Network


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

Hotspot


Dive into the research topics where Xuexian O. Yang is active.

Publication


Featured researches published by Xuexian O. Yang.


Nature Immunology | 2005

A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17.

Heon Park; Zhaoxia Li; Xuexian O. Yang; Seon Hee Chang; Roza Nurieva; Yi Hong Wang; Ying Wang; Leroy Hood; Zhou Zhu; Qiang Tian; Chen Dong

Interleukin 17 (IL-17) has been linked to autoimmune diseases, although its regulation and function have remained unclear. Here we have evaluated in vitro and in vivo the requirements for the differentiation of naive CD4 T cells into effector T helper cells that produce IL-17. This process required the costimulatory molecules CD28 and ICOS but was independent of the cytokines and transcription factors required for T helper type 1 or type 2 differentiation. Furthermore, both IL-4 and interferon-γ negatively regulated T helper cell production of IL-17 in the effector phase. In vivo, antibody to IL-17 inhibited chemokine expression in the brain during experimental autoimmune encephalomyelitis, whereas overexpression of IL-17 in lung epithelium caused chemokine production and leukocyte infiltration. Thus, IL-17 expression characterizes a unique T helper lineage that regulates tissue inflammation.


Nature | 2007

Essential autocrine regulation by IL-21 in the generation of inflammatory T cells

Roza Nurieva; Xuexian O. Yang; Gustavo J. Martinez; Yongliang Zhang; Athanasia D. Panopoulos; Li Ma; Kimberly S. Schluns; Qiang Tian; Stephanie S. Watowich; Anton M. Jetten; Chen Dong

After activation, CD4+ helper T (TH) cells differentiate into distinct effector subsets that are characterized by their unique cytokine expression and immunoregulatory function. During this differentiation, TH1 and TH2 cells produce interferon-γ and interleukin (IL)-4, respectively, as autocrine factors necessary for selective lineage commitment. A distinct TH subset, termed THIL-17, TH17 or inflammatory TH (THi), has been recently identified as a distinct TH lineage mediating tissue inflammation. TH17 differentiation is initiated by transforming growth factor-β and IL-6 (refs 5–7) and reinforced by IL-23 (ref. 8), in which signal transduction and activators of transcription (STAT)3 and retinoic acid receptor-related orphan receptor (ROR)-γ mediate the lineage specification. TH17 cells produce IL-17, IL-17F and IL-22, all of which regulate inflammatory responses by tissue cells but have no importance in TH17 differentiation. Here we show that IL-21 is another cytokine highly expressed by mouse TH17 cells. IL-21 is induced by IL-6 in activated T cells, a process that is dependent on STAT3 but not ROR-γ. IL-21 potently induces TH17 differentiation and suppresses Foxp3 expression, which requires STAT3 and ROR-γ, which is encoded by Rorc. IL-21 deficiency impairs the generation of TH17 cells and results in protection against experimental autoimmune encephalomyelitis. IL-21 is therefore an autocrine cytokine that is sufficient and necessary for TH17 differentiation, and serves as a target for treating inflammatory diseases.


Immunity | 2008

T Helper 17 Lineage Differentiation Is Programmed by Orphan Nuclear Receptors RORα and RORγ

Xuexian O. Yang; Bhanu P. Pappu; Roza Nurieva; Askar M. Akimzhanov; Hong Soon Kang; Yeonseok Chung; Li Ma; Bhavin Shah; Athanasia D. Panopoulos; Kimberly S. Schluns; Stephanie S. Watowich; Qiang Tian; Anton M. Jetten; Chen Dong

T cell functional differentiation is mediated by lineage-specific transcription factors. T helper 17 (Th17) has been recently identified as a distinct Th lineage mediating tissue inflammation. Retinoic acid receptor-related orphan receptor gamma (ROR gamma) was shown to regulate Th17 differentiation; ROR gamma deficiency, however, did not completely abolish Th17 cytokine expression. Here, we report Th17 cells highly expressed another related nuclear receptor, ROR alpha, induced by transforming growth factor-beta and interleukin-6 (IL-6), which is dependent on signal transducer and activator of transcription 3. Overexpression of ROR alpha promoted Th17 differentiation, possibly through the conserved noncoding sequence 2 in Il17-Il17f locus. ROR alpha deficiency resulted in reduced IL-17 expression in vitro and in vivo. Furthermore, ROR alpha and ROR gamma coexpression synergistically led to greater Th17 differentiation. Double deficiencies in ROR alpha and ROR gamma globally impaired Th17 generation and completely protected mice against experimental autoimmune encephalomyelitis. Therefore, Th17 differentiation is directed by two lineage-specific nuclear receptors, ROR alpha and ROR gamma.


Journal of Biological Chemistry | 2007

STAT3 Regulates Cytokine-mediated Generation of Inflammatory Helper T Cells

Xuexian O. Yang; Athanasia D. Panopoulos; Roza Nurieva; Seon Hee Chang; Demin Wang; Stephanie S. Watowich; Chen Dong

Interleukin-17 (IL-17)-producing helper T (TH) cells, named as THIL-17, TH17, or inflammatory TH (THi), have been recently identified as a novel effector lineage. However, how cytokine signals mediate THi differentiation is unclear. We found that IL-6 functioned to up-regulate IL-23R and that IL-23 synergized with IL-6 in promoting THi generation. STAT3, activated by both IL-6 and IL-23, plays a critical role in THi development. A hyperactive form of STAT3 promoted THi development, whereas this differentiation process was greatly impaired in STAT3-deficient T cells. Moreover, STAT3 regulated the expression of retinoic acid receptor-related orphan receptor γ-T (RORγt), a THi-specific transcriptional regulator; STAT3 deficiency impaired RORγt expression and led to elevated expression of T-box expressed in T cells (T-bet) and Forkhead box P3 (Foxp3). Our data thus demonstrate a pathway whereby cytokines regulate THi differentiation through a selective STAT transcription factor that functions to regulate lineage-specific gene expression.


Science | 2009

Bcl6 Mediates the Development of T Follicular Helper Cells

Roza Nurieva; Yeonseok Chung; Gustavo J. Martinez; Xuexian O. Yang; Shinya Tanaka; Tatyana D. Matskevitch; Yi Hong Wang; Chen Dong

T Follicular Helper Cell Differentiation When B cells respond to an infection, they often require help from CD4+ T cells to mount a proper response. It is thought that a subset of CD4+ effector T cells, called T follicular helper cells (TFH), performs this function. Several subsets of effector CD4+ T cells arise, depending on the type of infection, which have distinct transcriptional programs driving their differentiation. Whether this is also the case for TFH cells has not been clear (see the Perspective by Awasthi and Kuchroo). Nurieva et al. (p. 1001, published online 23 July) and Johnston et al. (p. 1006; published online 16 July) now demonstrate that the transcription factor Bcl6 is both necessary and sufficient for TFH differentiation and subsequent B cell–mediated immunity, suggesting that it is a master regulator of this lineage. Johnston et al. also show that expression of Bcl6 and the transcription factor, Blimp-1, are reciprocally regulated in TFH cells and that, when ectopically expressed, Blimp-1 inhibits TFH development. The transcription factors that regulate follicular T helper cell differentiation are identified. A fundamental function of CD4+ helper T (TH) cells is the regulation of B cell–mediated humoral immunity. Development of T follicular helper (TFH) cells that provide help to B cells is mediated by the cytokines interleukin-6 and interleukin-21 but is independent of TH1, TH2, and TH17 effector cell lineages. Here, we characterize the function of Bcl6, a transcription factor selectively expressed in TFH cells. Bcl6 expression is regulated by interleukin-6 and interleukin-21. Bcl6 overexpression induced TFH-related gene expression and inhibited other TH lineage cell differentiation in a DNA binding–dependent manner. Moreover, Bcl6 deficiency in T cells resulted in impaired TFH cell development and germinal center reactions, and altered production of other effector T cell subsets. Our data thus illustrate that Bcl6 is required for programming of TFH cell generation.


Immunity | 2008

Molecular Antagonism and Plasticity of Regulatory and Inflammatory T Cell Programs

Xuexian O. Yang; Roza Nurieva; Gustavo J. Martinez; Hong Soon Kang; Yeonseok Chung; Bhanu P. Pappu; Bhavin Shah; Seon Hee Chang; Kimberly S. Schluns; Stephanie S. Watowich; Xin-Hua Feng; Anton M. Jetten; Chen Dong

Regulatory T (Treg) and T helper 17 (Th17) cells were recently proposed to be reciprocally regulated during differentiation. To understand the underlying mechanisms, we utilized a Th17 reporter mouse with a red fluorescent protein (RFP) sequence inserted into the interleukin-17F (IL-17F) gene. Using IL-17F-RFP together with a Foxp3 reporter, we found that the development of Th17 and Foxp3(+) Treg cells was associated in immune responses. Although TGF-beta receptor I signaling was required for both Foxp3 and IL-17 induction, SMAD4 was only involved in Foxp3 upregulation. Foxp3 inhibited Th17 differentiation by antagonizing the function of the transcription factors RORgammat and ROR*. In contrast, IL-6 overcame this suppressive effect of Foxp3 and, together with IL-1, induced genetic reprogramming in Foxp3(+) Treg cells. STAT3 regulated Foxp3 downregulation, whereas STAT3, RORgamma, and ROR* were required for IL-17 expression in Treg cells. Our data demonstrate molecular antagonism and plasticity of Treg and Th17 cell programs.


Immunity | 2008

Generation of T Follicular Helper Cells Is Mediated by Interleukin-21 but Independent of T Helper 1, 2, or 17 Cell Lineages

Roza Nurieva; Yeonseok Chung; Daehee Hwang; Xuexian O. Yang; Hong Soon Kang; Li Ma; Yi Hong Wang; Stephanie S. Watowich; Anton M. Jetten; Qiang Tian; Chen Dong

After activation, CD4(+) helper T (Th) cells differentiate into distinct effector subsets. Although chemokine (C-X-C motif) receptor 5-expressing T follicular helper (Tfh) cells are important in humoral immunity, their developmental regulation is unclear. Here we show that Tfh cells had a distinct gene expression profile and developed in vivo independently of the Th1 or Th2 cell lineages. Tfh cell generation was regulated by ICOS ligand (ICOSL) expressed on B cells and was dependent on interleukin-21 (IL-21), IL-6, and signal transducer and activator of transcription 3 (STAT3). However, unlike Th17 cells, differentiation of Tfh cells did not require transforming growth factor beta (TGF-beta) or Th17-specific orphan nuclear receptors RORalpha and RORgamma in vivo. Finally, naive T cells activated in vitro in the presence of IL-21 but not TGF-beta signaling preferentially acquired Tfh gene expression and promoted germinal-center reactions in vivo. This study thus demonstrates that Tfh is a distinct Th cell lineage.


Journal of Experimental Medicine | 2008

Regulation of inflammatory responses by IL-17F

Xuexian O. Yang; Seon Hee Chang; Heon Park; Roza Nurieva; Bhavin Shah; Luis Acero; Yi-Hong Wang; Kimberly S. Schluns; Russell R. Broaddus; Zhou Zhu; Chen Dong

Although interleukin (IL) 17 has been extensively characterized, the function of IL-17F, which has an expression pattern regulated similarly to IL-17, is poorly understood. We show that like IL-17, IL-17F regulates proinflammatory gene expression in vitro, and this requires IL-17 receptor A, tumor necrosis factor receptor–associated factor 6, and Act1. In vivo, overexpression of IL-17F in lung epithelium led to infiltration of lymphocytes and macrophages and mucus hyperplasia, similar to observations made in IL-17 transgenic mice. To further understand the function of IL-17F, we generated and analyzed mice deficient in IL-17F or IL-17. IL-17, but not IL-17F, was required for the initiation of experimental autoimmune encephalomyelitis. Mice deficient in IL-17F, but not IL-17, had defective airway neutrophilia in response to allergen challenge. Moreover, in an asthma model, although IL-17 deficiency reduced T helper type 2 responses, IL-17F–deficient mice displayed enhanced type 2 cytokine production and eosinophil function. In addition, IL-17F deficiency resulted in reduced colitis caused by dextran sulfate sodium, whereas IL-17 knockout mice developed more severe disease. Our results thus demonstrate that IL-17F is an important regulator of inflammatory responses that seems to function differently than IL-17 in immune responses and diseases.


Immunity | 2009

T helper 17 cells promote cytotoxic T cell activation in tumor immunity.

Pawel Muranski; Yeonseok Chung; Xuexian O. Yang; Tomohide Yamazaki; Sijie Lu; Patrick Hwu; Nicholas P. Restifo; Willem W. Overwijk; Chen Dong

Although T helper 17 (Th17) cells have been found in tumor tissues, their function in cancer immunity is unclear. We found that interleukin-17A (IL-17A)-deficient mice were more susceptible to developing lung melanoma. Conversely, adoptive T cell therapy with tumor-specific Th17 cells prevented tumor development. Importantly, the Th17 cells retained their cytokine signature and exhibited stronger therapeutic efficacy than Th1 cells. Unexpectedly, therapy using Th17 cells elicited a remarkable activation of tumor-specific CD8(+) T cells, which were necessary for the antitumor effect. Th17 cells promoted dendritic cell recruitment into the tumor tissues and in draining lymph nodes increased CD8 alpha(+) dendritic cells containing tumor material. Moreover, Th17 cells promoted CCL20 chemokine production by tumor tissues, and tumor-bearing CCR6-deficient mice did not respond to Th17 cell therapy. Thus, Th17 cells elicited a protective inflammation that promotes the activation of tumor-specific CD8(+) T cells. These findings have important implications in antitumor immunotherapies.


Journal of Immunology | 2008

CCR6 Regulates the Migration of Inflammatory and Regulatory T Cells

Tomohide Yamazaki; Xuexian O. Yang; Yeonseok Chung; Atsushi Fukunaga; Roza Nurieva; Bhanu P. Pappu; Hong Soon Kang; Li Ma; Athanasia D. Panopoulos; Suzanne Craig; Stephanie S. Watowich; Anton M. Jetten; Qiang Tian; Chen Dong

Th17 and regulatory T (Treg) cells play opposite roles in autoimmune diseases. However, the mechanisms underlying their proper migration to inflammatory tissues are unclear. In this study, we report that these two T cell subsets both express CCR6. CCR6 expression in Th17 cells is regulated by TGF-β and requires two nuclear receptors, RORα and RORγ. Th17 cells also express the CCR6 ligand CCL20, which is induced synergistically by TGF-β and IL-6, which requires STAT3, RORγ and IL-21. Th17 cells, by producing CCL20, promote migration of Th17 and Treg cells in vitro in a CCR6-dependent manner. Lack of CCR6 in Th17 cells reduces the severity of experimental autoimmune encephalomyelitis and Th17 and Treg recruitment into inflammatory tissues. Similarly, CCR6 on Treg cells is also important for their recruitment into inflammatory tissues. Our data indicate an important role of CCR6 in Treg and Th17 cell migration.

Collaboration


Dive into the Xuexian O. Yang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roza Nurieva

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yeonseok Chung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seon Hee Chang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Stephanie S. Watowich

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anton M. Jetten

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qiang Tian

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gustavo J. Martinez

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Kimberly S. Schluns

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge