Network


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

Hotspot


Dive into the research topics where Dahui You is active.

Publication


Featured researches published by Dahui You.


Expert Review of Anti-infective Therapy | 2010

The use of a neonatal mouse model to study respiratory syncytial virus infections

Stephania A. Cormier; Dahui You; Srinivasa Honnegowda

Respiratory syncytial virus (RSV) infection is the most significant cause of viral death in infants worldwide. The significant morbidity and mortality associated with this disease underscores the urgent need for the development of an RSV vaccine. The development of an RSV vaccine has been hampered by our limited understanding of the human host immune system, which plays a significant role in RSV pathogenesis, susceptibility and vaccine efficacy. As a result, animal models have been developed to better understand the mechanisms by which RSV causes disease. Within the past few years, a revolutionary variation on these animal models has emerged – age at time of initial infection – and early studies in neonatal mice (aged <7 days at time of initial infection) indicate the validity of this model to understand RSV infection in infants. This article reviews available information on current murine and emerging neonatal mouse RSV models.


PLOS Pathogens | 2015

Respiratory Syncytial Virus Disease Is Mediated by Age-Variable IL-33.

Jordy Saravia; Dahui You; Bishwas Shrestha; Sridhar Jaligama; David Siefker; Greg I. Lee; Jeffrey N. Harding; Tamekia L. Jones; Cynthia Rovnaghi; Bindiya Bagga; John P. DeVincenzo; Stephania A. Cormier

Respiratory syncytial virus (RSV) is the most common cause of infant hospitalizations and severe RSV infections are a significant risk factor for childhood asthma. The pathogenic mechanisms responsible for RSV induced immunopathophysiology remain elusive. Using an age-appropriate mouse model of RSV, we show that IL-33 plays a critical role in the immunopathogenesis of severe RSV, which is associated with higher group 2 innate lymphoid cells (ILC2s) specifically in neonates. Infection with RSV induced rapid IL-33 expression and an increase in ILC2 numbers in the lungs of neonatal mice; this was not observed in adult mice. Blocking IL-33 with antibodies or using an IL-33 receptor knockout mouse during infection was sufficient to inhibit RSV immunopathogenesis (i.e., airway hyperresponsiveness, Th2 inflammation, eosinophilia, and mucus hyperproduction); whereas administration of IL-33 to adult mice during RSV infection was sufficient to induce RSV disease. Additionally, elevated IL-33 and IL-13 were observed in nasal aspirates from infants hospitalized with RSV; these cytokines declined during convalescence. In summary, IL-33 is necessary, either directly or indirectly, to induce ILC2s and the Th2 biased immunopathophysiology observed following neonatal RSV infection. This study provides a mechanism involving IL-33 and ILC2s in RSV mediated human asthma.


Respiratory Research | 2005

A role for airway remodeling during respiratory syncytial virus infection

David Becnel; Dahui You; Joshua Erskin; Dawn Dimina; Stephania A. Cormier

BackgroundSevere respiratory syncytial virus infection (RSV) during infancy has been shown to be a major risk factor for the development of subsequent wheeze. However, the reasons for this link remain unclear. The objective of this research was to determine the consequences of early exposure to RSV and allergen in the development of subsequent airway hyperreactivity (AHR) using a developmental time point in the mouse that parallels that of the human neonate.MethodsWeanling mice were sensitized and challenged with ovalbumin (Ova) and/or infected with RSV. Eight days after the last allergen challenge, various pathophysiological endpoints were examined.ResultsAHR in response to methacholine was enhanced only in weanling mice exposed to Ova and subsequently infected with RSV. The increase in AHR appeared to be unrelated to pulmonary RSV titer. Total bronchoalveolar lavage cellularity in these mice increased approximately two-fold relative to Ova alone and was attributable to increases in eosinophil and lymphocyte numbers. Enhanced pulmonary pathologies including persistent mucus production and subepithelial fibrosis were observed. Interestingly, these data correlated with transient increases in TNF-α, IFN-γ, IL-5, and IL-2.ConclusionThe observed changes in pulmonary structure may provide an explanation for epidemiological data suggesting that early exposure to allergens and RSV have long-term physiological consequences. Furthermore, the data presented here highlight the importance of preventative strategies against RSV infection of atopic individuals during neonatal development.


Environmental Toxicology and Pharmacology | 2010

In vitro and in vivo assessment of pulmonary risk associated with exposure to combustion generated fine particles

Baher Fahmy; Liren Ding; Dahui You; Slawo Lomnicki; Barry Dellinger; Stephania A. Cormier

Strong correlations exist between exposure to PM(2.5) and adverse pulmonary effects. PM(2.5) consists of fine (</=2.5 mum) and ultrafine (</=0.1 mum) particles with ultrafine particles accounting for >70% of the total particles. Environmentally persistent free radicals (EPFRs) have recently been identified in airborne PM(2.5). To determine the adverse pulmonary effects of EPFRs associated with exposure to elevated levels of PM(2.5), we engineered 2.5 mum surrogate EPFR-particle systems. We demonstrated that EPFRs generated greater oxidative stress in vitro, which was partly responsible for the enhanced cytotoxicity following exposure. In vivo studies using rats exposed to EPFRs containing particles demonstrated minimal adverse pulmonary effects. Additional studies revealed that fine particles failed to reach the alveolar region. Overall, our study implies qualitative differences between the health effects of PM size fractions.


Mucosal Immunology | 2014

Lung epithelial cells are essential effectors of inducible resistance to pneumonia

Jeffrey O. Cleaver; Dahui You; D. R. Michaud; F. A. Guzmán Pruneda; M. M. Leiva Juarez; Jiaqi Zhang; P. M. Weill; Roberto Adachi; Lei Gong; Seyed Javad Moghaddam; M. E. Poynter; Michael J. Tuvim; Scott E. Evans

Infectious pneumonias are the leading cause of death worldwide, particularly among immunocompromised patients. Therapeutic stimulation of the lungs’ intrinsic defenses with a unique combination of inhaled Toll-like receptor (TLR) agonists broadly protects mice against otherwise lethal pneumonias. As the survival benefit persists despite cytotoxic chemotherapy-related neutropenia, the cells required for protection were investigated. The inducibility of resistance was tested in mice with deficiencies of leukocyte lineages due to genetic deletions and in wild-type mice with leukocyte populations significantly reduced by antibodies or toxins. Surprisingly, these serial reductions in leukocyte lineages did not appreciably impair inducible resistance, but targeted disruption of TLR signaling in the lung epithelium resulted in complete abrogation of the protective effect. Isolated lung epithelial cells were also induced to kill pathogens in the absence of leukocytes. Proteomic and gene expression analyses of isolated epithelial cells and whole lungs revealed highly congruent antimicrobial responses. Taken together, these data indicate that lung epithelial cells are necessary and sufficient effectors of inducible resistance. These findings challenge conventional paradigms about the role of epithelia in antimicrobial defense and offer a novel potential intervention to protect patients with impaired leukocyte-mediated immunity from fatal pneumonias.


Journal of Leukocyte Biology | 2013

IL‐4Rα on CD4+ T cells plays a pathogenic role in respiratory syncytial virus reinfection in mice infected initially as neonates

Dahui You; Nico Marr; Jordy Saravia; Bishwas Shrestha; Greg I. Lee; Stuart E. Turvey; Frank Brombacher; DeˈBroski R. Herbert; Stephania A. Cormier

RSV is the major cause of severe bronchiolitis in infants, and severe bronchiolitis as a result of RSV is associated with subsequent asthma development. A biased Th2 immune response is thought to be responsible for neonatal RSV pathogenesis; however, molecular mechanisms remain elusive. Our data demonstrate, for the first time, that IL‐4Rα is up‐regulated in vitro on human CD4+ T cells from cord blood following RSV stimulation and in vivo on mouse pulmonary CD4+ T cells upon reinfection of mice, initially infected as neonates. Th cell‐specific deletion of Il4ra attenuated Th2 responses and abolished the immunopathophysiology upon reinfection, including airway hyper‐reactivity, eosinophilia, and mucus hyperproduction in mice infected initially as neonates. These findings support a pathogenic role for IL‐4Rα on Th cells following RSV reinfection of mice initially infected as neonates; more importantly, our data from human cells suggest that the same mechanism occurs in humans.


Mucosal Immunology | 2014

Early-life exposure to combustion-derived particulate matter causes pulmonary immunosuppression

Jordy Saravia; Dahui You; Paul Thevenot; Greg I. Lee; Bishwas Shrestha; Slawo Lomnicki; Stephania A. Cormier

Elevated levels of combustion-derived particulate matter (CDPM) are a risk factor for the development of lung diseases such as asthma. Studies have shown that CDPM exacerbates asthma, inducing acute lung dysfunction and inflammation; however, the impact of CDPM exposure on early immunological responses to allergens remains unclear. To determine the effects of early-life CDPM exposure on allergic asthma development in infants, we exposed infant mice to CDPM and then induced a mouse model of asthma using house dust mite (HDM) allergen. Mice exposed to CDPM+HDM failed to develop a typical asthma phenotype including airway hyper-responsiveness, T-helper type 2 (Th2) inflammation, Muc5ac expression, eosinophilia, and HDM-specific immunoglobulin (Ig) compared with HDM-exposed mice. Although HDM-specific IgE was attenuated, total IgE was twofold higher in CDPM+HDM mice compared with HDM mice. We further demonstrate that CDPM exposure during early life induced an immunosuppressive environment in the lung, concurrent with increases in tolerogenic dendritic cells and regulatory T cells, resulting in the suppression of Th2 responses. Despite having early immunosuppression, these mice develop severe allergic inflammation when challenged with allergen as adults. These findings demonstrate a mechanism whereby CDPM exposure modulates adaptive immunity, inducing specific antigen tolerance while amplifying total IgE, and leading to a predisposition to develop asthma upon rechallenge later in life.


Immunology and Cell Biology | 2015

Impaired gamma delta T cell-derived IL-17A and inflammasome activation during early respiratory syncytial virus infection in infants

Huaqiong Huang; Jordy Saravia; Dahui You; Aaron J. Shaw; Stephania A. Cormier

Respiratory syncytial virus (RSV) infection remains a significant global health burden disproportionately affecting infants and leading to long‐term lung disease. Interleukin (IL)‐17A has been shown to be involved in regulating viral and allergic lung inflammatory responses, which has led to a more recent interest in its role in RSV infection. Using a neonatal mouse model of RSV, we demonstrate that neonates fail to develop IL‐17A responses compared with adult mice; the main immediate IL‐17A contributor in adults were γδ T cells. Antibody neutralization of IL‐17A in adult mice caused increased lung inflammation and airway mucus from RSV, whereas exogenous IL‐17A administration to RSV‐infected neonates caused decreased inflammation but no change in airway mucus. We also observed a lack of pro‐inflammatory cytokine production (IL‐1β, IL‐6) from infected neonates. Using human cord blood mononuclear cells (CBMCs) and adult peripheral blood mononuclear cells (PBMCs), we compared inflammasome activation by direct retinoic acid‐inducible gene I agonism; CBMCs failed to induce pro‐inflammatory cytokines or IL‐17A+ γδ T cells compared with PBMCs. Our results indicate that RSV disease severity is in part mediated by a lack of inflammasome activation and IL‐17A production in neonates.


Particle and Fibre Toxicology | 2014

Exposure to combustion generated environmentally persistent free radicals enhances severity of influenza virus infection

Greg I. Lee; Jordy Saravia; Dahui You; Bishwas Shrestha; Sridhar Jaligama; Valerie Y Hebert; Tammy R. Dugas; Stephania A. Cormier

BackgroundExposures to elevated levels of particulate matter (PM) enhance severity of influenza virus infection in infants. The biological mechanism responsible for this phenomenon is unknown. The recent identification of environmentally persistent free radicals (EPFRs) associated with PM from a variety of combustion sources suggests its role in the enhancement of influenza disease severity.MethodsNeonatal mice (< seven days of age) were exposed to DCB230 (combustion derived PM with a chemisorbed EPFR), DCB50 (non-EPFR PM sample), or air for 30 minutes/day for seven consecutive days. Four days post-exposure, neonates were infected with influenza intranasally at 1.25 TCID50/neonate. Neonates were assessed for morbidity (% weight gain, peak pulmonary viral load, and viral clearance) and percent survival. Lungs were isolated and assessed for oxidative stress (8-isoprostanes and glutathione levels), adaptive immune response to influenza, and regulatory T cells (Tregs). The role of the EPFR was also assessed by use of transgenic mice expressing human superoxide dismutase 2.ResultsNeonates exposed to EPFRs had significantly enhanced morbidity and decreased survival following influenza infection. Increased oxidative stress was also observed in EPFR exposed neonates. This correlated with increased pulmonary Tregs and dampened protective T cell responses to influenza infection. Reduction of EPFR-induced oxidative stress attenuated these effects.ConclusionsNeonatal exposure to EPFR containing PM resulted in pulmonary oxidative stress and enhanced influenza disease severity. EPFR-induced oxidative stress resulted in increased presence of Tregs in the lungs and subsequent suppression of adaptive immune response to influenza.


Cell Reports | 2015

Acute Lung Injury Results from Innate Sensing of Viruses by an ER Stress Pathway

Eike R. Hrincius; Swantje Liedmann; David Finkelstein; Peter Vogel; Shane Gansebom; Amali E. Samarasinghe; Dahui You; Stephania A. Cormier; Jonathan A. McCullers

Summary Incursions of new pathogenic viruses into humans from animal reservoirs are occurring with alarming frequency. The molecular underpinnings of immune recognition, host responses, and pathogenesis in this setting are poorly understood. We studied pandemic influenza viruses to determine the mechanism by which increasing glycosylation during evolution of surface proteins facilitates diminished pathogenicity in adapted viruses. ER stress during infection with poorly glycosylated pandemic strains activated the unfolded protein response, leading to inflammation, acute lung injury, and mortality. Seasonal strains or viruses engineered to mimic adapted viruses displaying excess glycans on the hemagglutinin did not cause ER stress, allowing preservation of the lungs and survival. We propose that ER stress resulting from recognition of non-adapted viruses is utilized to discriminate “non-self” at the level of protein processing and to activate immune responses, with unintended consequences on pathogenesis. Understanding this mechanism should improve strategies for treating acute lung injury from zoonotic viral infections.

Collaboration


Dive into the Dahui You's collaboration.

Top Co-Authors

Avatar

Stephania A. Cormier

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Jordy Saravia

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Bishwas Shrestha

LSU Health Sciences Center New Orleans

View shared research outputs
Top Co-Authors

Avatar

Greg I. Lee

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

David Siefker

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sridhar Jaligama

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

David Becnel

Louisiana State University

View shared research outputs
Top Co-Authors

Avatar

John P. DeVincenzo

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Bindiya Bagga

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Cynthia Rovnaghi

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge