulin receptor substrate 1 (IRS-1) in the muscle and fat tissues. Our results strongly suggest a pivotal role for TNFR1-mediated TNF-alpha signaling in the pathogenesis of obesity and insulin resistance. Thus, TNFR1BP-Fc may be a good candidate for the treatment of this disease.


Journal of Immunology | 2014

Transmembrane TNF-α Promotes Suppressive Activities of Myeloid-Derived Suppressor Cells via TNFR2

Xin Hu; Baihua Li; Xiaoyan Li; Xianxian Zhao; Lin Wan; Guohong Lin; Min Yu; Jing Wang; Xiaodan Jiang; Wei Feng; Zhihai Qin; Bingjiao Yin; Zhuoya Li

It has been reported that TNFR2 is involved in regulatory T cell induction and myeloid-derived suppressor cell (MDSC) accumulation, two kinds of immunosuppressive cells contributing to tumor immune evasion. Because transmembrane TNF-α (tmTNF-α) is the primary ligand for TNFR2, we hypothesized that tmTNF-α is mainly responsible for the activation of MDSCs. Indeed, we found that tmTNF-α, rather than secretory TNF-α (sTNF-α), activated MDSCs with enhanced suppressive activities, including upregulating arginase-1 and inducible NO synthase transcription, promoting secretion of NO, reactive oxygen species, IL-10, and TGF-β, and enhancing inhibition of lymphocyte proliferation. This effect of tmTNF-α was mediated by TNFR2, as TNFR2 deficiency significantly impaired tmTNF-α–induced release of IL-10 and NO and inhibition of T cell proliferation by MDSC supernatant. Furthermore, tmTNF-α caused p38 phosphorylation and NF-κB activation, whereas inhibition of NF-κB or p38 with an inhibitor pyrrolidine dithiocarbamate or SB203580 abrogated tmTNF-α–mediated increased suppression of lymphocyte proliferation by MDSCs. Consistently, our in vivo study showed that ectopic expression of uncleavable tmTNF-α mutant by 4T1 cells significantly promoted tumor progression and angiogenesis, accompanied with more accumulation of MDSCs and regulatory T cells in the tumor site, increased production of NO, IL-10, and TGF-β, as well as poor lymphocyte infiltration. In contrast, enforced expression of sTNF-α mutant by 4T1 cells that only released sTNF-α without expression of surface tmTNF-α markedly reduced MDSC accumulation and induced more lymphocyte infiltration instead, showing obvious tumor regression. Our data suggest that tmTNF-α acts as a potent activator of MDSCs via TNFR2 and reveals another novel immunosuppressive effect of this membrane molecule that promotes tumor immune escape.


Cell Death & Differentiation | 2007

Downregulation of Bim by brain-derived neurotrophic factor activation of TrkB protects neuroblastoma cells from paclitaxel but not etoposide or cisplatin-induced cell death.

Zhuoya Li; Jianke Zhang; Zheng-gang Liu; Woo Cw; Thiele Cj

Chemoresistance and increased expression of TrkB and brain-derived neurotrophic factor (BDNF) are biomarkers of poor prognosis in tumors from patients with neuroblastoma (NB). Previously, we found BDNF activation of TrkB through PI3K/Akt protects NB from etoposide/cisplatin-induced cell death. In this study, the role of Bim, a proapoptotic protein, was investigated. Bim was involved in paclitaxel but not etoposide or cisplatin-induced cell death in NB cells. Pharmacological and genetic studies showed that BDNF-induced decreases in Bim were regulated by MAPK and not PI3K/Akt pathway. Both MAPK and PI3K pathways were involved in BDNF protection of NB cells from paclitaxel-induced cell death, while PI3K predominantly mediated BDNF protection of NB cells from etoposide or cisplatin-induced cell death. These data indicate that different chemotherapeutic drugs induce distinct death pathways and growth factors utilize different signal transduction pathways to modulate the effects of chemotherapy on cells.


Journal of Leukocyte Biology | 2008

Transmembrane TNF-α mediates “forward” and “reverse” signaling, inducing cell death or survival via the NF-κB pathway in Raji Burkitt lymphoma cells

Hailong Zhang; Dan Yan; Xu Shi; Huifang Liang; Yan Pang; Nalin Qin; Hui Chen; Jing Wang; Bingjiao Yin; Xiaodan Jiang; Wei Feng; Wenjie Zhang; Muxiang Zhou; Zhuoya Li

Interestingly, some lymphoma cells, expressing high levels of transmembrane (tm)TNF‐α, are resistant to secretory (s)TNF‐α‐induced necrosis but sensitive to tmTNF‐α‐mediated apoptosis. As tmTNF‐α mediates “forward” as well as “reverse” signaling, we hypothesize that a balanced signaling between forward and reverse directions may play a critical role in determining the fate of cells bearing tmTNF‐α. Using Raji cells as a model, we first added exogenous tmTNF‐α on fixed, transfected NIH3T3 cells onto Raji cells to examine tmTNF‐α forward signaling and its effects, showing that constitutive NF‐κB activity and cellular inhibitor‐of‐apoptosis protein 1 transcription were down‐regulated, paralleled with Raji cell death. As Raji cells express tmTNF‐α, an inhibition of their tmTNF‐α expression by antisense oligonucleotide caused down‐regulation of NF‐κB activity. Conversely, increasing tmTNF‐α expression by suppressing expression of TNF‐α‐converting enzyme that cleaves tmTNF‐α led to an enhanced activation of NF‐κB, indicating that tmTNF‐α, but not sTNF‐α, contributes to constitutive NF‐κB activation. We next transfected Raji cells with a mutant tmTNF‐α lacking the intracellular domain to competitively suppress reverse signaling via tmTNF‐α; as expected, constitutive NF‐κB activity was decreased. In contrast, treating Raji cells with sTNFR2 to stimulate reverse signaling via tmTNF‐α ehanced NF‐κB activation. We conclude that tmTNF‐α, when highly expressed on tumor cells and acting as a receptor, promotes NF‐κB activation through reverse signaling, which is helpful to maintain tumor cell survival. On the contrary, tmTNF‐α, when acting as a ligand, inhibits NF‐κB activity through forward signaling, which is inclined to induce tumor cell death.


Cancer Research | 2013

Targeting Transmembrane TNF-α Suppresses Breast Cancer Growth

Mingxia Yu; Xiaoxi Zhou; Lin Niu; Guohong Lin; Jin Huang; Wenjing Zhou; Hui Gan; Jing Wang; Xiaodan Jiang; Bingjiao Yin; Zhuoya Li

TNF antagonists may offer therapeutic potential in solid tumors, but patients who have high serum levels of TNF-α fail to respond to infliximab, suggesting consumption of the circulating antibody and loss of transmembrane TNF-α (tmTNF-α) on tumors by ectodomain shedding. Addressing this possibility, we developed a monoclonal antibody (mAb) that binds both full-length tmTNF-α and its N-terminal truncated fragment on the membrane after tmTNF-α processing but does not cross-react with soluble TNF-α. We documented high levels of tmTNF-α expression in primary breast cancers, lower levels in atypical hyperplasia or hyperplasia, but undetectable levels in normal breast tissue, consistent with the notion that tmTNF-α is a potential therapeutic target. Evaluations in vitro and in vivo further supported this assertion. tmTNF-α mAb triggered antibody-dependent cell-mediated cytotoxicity against tmTNF-α-expressing cells but not to tmTNF-α-negative cells. In tumor-bearing mice, tmTNF-α mAb delayed tumor growth, eliciting complete tumor regressions in some mice. Moreover, tmTNF-α mAb inhibited metastasis and expression of CD44v6, a prometastatic molecule. However, the antibody did not activate tmTNF-α-mediated reverse signaling, which facilitates tumor survival and resistance to apoptosis, but instead inhibited NF-κB activation and Bcl-2 expression by decreasing tmTNF-α-positive cells. Overall, our results established that tmTNF-α mAb exerts effective antitumor activities and offers a promising candidate to treat tmTNF-α-positive tumors, particularly in patients that are nonresponders to TNF antagonists.


European Journal of Cell Biology | 2009

Influence of reverse signaling via membrane TNF-α on cytotoxicity of NK92 cells

Mingxia Yu; Wenfang Shi; Jian Zhang; Lin Niu; Qizheng Chen; Dan Yan; Tao Liu; Wang Jing; Xiaodan Jiang; Feng Wei; Bingjiao Yin; Wenjie Zhang; Qingfen Li; Zhuoya Li

Membrane tumor necrosis factor-alpha (mTNF-alpha) serves as a receptor transducing signals into mTNF-alpha-bearing cells. Among human peripheral blood mononuclear cells, natural killer (NK) cells have been reported to be the only cell type constitutively expressing mTNF-alpha, which is involved in the cytotoxicity of resting NK cells. Using an IL-2-dependent human NK cell line, NK92, which constitutively expresses mTNF-alpha, we examined the effect of reverse signaling via mTNF-alpha on cellular activities. When the cells were prestimulated with soluble TNFR1 (sTNFR1) which activated mTNF-alpha-mediated reverse signaling, the cytotoxicity of NK92 cells was significantly increased. Further investigation demonstrated that prestimulation with sTNFR1 augmented exocytosis and mRNA transcription of two cytotoxic molecules, perforin and granzyme B, which could serve as underlying molecular mechanisms by which mTNF-alpha-mediated reverse signaling promoted cytotoxicity of NK cells toward K562 cells. On the other hand, pretreatment of NK92 with sTNFR1 boosted the expression of FasL and TNF-alpha, including both the secretory and membrane forms. These molecules also contribute to the NK-mediated cytotoxicity, although K562 cells are Fas-negative and sTNF-alpha-resistant. Interestingly, the mTNF-alpha reverse signaling was found to act synergistically with IL-2 on NK-mediated cytotoxicity. This synergy markedly promoted the production of secretory as well as membrane cytotoxic molecules which may be responsible for the enhanced NK92-mediated cytotoxicity. Our observations suggest that, via reverse signaling, constitutively expressed mTNF-alpha may sensitize NK cells to activating stimuli, such as IL-2, resulting in increased NK-mediated cytotoxicity through promoting the production of multiple cytotoxic effector molecules.


Breast Cancer Research and Treatment | 2009

Expression of TNF-α leader sequence renders MCF-7 tumor cells resistant to the cytotoxicity of soluble TNF-α

Dan Yan; Nalin Qin; Hailong Zhang; Tao Liu; Mingxia Yu; Xiaodan Jiang; Wei Feng; Jing Wang; Bingjiao Yin; Tao Zhang; Muxiang Zhou; Zhuoya Li

Transmembrane TNF-α (tmTNF-α) contains a leader sequence (LS) that can be phosphorylated and cleaved at its cytoplasmic portion, inducing IL-12 production. We observed that the breast cancer cell line MDA-MB-231 expressing transmembrane TNF-α (tmTNF-α) at high level was resistant to soluble TNF-α (sTNF-α)-induced cytotoxicity, accompanied by constitutive NF-κB activation. In contrast, MCF-7 cells expressing tmTNF-α at very low level were sensitive to sTNF-α-induced cell death and had no detectable NF-κB activation. Consistently, siRNA-mediated tmTNF-α knockdown blocked NF-κB activation and rendered MDA-MB-231 sensitive. To test our hypothesis that TNF-LS may play an important role in determining the sensitivity of tumor cells to sTNF-α, we stably transfected MCF-7 cells with TNF-LS. We found that transfection of TNF-LS or wild-type TNF-α containing LS constitutively activated NF-κB and conferred the cytotoxic resistance of MCF-7 cells, while transfection of a mutant tmTNF-α lacking the cytoplasmic segment of LS neither activated NF-κB nor affected the sensitivity. However, NF-κB inhibitor PDTC suppressed NF-κB activation and reconstituted sensitivity of TNF-LS/MCF-7 cells. To check whether TNF-LS is required to be cleaved or internalized for NF-κB activation to occur, we used signal peptide peptidase inhibitor (Z-LL)2-ketone and receptor internalization inhibitor MDC to treat cells. Interestingly, both inhibitors increased TNF-LS expression on the cell surface and enhanced NF-κB activation. These results indicate that membrane-anchored TNF-LS contributes to constitutive activation of NF-κB and resistance to sTNF-α-induced cell death. Therefore, TNF-LS appears to be responsible for tmTNF-α-induced resistance in the breast cancer cells.


European Journal of Pharmacology | 2011

Blocking TNF-α by combination of TNF-α- and TNFR-binding cyclic peptide ameliorates the severity of TNBS-induced colitis in rats.

Bingjiao Yin; Xin Hu; Jing Wang; Huifang Liang; Xiaoyan Li; Nin Niu; Baihua Li; Xiaodan Jiang; Zhuoya Li

Tumor necrosis factor alpha (TNF-α) has been implicated in the pathogenesis of Crohns disease. TNF antagonists are effectively used to treat these patients, although the efficiency of different antagonists varies. In the present study we combined TNF-α binding cyclic peptide (TBCP) and TNFR1 binding cyclic peptide (TRBCP) to treat TNBS-induced colitis in rats for one week. The symptoms of colitis including bloody diarrhea, rectal prolapse, and a profound and sustained weight loss were significantly ameliorated and the colon inflammatory damage, both macroscopic and histological scores, MPO activity, and NO production were markedly decreased in rats by neutralization of TNF-α and blocking TNFR1, as compared with those in rats treated with irrelevant peptide or normal saline (P<0.05). The transcripts of IL-1β and IL-8, and the protein expression of TNF-α in rats treated with both TBCP and TRBCP were also down-regulated (P<0.05), while these proinflammatory cytokines remained unchanged in rats treated with irrelevant peptide or normal saline. These findings suggest that the combination of TNF-α- and TNFR1-binding peptide effectively improves the symptoms of TNBS-induced colitis and alleviates colonic pathological damages in rats. This combination may be a potent candidate for clinical treatment of the inflammatory bowel disease.


Molecular Immunology | 2009

Lipid rafts uncouple surface expression of transmembrane TNF-α from its cytotoxicity associated with ICAM-1 clustering in Raji cells

Shu Zhang; Tao Liu; Huifang Liang; Hailong Zhang; Dan Yan; Nidan Wang; Xiaodan Jiang; Wei Feng; Jing Wang; Pinlan Li; Zhuoya Li

Since transmembrane tumor necrosis factor-alpha (tmTNF-alpha) has been reported to have a palmitoylated site at Cys(-47), and therefore its functions may be linked to lipid raft membrane microdomains. The present study tested a hypothesis that lipid rafts may serve as a signaling platform to mediate the bioactivity of tmTNF-alpha. We found that destruction of lipid rafts with methyl-beta-cyclodextrin (MCD) in Raji cells almost completely blocked the cytotoxicity of tmTNF-alpha, as did an anti-TNF-alpha antibody. Although a proportion of tmTNF-alpha was colocated with lipid rafts, either the replacement of Cys at -47 by Ala, destructing its possible lipid rafts-attaching site or the displacement of its cytoplasmic domain by the C-terminal sequence (131-157) of caveolin-1, making all tmTNF-alpha target to lipid rafts, had no effect on tmTNF-alpha cytotoxicity. The data suggest that the cytotoxicity of tmTNF-alpha is not associated with its lipid rafts location. Unparallel to decreased cytotoxicity, moreover, MCD significantly increased tmTNF-alpha expression on the cell surface, and these increased tmTNF-alpha molecules were capable of binding to sTNFR1. To further explore the mechanism of lipid rafts-mediated cytotoxicity of tmTNF-alpha, we demonstrated that MCD led to a marked decrease in adhesion of Raji cells to T24 cells, which was due to dissociation of adhesion molecule ICAM-1 from lipid rafts. These results indicate that lipid rafts importantly participate in the cytotoxicity of tmTNF-alpha through ICAM-1 clustering and consequent enhancement of the cell-cell contact. The data suggest that lipid rafts are essential for the killing of tmTNF-alpha through the cell-cell contact mediated by ICAM-1. However, lipid rafts may limit exposure of tmTNF-alpha to the cell surface.


Cell and Tissue Research | 2016

Kupffer-cell-expressed transmembrane TNF-α is a major contributor to lipopolysaccharide and D-galactosamine-induced liver injury

Peng Yang; Wenjing Zhou; Meng Zhang; Yaping Jiang; Rui Jiang; Hongping Ba; Cheng Li; Jing Wang; Bingjiao Yin; Feili Gong; Zhuoya Li

Tumor necrosis factor (TNF)-α exists in two bioactive forms, a 26-kDa transmembrane form (tmTNF-α) and a 17-kDa soluble form (sTNF-α). sTNF-α has been recognized as a key regulator of hepatitis; however, serum sTNF-α disappears in mice during the development of severe liver injury, and high levels of serum sTNF-α do not necessarily result in liver damage. Interestingly, in a mouse model of acute hepatitis, we have found that tmTNF-α expression on Kupffer cells (KCs) significantly increases when mice develop severe liver injury caused by lipopolysaccharide (LPS)/D-galactosamine (D-gal), and the level of tmTNF-α expression is positively related to the activity of serum transaminases. Therefore, we hypothesized that KC-expressed tmTNF-α constitutes a pathomechanism in hepatitis and have explored the role of tmTNF-α in this disease model. Here, we have compared the impact of KCstmTNFlow and KCstmTNFhigh on acute hepatitis in vivo and ex vivo and have further demonstrated that KCstmTNFhigh, rather than KCstmTNFlow, not only exhibit an imbalance in secretion of pro- and anti-inflammatory cytokines, favoring inflammatory response and exacerbating liver injury, but also induce hepatocellular apoptosis via tmTNF-α and the expression of another pro-apoptotic factor, Fas ligand. Our data suggest that KCtmTNFhigh is a major contributor to liver injury in LPS/D-gal-induced hepatitis.

Collaboration


Dive into the Zhuoya Li's collaboration.

Top Co-Authors

Avatar

Jing Wang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Bingjiao Yin

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiaodan Jiang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Feili Gong

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hailong Zhang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Huifang Liang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Dan Yan

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hongping Ba

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Wei Feng

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Mingxia Yu

Huazhong University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge