Network


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

Hotspot


Dive into the research topics where Xindong Liu is active.

Publication


Featured researches published by Xindong Liu.


Nature | 2014

Transcription factor achaete-scute homologue 2 initiates follicular T-helper-cell development

Xindong Liu; Xin Chen; Bo Zhong; Aibo Wang; Xiaohu Wang; Fuliang Chu; Roza Nurieva; Xiaowei Yan; Ping Chen; Laurens G. van der Flier; Hiroko Nakatsukasa; Sattva S. Neelapu; WanJun Chen; Hans Clevers; Qiang Tian; Hai Qi; Lai Wei; Chen Dong

In immune responses, activated T cells migrate to B-cell follicles and develop into follicular T-helper (TFH) cells, a recently identified subset of CD4+ T cells specialized in providing help to B lymphocytes in the induction of germinal centres. Although Bcl6 has been shown to be essential in TFH-cell function, it may not regulate the initial migration of T cells or the induction of the TFH program, as exemplified by C-X-C chemokine receptor type 5 (CXCR5) upregulation. Here we show that expression of achaete-scute homologue 2 (Ascl2)—a basic helix–loop–helix (bHLH) transcription factor—is selectively upregulated in TFH cells. Ectopic expression of Ascl2 upregulates CXCR5 but not Bcl6, and downregulates C-C chemokine receptor 7 (CCR7) expression in T cells in vitro, as well as accelerating T-cell migration to the follicles and TFH-cell development in vivo in mice. Genome-wide analysis indicates that Ascl2 directly regulates TFH-related genes whereas it inhibits expression of T-helper cell 1 (TH1) and TH17 signature genes. Acute deletion of Ascl2, as well as blockade of its function with the Id3 protein in CD4+ T cells, results in impaired TFH-cell development and germinal centre response. Conversely, mutation of Id3, known to cause antibody-mediated autoimmunity, greatly enhances TFH-cell generation. Thus, Ascl2 directly initiates TFH-cell development.


Journal of Experimental Medicine | 2012

Bcl6 expression specifies the T follicular helper cell program in vivo

Xindong Liu; Xiaowei Yan; Bo Zhong; Roza Nurieva; Aibo Wang; Xiaohu Wang; Yi-Hong Wang; Seon Hee Chang; Enric Esplugues; Richard A. Flavell; Qiang Tian; Chen Dong

A novel Bcl6 reporter mouse is used to dissect the developmental requirements, plasticity, and genetic profile of Tfh cells.


Immunological Reviews | 2013

Transcriptional regulation of follicular T-helper (Tfh) cells.

Xindong Liu; Roza Nurieva; Chen Dong

T‐follicular helper (Tfh) cells are a new subset of effector CD4+ T cells that are specialized in helping B cells in the germinal center reaction. Tfh cells are distinct from other established CD4+ T‐cell lineages, Th1, Th2, Th17, and T‐regulatory cells, in their gene expression profiles. Tfh cell differentiation results from a network of transcriptional regulation by a master transcriptional factor Bcl6 as well as IRF4, c‐Maf, Batf, and STAT3/5. During Tfh cell ontogeny, increased CXCR5 expression directs activated T‐cell migration to the follicles, and their interaction with B cells leads to Bcl6 upregulation, which helps establish effector and memory Tfh cell program. This review summarizes the recent progress in molecular mechanisms underlying Tfh differentiation and discusses the future perspectives for this important area of research.


Nature Immunology | 2012

Negative regulation of IL-17-mediated signaling and inflammation by the ubiquitin-specific protease USP25

Bo Zhong; Xikui Liu; Xiaohu Wang; Seon Hee Chang; Xindong Liu; Aibo Wang; Joseph M. Reynolds; Chen Dong

Interleukin 17 (IL-17) is important in infection and autoimmunity; how it signals remains poorly understood. In this study, we identified the ubiquitin-specific protease USP25 as a negative regulator of IL-17-mediated signaling and inflammation. Overexpression of USP25 inhibited IL-17-triggered signaling, whereas USP25 deficiency resulted in more phosphorylation of the inhibitor IκBα and kinase Jnk and higher expression of chemokines and cytokines, as well as a prolonged half-life for chemokine CXCL1–encoding mRNA after treatment with IL-17. Consistent with that, Usp25−/− mice showed greater sensitivity to IL-17-dependent inflammation and autoimmunity in vivo. Mechanistically, stimulation with IL-17 induced the association of USP25 with the adaptors TRAF5 and TRAF6, and USP25 induced removal of Lys63-linked ubiquitination in TRAF5 and TRAF6 mediated by the adaptor Act1. Thus, our results demonstrate that USP25 is a deubiquitinating enzyme (DUB) that negatively regulates IL-17-triggered signaling.


Journal of Immunology | 2007

NF-κB Signaling Regulates Functional Expression of the MHC Class I-Related Neonatal Fc Receptor for IgG via Intronic Binding Sequences

Xindong Liu; Lilin Ye; Gregory J. Christianson; Jun Qi Yang; Derry C. Roopenian; Xiaoping Zhu

The neonatal Fc receptor for IgG (FcRn) functions to transport maternal IgG to a fetus or newborn and to protect IgG from degradation. Although FcRn is expressed in a variety of tissues and cell types, the extent to which FcRn expression is regulated by immunological and inflammatory events remains unknown. Stimulation of intestinal epithelial cell lines, macrophage-like THP-1, and freshly isolated human monocytes with the cytokine TNF-α rapidly up-regulated FcRn gene expression. In addition, the TLR ligands LPS and CpG oligodeoxynucleotide enhanced the level of FcRn expression in THP-1 and monocytes. Treatment of TNF-stimulated THP-1 cells with the NF-κB-specific inhibitor or overexpression of a dominant negative mutant inhibitory NF-κB (IκBα; S32A/S36A) resulted in down-regulation of FcRn expression. By using chromatin immunoprecipitation we identified three NF-κB binding sequences within introns 2 and 4 of the human FcRn gene. An EMSA confirmed the p50/p50 and/or p65/p50 complex (s) bound to intron 2- or 4-derived oligonucleotides containing putative NF-κB binding sequences, respectively. The intronic NF-κB sequences in combination with the promoter or alone regulated the expression of a luciferase reporter gene in response to TNF-α stimulation or overexpression of NF-κB p65 and p50. DNA looping interactions potentially occurred after the stimulation between intronic NF-κB sequences and the FcRn promoter as shown by a chromosome conformation capture assay. Finally, TNF-α stimulations enhanced IgG transport across an intestinal Caco-2 epithelial monolayer. Together, these data provide the first evidence that NF-κB signaling via intronic sequences regulates FcRn expression and function.


Journal of Immunology | 2011

The Neonatal FcR-Mediated Presentation of Immune-Complexed Antigen Is Associated with Endosomal and Phagosomal pH and Antigen Stability in Macrophages and Dendritic Cells

Xindong Liu; Li Lu; Ziyan Yang; Senthilkumar Palaniyandi; Rongyu Zeng; Lian Yong Gao; David M. Mosser; Derry C. Roopenian; Xiaoping Zhu

The FcγRs found on macrophages (Mϕs) and dendritic cells (DCs) efficiently facilitate the presentation or cross-presentation of immune-complexed Ags to T cells. We found that the MHC class I-related neonatal FcR for IgG (FcRn) in both Mϕs and DCs failed to have a strong effect on the cross-presentation of immune complex (IC) OVA Ag to CD8+ T cells. Interestingly, endosomal FcRn enhanced the presentation of the monomeric OVA-IC to CD4+ T cells robustly, whereas FcRn in phagosomes exerted distinctive effects on Ag presentation between Mϕs and DCs. The presentation of phagocytosed OVA-ICs to CD4+ T cells was considerably enhanced on wild-type versus FcRn-deficient Mϕs, but was not affected in FcRn-deficient DCs. This functional discrepancy was associated with the dependence of IgG–FcRn binding in an acidic pH. Following phagocytosis, the phagosomal pH dropped rapidly to <6.5 in Mϕs but remained in the neutral range in DCs. This disparity in pH determined the rate of degradation of phagocytosed ICs. Thus, our findings reveal that FcRn expression has a different effect on Ag processing and presentation of ICs to CD4+ T cells in the endosomal versus phagosomal compartments of Mϕs versus DCs.


Journal of Virology | 2011

A Neonatal Fc Receptor-Targeted Mucosal Vaccine Strategy Effectively Induces HIV-1 Antigen-Specific Immunity to Genital Infection

Li Lu; Senthilkumar Palaniyandi; Rongyu Zeng; Yu Bai; Xindong Liu; Yunsheng Wang; C. David Pauza; Derry C. Roopenian; Xiaoping Zhu

ABSTRACT Strategies to prevent the sexual transmission of HIV include vaccines that elicit durable, protective mucosal immune responses. A key to effective mucosal immunity is the capacity for antigens administered locally to cross epithelial barriers. Given the role of neonatal Fc receptor (FcRn) in transferring IgG across polarized epithelial cells which line mucosal surfaces, FcRn might be useful for delivering HIV vaccine antigens across mucosal epithelial barriers to the underlying antigen-presenting cells. Chimeric proteins composed of HIV Gag (p24) fused to the Fc region of IgG (Gag-Fc) bind efficiently to airway mucosa and are transported across this epithelial surface. Mice immunized intranasally with Gag-Fc plus CpG adjuvant developed local and systemic immunity, including durable B and T cell memory. Gag-specific immunity was sufficiently potent to protect against an intravaginal challenge with recombinant vaccinia virus expressing the HIV Gag protein. Intranasal administration of a Gag-Fc/CpG vaccine protected at a distal mucosal site. Our data suggest that targeting of FcRn with chimeric immunogens may be an important strategy for mucosal immunization and should be considered a new approach for preventive HIV vaccines.


Journal of Immunology | 2008

Activation of the JAK/STAT-1 signaling pathway by IFN-γ can down-regulate functional expression of the MHC class I-related neonatal Fc receptor for IgG

Xindong Liu; Lilin Ye; Yu Bai; Habi Mojidi; Neil E. Simister; Xiaoping Zhu

Expression of many MHC genes is enhanced at the transcriptional or posttranscriptional level following exposure to the cytokine IFN-γ. However, in this study we found that IFN-γ down-regulated the constitutive expression of the neonatal Fc receptor (FcRn), an MHC class I-related molecule that functions to transport maternal IgG and protect IgG and albumin from degradation. Epithelial cell, macrophage-like THP-1 cell, and freshly isolated human PBMC exposure to IFN-γ resulted in a significant decrease of FcRn expression as assessed by real-time RT-PCR and Western blotting. The down-regulation of FcRn was not caused by apoptosis or the instability of FcRn mRNA. Chromatin immunoprecipitation and gel mobility shift assays showed that STAT-1 bound to an IFN-γ activation site in the human FcRn promoter region. Luciferase expression from an FcRn promoter-luciferase reporter gene construct was not altered in JAK1- and STAT-1-deficient cells following exposure to IFN-γ, whereas expression of JAK1 or STAT-1 protein restored the IFN-γ inhibitory effect on luciferase activity. The repressive effect of IFN-γ on the FcRn promoter was selectively reversed or blocked by mutations of the core nucleotides in the IFN-γ activation site sequence and by overexpression of the STAT-1 inhibitor PIAS1 or the dominant negative phospho-STAT-1 mutations at Tyr-701 and/or Ser-727 residues. Furthermore, STAT-1 might down-regulate FcRn transcription through sequestering the transcriptional coactivator CREB binding protein/p300. Functionally, IFN-γ stimulation dampened bidirectional transport of IgG across a polarized Calu-3 lung epithelial monolayer. Taken together, our results indicate that the JAK/STAT-1 signaling pathway was necessary and sufficient to mediate the down-regulation of FcRn gene expression by IFN-γ.


Immunological Reviews | 2011

Molecular mechanisms of T-cell tolerance

Roza Nurieva; Xindong Liu; Chen Dong

Summary:  CD4+ T cells are the master regulators of adaptive immune responses, and many autoimmune diseases arise due to a breakdown of self‐tolerance in CD4+ T cells. Activation of CD4+ T cells is regulated by not only the binding of peptide‐major histocompatibility complexes to T‐cell receptor but also costimulatory signals from antigen‐presenting cells. Recently, there has been progress in understanding the extracellular and intracellular mechanisms that are required for implementation and maintenance of T‐cell tolerance. Understanding of the molecular mechanisms underlying T‐cell tolerance will lead to development of pharmacological approaches either to promote the tolerance state in terms of autoimmunity or to break tolerance in cancer.


Journal of Immunology | 2008

The MHC Class II-Associated Invariant Chain Interacts with the Neonatal Fcγ Receptor and Modulates Its Trafficking to Endosomal/Lysosomal Compartments

Lilin Ye; Xindong Liu; Subrat N. Rout; Zili Li; Yongqi Yan; Li Lu; Tirumalai Kamala; Navreet K. Nanda; Wenxia Song; Siba K. Samal; Xiaoping Zhu

The neonatal Fc receptor for IgG (FcRn) transfers maternal IgG to the offspring and protects IgG from degradation. The FcRn resides in an acidic intracellular compartment, allowing it to bind IgG. In this study, we found the association of FcRn and invariant chain (Ii). The interaction was initiated within the endoplasmic reticulum by Ii binding to either the FcRn H chain alone or FcRn H chain-β2-microglobulin complex and appeared to be maintained throughout the endocytic pathway. The CLIP in Ii was not required for FcRn-Ii association. The interaction was also detected in IFN-γ-treated THP-1, epithelial and endothelial cells, and immature mouse DCs. A truncated FcRn without the cytoplasmic tail was unable to traffic to early endosomes; however, its location in early endosomes was restored by Ii expression. FcRn was also detected in the late endosome/lysosome only in the presence of Ii or on exposure to IFN-γ. In immature human or mouse DCs, FcRn was barely detected in the late endosome/lysosome in the absence of Ii. Furthermore, the cytoplasmic tail of Ii conferred tailless FcRn to route to both the early endosome and late endosome/lysosome in a hybrid molecule. Because the FcRn is expressed in macrophages and DCs or epithelial and endothelial cells where Ii is induced under inflammation and infection, these results reveal the complexity of FcRn trafficking in which Ii is capable of expanding the boundary of FcRn trafficking. Taken together, the intracellular trafficking of FcRn is regulated by its intrinsic sorting information and/or an interaction with Ii chain.

Collaboration


Dive into the Xindong Liu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aibo Wang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xia Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Roza Nurieva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xiu-wu Bian

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
Top Co-Authors

Avatar

Qiang Tian

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge