Network


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

Hotspot


Dive into the research topics where Xiaoyi Yuan is active.

Publication


Featured researches published by Xiaoyi Yuan.


Science | 2013

Cleavage of Fibrinogen by Proteinases Elicits Allergic Responses Through Toll-Like Receptor 4

Valentine Millien; Wen Lu; Joanne Shaw; Xiaoyi Yuan; Garbo Mak; Luz Roberts; Li Zhen Song; J. Morgan Knight; Chad J. Creighton; Amber Luong; Farrah Kheradmand; David B. Corry

Allergy Induction Proteinases found in fungi and other allergens elicit allergic inflammation, but how they do so is far from clear. It is also unclear how pattern recognition receptors, which detect invading microbes, drive allergic inflammation. Millien et al. (p. 792) shed light on this puzzle by showing that, in mice, induction of allergic inflammation requires proteinase-dependent cleavage of the clotting factor fibrinogen, leading to generation of a ligand that activates the pattern-recognition receptor, Toll-like receptor 4 (TLR4). Cleaved fibrinogen signals through TLR4 to activate the innate immune system and recruit cells to the airway, which drives both allergic responses and antifungal immunity. Allergic inflammation requires proteinase-dependent cleavage of fibrinogen that activates innate immunity through Toll-like receptor 4. Proteinases and the innate immune receptor Toll-like receptor 4 (TLR4) are essential for expression of allergic inflammation and diseases such as asthma. A mechanism that links these inflammatory mediators is essential for explaining the fundamental basis of allergic disease but has been elusive. Here, we demonstrate that TLR4 is activated by airway proteinase activity to initiate both allergic airway disease and antifungal immunity. These outcomes were induced by proteinase cleavage of the clotting protein fibrinogen, yielding fibrinogen cleavage products that acted as TLR4 ligands on airway epithelial cells and macrophages. Thus, allergic airway inflammation represents an antifungal defensive strategy that is driven by fibrinogen cleavage and TLR4 activation. These findings clarify the molecular basis of allergic disease and suggest new therapeutic strategies.


Science Translational Medicine | 2012

Cigarette Smoke Induction of Osteopontin (SPP1) Mediates T H 17 Inflammation in Human and Experimental Emphysema

Ming Shan; Xiaoyi Yuan; Li Zhen Song; Luz Roberts; Nazanin Zarinkamar; Alexander Seryshev; Yiqun Zhang; Susan G. Hilsenbeck; Seon Hee Chang; Chen Dong; David B. Corry; Farrah Kheradmand

The destruction of lung tissue in emphysema is orchestrated by antigen-presenting cells that have been activated by smoke. When Smoke Gets in Your Lungs Even without the warning label on the cigarette box, everyone knows that tobacco smoke is bad for you. But what really happens inside the lung to cause disease? Shan et al. have married studies in patients with emphysema and in mice exposed to tobacco smoke for several months to identify some of the crucial events that cause one serious lung disease—emphysema. They find that the antigen-presenting cells of the immune system are culpable; indeed, transfer of these cells from a mouse with emphysema into a healthy mouse induces disease. Further, they identify the signaling molecules in these cells that are activated by smoke (osteopontin and the transcription factor Irf7), which may prove to be useful targets for therapies. First, the authors verified that mice exposed to smoke carried similar immune cells in their lungs as do human smokers. These cells, including their secretory products IFN-γ and IL-17A, trigger the lung tissue destruction that robs emphysema patients of their ability to breathe. What activates these immune responses? The authors finger antigen-presenting cells by transferring these cells from diseased mice to healthy ones, showing that the disease transfers along with them. Then, tracing the process one step back, the authors look for smoke-induced genes in these antigen-presenting cells and identify osteopontin and Irf7, a transcription factor regulated by Spp1, as key mediators of smoke-induced cell activation. There are earlier steps in the process that have yet to be elucidated, but the cause of the havoc wreaked inside the lung by tobacco smoke is getting clearer. As a bonus, another cell type that participates in the mayhem unleashed in the emphysemic lung was uncovered. The γδ T cell, usually very rare, was unexpectedly found to be a good guy. Induced along with the destructive cells, these cells curtailed the damage but ultimately lost the battle. Perhaps bolstering the abilities of the helpful γδ T cells could ultimately help to treat serious immunogenic destructive processes in the lung. Smoking-related lung diseases are among the leading causes of death worldwide, underscoring the need to understand their pathogenesis and develop new effective therapies. We have shown that CD1a+ antigen-presenting cells (APCs) from lungs of patients with emphysema can induce autoreactive T helper 1 (TH1) and TH17 cells. Similarly, the canonical cytokines interferon-γ (IFN-γ) and interleukin-17A (IL-17A) are specifically linked to lung destruction in smokers, but how smoke activates APCs to mediate emphysema remains unknown. Here, we show that, in addition to increasing IFN-γ expression, cigarette smoke increased the expression of IL-17A in both CD4+ and γδ T cells from mouse lung. IL-17A deficiency resulted in attenuation of, whereas lack of γδ T cells exacerbated, smoke-induced emphysema in mice. Adoptive transfer of lung APCs isolated from mice with emphysema revealed that this cell population was capable of transferring disease even in the absence of active smoke exposure, a process that was dependent on IL-17A expression. Spp1 (the gene for osteopontin) was highly expressed in the pathogenic lung APCs of smoke-exposed mice and was required for the TH17 responses and emphysema in vivo, in part through its inhibition of the expression of the transcription factor Irf7. Thus, the Spp1-Irf7 axis is critical for induction of pathological TH17 responses, revealing a major mechanism by which smoke activates lung APCs to induce emphysema and identifying a pathway that could be targeted for therapeutic purposes.


Journal of Clinical Investigation | 2014

Agonistic induction of PPARγ reverses cigarette smoke–induced emphysema

Ming Shan; Ran You; Xiaoyi Yuan; Michael V. Frazier; Paul Porter; Alexander Seryshev; Jeong Soo Hong; Li Zhen Song; Yiqun Zhang; Susan G. Hilsenbeck; Lawrence W. Whitehead; Nazanin Zarinkamar; Sarah Perusich; David B. Corry; Farrah Kheradmand

The development of emphysema in humans and mice exposed to cigarette smoke is promoted by activation of an adaptive immune response. Lung myeloid dendritic cells (mDCs) derived from cigarette smokers activate autoreactive Th1 and Th17 cells. mDC-dependent activation of T cell subsets requires expression of the SPP1 gene, which encodes osteopontin (OPN), a pleiotropic cytokine implicated in autoimmune responses. The upstream molecular events that promote SPP1 expression and activate mDCs in response to smoke remain unknown. Here, we show that peroxisome proliferator-activated receptor γ (PPARG/Pparg) expression was downregulated in mDCs of smokers with emphysema and mice exposed to chronic smoke. Conditional knockout of PPARγ in APCs using Cd11c-Cre Pparg(flox/flox) mice led to spontaneous lung inflammation and emphysema that resembled the phenotype of smoke-exposed mice. The inflammatory phenotype of Cd11c-Cre Pparg(flox/flox) mice required OPN, suggesting an antiinflammatory mechanism in which PPARγ negatively regulates Spp1 expression in the lung. A 2-month treatment with a PPARγ agonist reversed emphysema in WT mice despite continual smoke exposure. Furthermore, endogenous PPARγ agonists were reduced in the plasma of smokers with emphysema. These findings reveal a proinflammatory pathway, in which reduced PPARγ activity promotes emphysema, and suggest that targeting this pathway in smokers could prevent and reverse emphysema.


Nature Immunology | 2015

The microRNA miR-22 inhibits the histone deacetylase HDAC4 to promote TH17 cell-dependent emphysema

Wen Lu; Ran You; Xiaoyi Yuan; Tianshu Yang; Errol L. G. Samuel; Daniela C. Marcano; William K. A. Sikkema; James M. Tour; Antony Rodriguez; Farrah Kheradmand; David B. Corry

Smoking-related emphysema is a chronic inflammatory disease driven by the TH17 subset of helper T cells through molecular mechanisms that remain obscure. Here we explored the role of the microRNA miR-22 in emphysema. We found that miR-22 was upregulated in lung myeloid dendritic cells (mDCs) of smokers with emphysema and antigen-presenting cells (APCs) of mice exposed to smoke or nanoparticulate carbon black (nCB) through a mechanism that involved the transcription factor NF-κB. Mice deficient in miR-22, but not wild-type mice, showed attenuated TH17 responses and failed to develop emphysema after exposure to smoke or nCB. We further found that miR-22 controlled the activation of APCs and TH17 responses through the activation of AP-1 transcription factor complexes and the histone deacetylase HDAC4. Thus, miR-22 is a critical regulator of both emphysema and TH17 responses.


eLife | 2015

Nanoparticulate carbon black in cigarette smoke induces DNA cleavage and Th17-mediated emphysema

Ran You; Wen Lu; Ming Shan; Jacob M. Berlin; Errol L. G. Samuel; Daniela C. Marcano; Zhengzong Sun; William K. A. Sikkema; Xiaoyi Yuan; Li-zhen Song; Amanda Y. Hendrix; James M. Tour; David B. Corry; Farrah Kheradmand

Chronic inhalation of cigarette smoke is the major cause of sterile inflammation and pulmonary emphysema. The effect of carbon black (CB), a universal constituent of smoke derived from the incomplete combustion of organic material, in smokers and non-smokers is less known. In this study, we show that insoluble nanoparticulate carbon black (nCB) accumulates in human myeloid dendritic cells (mDCs) from emphysematous lung and in CD11c+ lung antigen presenting cells (APC) of mice exposed to smoke. Likewise, nCB intranasal administration induced emphysema in mouse lungs. Delivered by smoking or intranasally, nCB persisted indefinitely in mouse lung, activated lung APCs, and promoted T helper 17 cell differentiation through double-stranded DNA break (DSB) and ASC-mediated inflammasome assembly in phagocytes. Increasing the polarity or size of CB mitigated many adverse effects. Thus, nCB causes sterile inflammation, DSB, and emphysema and explains adverse health outcomes seen in smokers while implicating the dangers of nCB exposure in non-smokers. DOI: http://dx.doi.org/10.7554/eLife.09623.001


Mucosal Immunology | 2015

Activation of C3a receptor is required in cigarette smoke-mediated emphysema

Xiaoyi Yuan; Ming Shan; Ran You; Michael V. Frazier; Monica Jeongsoo Hong; Rick A. Wetsel; Scott M. Drouin; Alexander Seryshev; Li-zhen Song; Lorraine D. Cornwell; Roger D. Rossen; David B. Corry; Farrah Kheradmand

Exposure to cigarette smoke can initiate sterile inflammatory responses in the lung and activate myeloid dendritic cells (mDCs) that induce differentiation of T helper type 1 (Th1) and Th17 cells in the emphysematous lungs. Consumption of complement proteins increases in acute inflammation, but the contribution of complement protein 3 (C3) to chronic cigarette smoke-induced immune responses in the lung is not clear. Here, we show that following chronic exposure to cigarette smoke, C3-deficient (C3−/−) mice develop less emphysema and have fewer CD11b+CD11c+ mDCs infiltrating the lungs as compared with wild-type mice. Proteolytic cleavage of C3 by neutrophil elastase releases C3a, which in turn increases the expression of its receptor (C3aR) on lung mDCs. Mice deficient in the C3aR (C3ar−/−) partially phenocopy the attenuated responses to chronic smoke observed in C3−/− mice. Consistent with a role for C3 in emphysema, C3 and its active fragments are deposited on the lung tissue of smokers with emphysema, and smoke-exposed mice. Together, these findings suggest a critical role for C3a through autocrine/paracrine induction of C3aR in the pathogenesis of cigarette smoke-induced sterile inflammation and provide new therapeutic targets for the treatment of emphysema.


Annals of the American Thoracic Society | 2014

Airway fibrinogenolysis and the initiation of allergic inflammation.

Valentine Millien; Wen Lu; Garbo Mak; Xiaoyi Yuan; J. Morgan Knight; Paul Porter; Farrah Kheradmand; David B. Corry

The past 15 years of allergic disease research have produced extraordinary improvements in our understanding of the pathogenesis of airway allergic diseases such as asthma. Whereas it was previously viewed as largely an immunoglobulin E-mediated process, the gradual recognition that T cells, especially Type 2 T helper (Th2) cells and Th17 cells, play a major role in asthma and related afflictions has inspired clinical trials targeting cytokine-based inflammatory pathways that show great promise. What has yet to be clarified about the pathogenesis of allergic inflammatory disorders, however, are the fundamental initiating factors, both exogenous and endogenous, that drive and sustain B- and T-cell responses that underlie the expression of chronic disease. Here we review how proteinases derived from diverse sources drive allergic responses. A central discovery supporting the proteinase hypothesis of allergic disease pathophysiology is the role played by airway fibrinogen, which in part appears to serve as a sensor of unregulated proteinase activity and which, when cleaved, both participates in a novel allergic signaling pathway through Toll-like receptor 4 and forms fibrin clots that contribute to airway obstruction. Unresolved at present is the ultimate source of airway allergenic proteinases. From among many potential candidates, perhaps the most intriguing is the possibility such enzymes derive from airway fungi. Together, these new findings expand both our knowledge of allergic disease pathophysiology and options for therapeutic intervention.


PLOS ONE | 2015

Long-Acting Beta Agonists Enhance Allergic Airway Disease.

John M. Knight; Garbo Mak; Joanne Shaw; Paul Porter; Catherine McDermott; Luz Roberts; Ran You; Xiaoyi Yuan; Valentine Millien; Yuping Qian; Li Zhen Song; Vincent Frazier; Choel Kim; Jeong Joo Kim; Richard A. Bond; Joshua D. Milner; Yuan Zhang; Pijus K. Mandal; Amber Luong; Farrah Kheradmand; John S. McMurray; David B. Corry

Asthma is one of the most common of medical illnesses and is treated in part by drugs that activate the beta-2-adrenoceptor (β2-AR) to dilate obstructed airways. Such drugs include long acting beta agonists (LABAs) that are paradoxically linked to excess asthma-related mortality. Here we show that LABAs such as salmeterol and structurally related β2-AR drugs such as formoterol and carvedilol, but not short-acting agonists (SABAs) such as albuterol, promote exaggerated asthma-like allergic airway disease and enhanced airway constriction in mice. We demonstrate that salmeterol aberrantly promotes activation of the allergic disease-related transcription factor signal transducer and activator of transcription 6 (STAT6) in multiple mouse and human cells. A novel inhibitor of STAT6, PM-242H, inhibited initiation of allergic disease induced by airway fungal challenge, reversed established allergic airway disease in mice, and blocked salmeterol-dependent enhanced allergic airway disease. Thus, structurally related β2-AR ligands aberrantly activate STAT6 and promote allergic airway disease. This untoward pharmacological property likely explains adverse outcomes observed with LABAs, which may be overcome by agents that antagonize STAT6.


Mucosal Immunology | 2018

Protective role of γδ T cells in cigarette smoke and influenza infection

M J Hong; B H Gu; M C Madison; C Landers; H Y Tung; M Kim; Xiaoyi Yuan; Ran You; A A Machado; B E Gilbert; P Soroosh; M Elloso; Li-zhen Song; M Chen; David B. Corry; G Diehl; Farrah Kheradmand

Airborne pathogens commonly trigger severe respiratory failure or death in smokers with lung disease. Cigarette smoking compromises the effectiveness of innate immunity against infections but the underlying mechanisms responsible for defective acquired immune responses in smokers remains less clear. We found that mice exposed to chronic cigarette smoke recovered poorly from primary Influenza A pneumonia with reduced type I and II interferons (IFNs) and viral-specific immunoglobulins, but recruited γδ T cells to the lungs that predominantly expressed interleukin 17A (IL-17A). Il-17a−/− mice exposed to smoke and infected with Influenza A also recruited γδ T cells to the lungs, but in contrast to wild-type mice, expressed increased IFNs, made protective influenza-specific antibodies, and recovered from infection. Depletion of IL-17A with blocking antibodies significantly increased T-bet expression in γδ T cells and improved recovery from acute Influenza A infection in air, but not smoke-exposed mice. In contrast, when exposed to smoke, γδ T cell deficient mice failed to mount an effective immune response to Influenza A and showed increased mortality. Our findings demonstrate a protective role for γδ T cells in smokers and suggest that smoke-induced increase in IL-17A inhibits the transcriptional programs required for their optimal anti-viral responses. Cigarette smoke induces IL-17A expression in the lungs and inhibits γδ T-cell-mediated protective anti-viral immune responses.


Cancer immunology research | 2018

IL17A Regulates Tumor Latency and Metastasis in Lung Adeno and Squamous SQ.2b and AD.1 Cancer

Ran You; Francesco J. DeMayo; Jian Liu; Sung-Nam Cho; Bryan M. Burt; Chad J. Creighton; Roberto F. Casal; Donald R. Lazarus; Wen Lu; Hui-Ying Tung; Xiaoyi Yuan; Andrea Hill-McAlester; Myunghoo Kim; Sarah Perusich; Loraine Cornwell; Daniel G. Rosen; Li-zhen Song; Silke Paust; Gretchen E. Diehl; David B. Corry; Farrah Kheradmand

The pro-inflammatory cytokine IL17A has antitumor effects in certain subtypes of non–small cell lung cancer. Mice whose tumors were infiltrated with IL17A-expressing T cells were associated with increased lung cancer latency and had fewer metastasis. Somatic mutations can promote malignant transformation of airway epithelial cells and induce inflammatory responses directed against resultant tumors. Tumor-infiltrating T lymphocytes (TIL) in early-stage non–small cell lung cancer (NSCLC) secrete distinct proinflammatory cytokines, but the contribution of these TILs to tumor development and metastasis remains unknown. We show here that TILs in early-stage NSCLC are biased toward IL17A expression (Th17) when compared with adjacent tumor-free tissue, whereas Th17 cells are decreased in tumor infiltrating locoregional lymph nodes in advanced NSCLC. Mice in which Pten and Smad4 (Pts4d/d) are deleted from airway epithelial cells develop spontaneous tumors, that share genetic signatures with squamous- (SQ.2b), and adeno- (AD.1) subtypes of human NSCLC. Pts4d/d mice globally lacking in IL17a (Pts4d/dIl17a–/–) showed decreased tumor latency and increased metastasis. Th17 cells were required for recruitment of CD103+ dendritic cells, and adoptive transfer of IL17a-sufficient CD4+ T cells reversed early tumor development and metastasis in Pts4d/dIl17a–/– mice. Together, these findings support a key role for Th17 cells in TILs associated with the Pts4d/d model of NSCLC and suggest therapeutic and biomarker strategies for human SQ2b and AD1 lung cancer. Cancer Immunol Res; 6(6); 645–57. ©2018 AACR.

Collaboration


Dive into the Xiaoyi Yuan's collaboration.

Top Co-Authors

Avatar

David B. Corry

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Farrah Kheradmand

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ran You

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Li-zhen Song

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Garbo Mak

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Luz Roberts

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ming Shan

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Valentine Millien

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Wen Lu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Amber Luong

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge