Network


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

Hotspot


Dive into the research topics where Li-Juan Yao is active.

Publication


Featured researches published by Li-Juan Yao.


Experimental Lung Research | 1999

An examination of the different variables affecting surfactant aggregate conversion in vitro

Ruud A. W. Veldhuizen; Li-Juan Yao; James F. Lewis

Pulmonary surfactant exists in 2 major subtypes, the freshly secreted, surface-active large surfactant aggregates (LA) and their metabolic product, the less surface active, small aggregates (SA). Conversion of LA into SA can be studied using an in vitro technique, surface area cycling, which involves the rotation of a suspension of LA end-over-end so that the surface area of the liquid changes twice each cycle. In order to further elucidate the mechanisms involved in aggregate conversion, we have examined the effects of time, temperature, change in surface area, cycling speed, surfactant concentration, and albumin on aggregate conversion in vitro. Three different surfactant preparations were used; rabbit LA, sheep LA, and an exogenous surfactant preparation, bovine lipid extract surfactant (BLES). Based on our results that showed that these variables affected aggregate conversion, we concluded that the adsorption of surfactant at the changing air-liquid interface was an important step in aggregate conversion. However, the results also indicated that aggregate conversion was not solely due to the adsorption characteristics of the surfactant. Other surfactant properties, such as the activity of serine protease or film stability at the air-liquid interface, may also be important in aggregate conversion.


Experimental Lung Research | 2001

SURFACTANT TREATMENT FOR VENTILATION-INDUCED LUNG INJURY IN RATS: EFFECTS ON LUNG COMPLIANCE AND CYTOKINES

Blayne Welk; Jaret L. Malloy; Mariamma Joseph; Li-Juan Yao; Ruud A. W. Veldhuizen

The objective of this study was to determine if exogenous surfactant therapy could prevent the harmful effects ofventilation at high tidal volumes without positive end-expiratory pressure (PEEP). Rats were randomized to either a nontreated controlgroup (8mL/kg 4cm H 2 O PEEP), a nontreated injuriously ventilated group (20mL/kg 0cm H 2 O PEEP)or a treatment group of either 50 mg/kg, 50 mg/kg + 5% surfactant-associated protein A, or 100 mg/kg exogenous surfactant followed by injurious ventilation. Isolated lungs from animals in all 5 groups were ventilated in a humidified box at 37° C for 2 hours. Pressure-volume curves and light microscopy showed that surfactant treatment reduced the ventilation-induced lung injury (VILI). Inflammatory cytokines (tumor necrosis factor- α [TNF α ], interleukin [IL]-1 β, and IL-6) in the lavage were significantly higher in injuriously ventilated lungs compared to the control group. However, the 3 treatment groups had cytokine concentrations that were similar to the injuriously ventilated group. Weconclude that surfactant treatment is beneficial in preventing VILI; however, it does not prevent the release of inflammatory cytokines during mechanical ventilation.


PLOS ONE | 2013

Human Alveolar Epithelial Cells Attenuate Pulmonary Microvascular Endothelial Cell Permeability under Septic Conditions

Lefeng Wang; Ravi Taneja; Wei Wang; Li-Juan Yao; Ruud A. W. Veldhuizen; Sean E. Gill; Dalilah Fortin; Richard Inculet; Richard A. Malthaner; Sanjay Mehta

Acute lung injury (ALI) and its most severe form, acute respiratory distress syndrome (ARDS), are characterised by high-protein pulmonary edema and severe hypoxaemic respiratory failure due to increased permeability of pulmonary microvascular endothelial cells (PMVEC). Alveolar epithelial cells (AEC) contribute importantly to normal alveolar function, and AEC dysfunction in ALI/ARDS is associated with worse outcomes. We hypothesized that AEC can modulate human PMVEC barrier function, and investigated the effects of AEC presence on human PMVEC barrier under septic conditions in vitro. PMVEC isolated from human lung were treated in vitro with septic stimulation (lipopolysaccharide [LPS], a mixture of clinically-relevant cytokines [cytomix], or plasma from patients with severe sepsis), and the trans-PMVEC leak of Evans Blue dye-labeled albumin assessed. PMVEC septic responses were compared in the presence/absence of co-cultured A549 epithelial cell line or primary human AEC. Septic stimulation with LPS, cytomix, or septic plasma induced marked PMVEC hyper-permeability (10.2±1.8, 8.9±2.2, and 3.7±0.2 fold-increase vs. control, respectively, p<0.01 for all). The presence of A549 cells or primary human AEC in a non-contact co-culture model attenuated septic PMVEC hyper-permeability by 39±4% to 100±3%, depending on the septic stimulation (p<0.05). Septic PMVEC hyper-permeability was also attenuated following treatment with culture medium conditioned by previous incubation with either naïve or cytomix-treated A549 cells (p<0.05), and this protective effect of A549 cell-conditioned medium was both heat-stable and transferable following lipid extraction. Cytomix-stimulated PMN-dependent PMVEC hyper-permeability and trans-PMVEC PMN migration were also inhibited in the presence of A549 cells or A549 cell-conditioned medium (p<0.05). Human AEC appear to protect human PMVEC barrier function under septic conditions in vitro, through release of a soluble mediator(s), which are at least partly lipid in nature. This study suggests a scientific and potential clinical therapeutic importance of epithelial-endothelial cross talk in maintaining alveolar integrity in ALI/ARDS.


Experimental Lung Research | 2009

THE EFFECTS OF HYPEROXIA EXPOSURE ON LUNG FUNCTION AND PULMONARY SURFACTANT IN A RAT MODEL OF ACUTE LUNG INJURY

Patrick W. Pace; Li-Juan Yao; John X. Wilson; Fred Possmayer; Ruud A. W. Veldhuizen; James F. Lewis

The objective of this study was to determine if prolonged hyperoxia exposure would deplete antioxidants, resulting in excessive oxidative stress that would lead to oxidation of pulmonary surfactant and contribute to lung dysfunction. Rats were exposed to either hyperoxic (> 95% O2) or normoxic (21% O2) oxygen concentrations for 48 or 60 hours. Pulmonary compliance, inflammatory cells, and total protein levels were measured as indicators of lung injury. Bronchoalveolar lavage (BAL) samples were analyzed for surfactant composition, antioxidant content, and markers of oxidative stress. Antioxidants were also measured in lung tissue and plasma samples. Hyperoxia exposure for 60 hours resulted in increased protein and inflammatory cells in BAL, and lower pulmonary compliance, compared to all other groups. Total surfactant and surfactant large aggregates were increased following 48 hours of hyperoxia exposure, with a further increase following 60 hours. Animals exposed to 60 hours of hyperoxia also demonstrated lower ascorbate levels in lung tissue, increased lipid peroxides in BAL, and increased oxidation of phosphatidylglycerol species in surfactant. This study demonstrates that the balance of oxidant/antioxidant components is disrupted within the lung during periods of hyperoxia, and that although surfactant lipids may be susceptible to oxidative damage, they do not likely represent a major mechanism for the lung dysfunction observed.


Experimental Lung Research | 2009

ELEVATED ENDOGENOUS SURFACTANT REDUCES INFLAMMATION IN AN ACUTE LUNG INJURY MODEL

Melissa G. Walker; Jenna M. Tessolini; Lynda McCaig; Li-Juan Yao; James F. Lewis; Ruud A. W. Veldhuizen

Acute lung injury (ALI) is associated with severe pulmonary inflammation and alterations to surfactant, and often results in overwhelming systemic inflammation, leading to multiple organ failure. The objective of this study was to determine the effect of increased endogenous surfactant pools on pulmonary and systemic inflammation in a model of lipopolysaccharide (LPS)-induced ALI. Mice received an instillation of liposome-encapsulated (i) dichloromethylene diphosphonic acid (DMDP) to increase surfactant pools via depletion of alveolar macrophages, or (ii) phosphate-buffered saline (PBS). Seven days after instillation, mice received an intranasal administration of LPS or saline. Following a 4-hour recovery period, mice were sacrificed and their lungs were isolated, mechanically ventilated, and perfused with 8 mL of recirculated perfusate through the pulmonary circulation for 2 hours. Perfusate and lavage fluid were collected for analysis of inflammatory mediators. Lavage analysis revealed a 5-fold increase in surfactant pools in DMDP-treated mice compared to PBS-treated controls. Lavage and perfusate analyses showed significant decreases in the concentrations of interleukin (IL)-6, tumor necrosis factor (TNF)-α, macrophage inflammatory protein (MIP)-1α, and IL-1β cytokines in DMDP-LPS mice compared to PBS-LPS controls. Elevated endogenous surfactant pools are protective against both LPS- and mechanical ventilation–induced inflammation, in addition to inflammation associated with the combination of these two insults.


Critical Care Medicine | 2008

The effects of long-term conventional mechanical ventilation on the lungs of adult rats.

Timothy C. Bailey; Adam A. Maruscak; Erica L. Martin; Amy Forbes; Anne Petersen; Lynda McCaig; Li-Juan Yao; James F. Lewis; Ruud A. W. Veldhuizen

Background:Ventilation-induced lung injury is often studied in animal models by using ventilation strategies with high-tidal volumes and high-oxygen concentration over a relatively short period of time. The injury induced by these ventilation strategies includes alterations to the surfactant system and up-regulation of inflammatory markers. Whether these responses to ventilation occur with more clinically relevant ventilation strategies is not known. Objective:To assess how healthy adult rats respond to 24 hrs of conventional mechanical ventilation with respect to lung physiology, markers of inflammation, and alterations to pulmonary surfactant, and how this is affected by the oxygen concentration. Interventions:Adult rats were mechanically ventilated for 24 hrs with a tidal volume of 8 mL/kg, 5 cm H2O positive end-expiratory pressure, at 60 breaths/min with either 21% or 100% oxygen. Animals were monitored for blood oxygenation and other physiologic parameters. After ventilation, lungs were lavaged and analyzed for inflammatory markers and pulmonary surfactant. These outcomes were compared with measurements obtained from spontaneously breathing rats exposed to either 21% or 100% oxygen for 24 hrs. Main Results:Twenty-four hours of ventilation did not result in significant changes in blood oxygenation. Inflammatory markers, such as interleukin-6 concentration and the number of neutrophils in the lavage, were increased in ventilated animals compared with the nonventilated controls, regardless of the level of inspired oxygen. The amount of active surfactant was increased after ventilation; however, the surface activity of this material was impaired as compared with controls. Conclusion:Prolonged mechanical ventilation of health lungs with a physiologically benign strategy can contribute to the inflammatory response and cause alterations to pulmonary surfactant.


Respiration | 2011

The Effect of Tidal Volume on Systemic Inflammation in Acid-Induced Lung Injury

Melissa G. Walker; Li-Juan Yao; Eric K. Patterson; Mariamma Joseph; Gediminas Cepinskas; Ruud A. W. Veldhuizen; James F. Lewis; Cory M. Yamashita

Background: Overwhelming systemic inflammation has been implicated in the progression of acute lung injury (ALI) leading to multiple organ failure (MOF) and death. Previous studies suggest that mechanical ventilation (MV) may be a key mediator of MOF through an upregulation of the systemic inflammatory response. Objectives: It was the aim of this study to investigate mechanisms whereby mechanical stress induced by different tidal volumes may contribute to the development of systemic inflammation and maladaptive peripheral organ responses in the setting of ALI. Methods: An acid aspiration model of ALI was employed in 129X1/SVJ mice through an intratracheal administration of hydrochloric acid followed by MV employing either a low (5 ml/kg) or high (12.5 ml/kg) tidal volume ventilation for 120 min. The isolated perfused mouse lung setup was used to assess the specific contribution of the lung to systemic inflammation during MV. Furthermore, lung perfusate collected over the course of MV was used to assess the effects of lung-derived mediators on activation (expression of a proadhesive phenotype) of liver endothelial cells. Results: High tidal volume MV of acid-injured lungs resulted in greater physiologic and histological indices of lung injury compared to control groups. Additionally, there was an immediate and significant release of multiple inflammatory mediators from the lung into the systemic circulation which resulted in greater levels of mRNA adhesion molecule expression in liver endothelial cells in vitro. Conclusions: This study suggests that MV, specifically tidal volume strategy, influences the development of MOF through an upregulation of lung-derived systemic inflammation resulting in maladaptive cellular changes in peripheral organs.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2012

Surfactant protein B inhibits secretory phospholipase A2 hydrolysis of surfactant phospholipids

R. Duncan Hite; Bonnie L. Grier; B. Moseley Waite; Ruud A. W. Veldhuizen; Fred Possmayer; Li-Juan Yao; Michael C. Seeds

Hydrolysis of surfactant phospholipids (PL) by secretory phospholipases A(2) (sPLA(2)) contributes to surfactant damage in inflammatory airway diseases such as acute lung injury/acute respiratory distress syndrome. We and others have reported that each sPLA(2) exhibits specificity in hydrolyzing different PLs in pulmonary surfactant and that the presence of hydrophilic surfactant protein A (SP-A) alters sPLA(2)-mediated hydrolysis. This report tests the hypothesis that hydrophobic SP-B also inhibits sPLA(2)-mediated surfactant hydrolysis. Three surfactant preparations were used containing varied amounts of SP-B and radiolabeled tracers of phosphatidylcholine (PC) or phosphatidylglycerol (PG): 1) washed ovine surfactant (OS) (pre- and postorganic extraction) compared with Survanta (protein poor), 2) Survanta supplemented with purified bovine SP-B (1-5%, wt/wt), and 3) a mixture of dipalmitoylphosphatidylcholine (DPPC), 1-palmitoyl-2-oleoyl-phosphatidylcholine (POPC), and 1-palmitoyl-2-oleoyl-phosphatidylglycerol (POPG) (DPPC:POPC:POPG, 40:40:20) prepared as vesicles and monomolecular films in the presence or absence of SP-B. Hydrolysis of PG and PC by Group IB sPLA(2) (PLA2G1A) was significantly lower in the extracted OS, which contains SP-B, compared with Survanta (P = 0.005), which is SP-B poor. Hydrolysis of PG and PC in nonextracted OS, which contains all SPs, was lower than both Survanta and extracted OS. When Survanta was supplemented with 1% SP-B, PG and PC hydrolysis by PLA2G1B was significantly lower (P < 0.001) than in Survanta alone. When supplemented into pure lipid vesicles and monomolecular films composed of PG and PC mixtures, SP-B also inhibited hydrolysis by both PLA2G1B and Group IIA sPLA2 (PLA2G2A). In films, PLA2G1B hydrolyzed surfactant PL monolayers at surface pressures ≤30 mN/m (P < 0.01), and SP-B lowered the surface pressure range at which hydrolysis can occur. These results suggest the hydrophobic SP, SP-B, protects alveolar surfactant PL from hydrolysis mediated by multiple sPLA(2) in both vesicles (alveolar subphase) and monomolecular films (air-liquid interface).


Respiration | 2014

Apolipoprotein E-Deficient Mice Are Susceptible to the Development of Acute Lung Injury

Cory M. Yamashita; Michael B. Fessler; Lakshman Vasanthamohan; Joanne Lac; Jennifer H. Madenspacher; Lynda McCaig; Li-Juan Yao; Lefeng Wang; Valeria Puntorieri; Sanjay Mehta; Jim Lewis; Ruud A. W. Veldhuizen

Background: Apolipoprotein E (apoE) has been shown to play a pivotal role in the development of cardiovascular disease, attributable to its function in lipid trafficking and immune modulating properties; however, its role in modulating inflammation in the setting of acute lung injury (ALI) is unknown. Objective: To determine whether apoE-deficient mice (apoE-/-) are more susceptible to ALI compared to wild-type (WT) animals. Methods: Two independent models of ALI were employed. Firstly, WT and apoE-/- mice were randomized to acid aspiration (50 μl of 0.1 N hydrochloric acid) followed by 4 h of mechanical ventilation. Secondly, WT and apoE-/- mice were randomized to 72 h of hyperoxia exposure or room air. Thereafter, the intrinsic responses of WT and apoE-/- mice were assessed using the isolated perfused mouse lung (IPML) setup. Finally, based on elevated levels of oxidized low-density lipoprotein (oxLDL) in apoE-/-, the effect of oxLDL on lung endothelial permeability and inflammation was assessed. Results: In both in vivo models, apoE-/- mice demonstrated greater increases in lung lavage protein levels, neutrophil counts, and cytokine expression (p < 0.05) compared to WT mice. Experiments utilizing the IPML setup demonstrated no differences in intrinsic lung responses to injury between apoE-/- and WT mice, suggesting the presence of a circulating factor as being responsible for the in vivo observations. Finally, the exposure of lung endothelial cells to oxLDL resulted in increased monolayer permeability and IL-6 release compared to native (nonoxidized) LDL. Conclusions: Our findings demonstrate a susceptibility of apoE-/- animals to ALI that may occur, in part, due to elevated levels of oxLDL.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2008

Protective effects of elevated endogenous surfactant pools to injurious mechanical ventilation.

Cory M. Yamashita; Amy Forbes; Jenna M. Tessolini; Li-Juan Yao; James F. Lewis; Ruud A. W. Veldhuizen

Depletion of alveolar macrophages (AM) leads to an increase in endogenous surfactant that lasts several days beyond the repletion of AM. Furthermore, impairment to the endogenous pulmonary surfactant system contributes to ventilation-induced lung injury. The objective of the current study was to determine whether increased endogenous surfactant pools induced via AM depletion was protective against ventilation-induced lung injury. Adult rats were intratracheally instilled with either control or dichloromethylene diphosphonic acid (DMDP) containing liposomes to deplete AMs and thereby increase endogenous surfactant pools. Either 3 or 7 days following instillation, rats were exposed to 2 h of injurious ventilation using either an ex vivo or in vivo ventilation protocol and were compared with nonventilated controls. The measured outcomes were oxygenation, lung compliance, lavage protein, and inflammatory cytokine concentrations. Compared with controls, the DMDP-treated animals had significantly reduced AM numbers and increased surfactant pools 3 days after instillation. Seven days after instillation, AM numbers had returned to normal, but surfactant pools were still elevated. DMDP-treated animals at both time points exhibited protection against ventilation-induced lung injury, which included superior physiological parameters, lower protein leakage, and lower inflammatory mediator release into the air space, compared with animals not receiving DMDP. It is concluded that DMDP-liposome administration protects against ventilation-induced lung injury. This effect appears to be due to the presence of elevated endogenous surfactant pools.

Collaboration


Dive into the Li-Juan Yao's collaboration.

Top Co-Authors

Avatar

Ruud A. W. Veldhuizen

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

James F. Lewis

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lynda McCaig

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Cory M. Yamashita

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Fred Possmayer

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jim Lewis

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Mariamma Joseph

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Yushi Ito

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Melissa G. Walker

Lawson Health Research Institute

View shared research outputs
Top Co-Authors

Avatar

Valeria Puntorieri

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge