Network


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

Hotspot


Dive into the research topics where Bin Teng is active.

Publication


Featured researches published by Bin Teng.


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

Regulation and function of the two-pore-domain (K2P) potassium channel Trek-1 in alveolar epithelial cells

Andreas Schwingshackl; Bin Teng; Manik C. Ghosh; Alina West; Patrudu S. Makena; Vijay K. Gorantla; Scott E. Sinclair; Christopher M. Waters

Hyperoxia can lead to a myriad of deleterious effects in the lung including epithelial damage and diffuse inflammation. The specific mechanisms by which hyperoxia promotes these pathological changes are not completely understood. Activation of ion channels has been proposed as one of the mechanisms required for cell activation and mediator secretion. The two-pore-domain K(+) channel (K2P) Trek-1 has recently been described in lung epithelial cells, but its function remains elusive. In this study we hypothesized that hyperoxia affects expression of Trek-1 in alveolar epithelial cells and that Trek-1 is involved in regulation of cell proliferation and cytokine secretion. We found gene expression of several K2P channels in mouse alveolar epithelial cells (MLE-12), and expression of Trek-1 was significantly downregulated in cultured cells and lungs of mice exposed to hyperoxia. Similarly, proliferation cell nuclear antigen (PCNA) and Cyclin D1 expression were downregulated by exposure to hyperoxia. We developed an MLE-12 cell line deficient in Trek-1 expression using shRNA and found that Trek-1 deficiency resulted in increased cell proliferation and upregulation of PCNA but not Cyclin D1. Furthermore, IL-6 and regulated on activation normal T-expressed and presumably secreted (RANTES) secretion was decreased in Trek-1-deficient cells, whereas release of monocyte chemoattractant protein-1 was increased. Release of KC/IL-8 was not affected by Trek-1 deficiency. Overall, deficiency of Trek-1 had a more pronounced effect on mediator secretion than exposure to hyperoxia. This is the first report suggesting that the K(+) channel Trek-1 could be involved in regulation of alveolar epithelial cell proliferation and cytokine secretion, but a direct association with hyperoxia-induced changes in Trek-1 levels remains elusive.


PLOS ONE | 2014

The 2-pore domain potassium channel TREK-1 regulates stretch-induced detachment of alveolar epithelial cells.

Esra Roan; Christopher M. Waters; Bin Teng; Manik C. Ghosh; Andreas Schwingshackl

Acute Respiratory Distress Syndrome remains challenging partially because the underlying mechanisms are poorly understood. While inflammation and loss of barrier function are associated with disease progression, our understanding of the biophysical mechanisms associated with ventilator-associated lung injury is incomplete. In this line of thinking, we recently showed that changes in the F-actin content and deformability of AECs lead to cell detachment with mechanical stretch. Elsewhere, we discovered that cytokine secretion and proliferation were regulated in part by the stretch-activated 2-pore domain K+ (K2P) channel TREK-1 in alveolar epithelial cells (AECs). As such, the aim of the current study was to determine whether TREK-1 regulated the mechanobiology of AECs through cytoskeletal remodeling and cell detachment. Using a TREK-1-deficient human AEC line (A549), we examined the cytoskeleton by confocal microscopy and quantified differences in the F-actin content. We used nano-indentation with an atomic force microscope to measure the deformability of cells and detachment assays to quantify the level of injury in our monolayers. We found a decrease in F-actin and an increase in deformability in TREK-1 deficient cells compared to control cells. Although total vinculin and focal adhesion kinase (FAK) levels remained unchanged, focal adhesions appeared to be less prominent and phosphorylation of FAK at the Tyr925 residue was greater in TREK-1 deficient cells. TREK-1 deficient cells have less F-actin and are more deformable making them more resistant to stretch-induced injury.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2013

Regulation of interleukin-6 secretion by the two-pore-domain potassium channel Trek-1 in alveolar epithelial cells.

Andreas Schwingshackl; Bin Teng; Manik C. Ghosh; Keng Gat Lim; Gabor Tigyi; Damodaran Narayanan; Jonathan H. Jaggar; Christopher M. Waters

We recently proposed a role for the two-pore-domain K(+) (K2P) channel Trek-1 in the regulation of cytokine release from mouse alveolar epithelial cells (AECs) by demonstrating decreased interleukin-6 (IL-6) secretion from Trek-1-deficient cells, but the underlying mechanisms remained unknown. This study was designed to investigate the mechanisms by which Trek-1 decreases IL-6 secretion. We hypothesized that Trek-1 regulates tumor necrosis factor-α (TNF-α)-induced IL-6 release via NF-κB-, p38-, and PKC-dependent pathways. We found that Trek-1 deficiency decreased IL-6 secretion from mouse and human AECs at both transcriptional and translational levels. While NF-κB/p65 phosphorylation was unchanged, p38 phosphorylation was decreased in Trek-1-deficient cells, and pharmacological inhibition of p38 decreased IL-6 secretion in control but not Trek-1-deficient cells. Similarly, pharmacological inhibition of PKC also decreased IL-6 release, and we found decreased phosphorylation of the isoforms PKC/PKDμ (Ser(744/748)), PKCθ, PKCδ, PKCα/βII, and PKCζ/λ, but not PKC/PKDμ (Ser(916)) in Trek-1-deficient AECs. Phosphorylation of PKCθ, a Ca(2+)-independent isoform, was intact in control cells but impaired in Trek-1-deficient cells. Furthermore, TNF-α did not elevate the intracellular Ca(2+) concentration in control or Trek-1-deficient cells, and removal of extracellular Ca(2+) did not impair IL-6 release. In summary, we report the expression of Trek-1 in human AECs and propose that Trek-1 deficiency may alter both IL-6 translation and transcription in AECs without affecting Ca(2+) signaling. The results of this study identify Trek-1 as a new potential target for the development of novel treatment strategies against acute lung injury.


Critical Care Medicine | 2013

Deficiency of the two-pore-domain potassium channel TREK-1 promotes hyperoxia-induced lung injury.

Andreas Schwingshackl; Bin Teng; Patrudu S. Makena; Manik C. Ghosh; Scott E. Sinclair; Charlean L. Luellen; Louisa Balasz; Cynthia Rovnaghi; Robert M. Bryan; Eric E. Lloyd; Elizabeth A. Fitzpatrick; Jordy Saravia; Stephania A. Cormier; Christopher M. Waters

Objectives: We previously reported the expression of the two-pore-domain K+ channel TREK-1 in lung epithelial cells and proposed a role for this channel in the regulation of alveolar epithelial cytokine secretion. In this study, we focused on investigating the role of TREK-1 in vivo in the development of hyperoxia-induced lung injury. Design: Laboratory animal experiments. Setting: University research laboratory. Subjects: Wild-type and TREK-1-deficient mice. Interventions: Mice were anesthetized and exposed to 1) room air, no mechanical ventilation, 2) 95% hyperoxia for 24 hours, and 3) 95% hyperoxia for 24 hours followed by mechanical ventilation for 4 hours. Measurements and Main Results: Hyperoxia exposure accentuated lung injury in TREK-1-deficient mice but not controls, resulting in increase in lung injury scores, bronchoalveolar lavage fluid cell numbers, and cellular apoptosis and a decrease in quasi-static lung compliance. Exposure to a combination of hyperoxia and injurious mechanical ventilation resulted in further morphological lung damage and increased lung injury scores and bronchoalveolar lavage fluid cell numbers in control but not TREK-1-deficient mice. At baseline and after hyperoxia exposure, bronchoalveolar lavage cytokine levels were unchanged in TREK-1-deficient mice compared with controls. Exposure to hyperoxia and mechanical ventilation resulted in an increase in bronchoalveolar lavage interleukin-6, monocyte chemotactic protein-1, and tumor necrosis factor-&agr; levels in both mouse types, but the increase in interleukin-6 and monocyte chemotactic protein-1 levels was less prominent in TREK-1-deficient mice than in controls. Lung tissue macrophage inflammatory protein-2, keratinocyte-derived cytokine, and interleukin-1&bgr; gene expression was not altered by hyperoxia in TREK-1-deficient mice compared with controls. Furthermore, we show for the first time TREK-1 expression on alveolar macrophages and unimpaired tumor necrosis factor-&agr; secretion from TREK-1-deficient macrophages. Conclusions: TREK-1 deficiency resulted in increased sensitivity of lungs to hyperoxia, but this effect is less prominent if overwhelming injury is induced by the combination of hyperoxia and injurious mechanical ventilation. TREK-1 may constitute a new potential target for the development of novel treatment strategies against hyperoxia-induced lung injury.


Cytokine | 2016

Regulation of inflammatory biomarkers by intravenous methylprednisolone in pediatric ARDS patients: Results from a double-blind, placebo-controlled randomized pilot trial.

Andreas Schwingshackl; Dai Kimura; Cynthia Rovnaghi; Jordy Saravia; Stephania A. Cormier; Bin Teng; Alina West; Umberto G. Meduri; K.J.S. Anand

OBJECTIVE A double-blind, randomized controlled trial showed that low-dose glucocorticoid therapy in pediatric ARDS patients is feasible and may improve both ventilation and oxygenation indices in these patients. However, the molecular mechanisms underlying potential changes in outcomes remain unclear. Based on these clinical findings, this study was designed to examine the effects of intravenous methylprednisolone on circulating inflammatory biomarkers in pediatric ARDS patients. DESIGN Double-blind, placebo-controlled randomized trial with blood collection on study entry and day 7. SETTING Tertiary care childrens hospital. PATIENTS Children (0-18years) with ARDS undergoing mechanical ventilation. INTERVENTIONS 35 children were randomized within 72h of mechanical ventilation. The glucocorticoid group received methylprednisolone 2mg/kg loading dose followed by 1mg/kg/day continuous infusion from days 1 to 7. Both groups were ventilated following the ARDSnet recommendations. WBC and differential cell counts, plasma cytokines and CRP levels, and coagulation parameters were analyzed on days 0 and 7. RESULTS At study entry, the placebo group had higher IL-15 and basophil levels. On day 7, in comparison to study entry, the placebo group had lower IL-1α, IFN-γ and IL-10 levels. The glucocorticoid group had lower INF-α, IL-6, IL-10, MCP-1, G-CSF and GM-CSF levels, and higher IL-17α levels on day 7 in comparison to study entry. Total and differential cell counts remained unchanged within the placebo group between days 0 and 7, whereas in the glucocorticoid group total WBC and platelets counts were increased on day 7. Pearsons correlation studies within the placebo and glucocorticoid groups revealed positive and negative correlations between cytokine levels, cell counts, coagulation parameters and relevant clinical parameters of disease severity identified in our previous study. Multiple regression models identified several cytokines as predictors for alterations in clinical parameters of disease severity. CONCLUSION This pilot study shows the feasibility of simultaneously measuring multiple inflammatory cytokines, cell counts and coagulation parameters in pediatric ARDS patients. We report statistical models that may be useful for future, larger trials to predict ARDS severity and outcomes.


PLOS ONE | 2015

TREK-1 Regulates Cytokine Secretion from Cultured Human Alveolar Epithelial Cells Independently of Cytoskeletal Rearrangements

Andreas Schwingshackl; Esra Roan; Bin Teng; Christopher M. Waters

Background TREK-1 deficient alveolar epithelial cells (AECs) secrete less IL-6, more MCP-1, and contain less F-actin. Whether these alterations in cytokine secretion and F-actin content are related remains unknown. We now hypothesized that cytokine secretion from TREK-1-deficient AECs was regulated by cytoskeletal rearrangements. Methods We determined F-actin and α-tubulin contents of control, TREK-1-deficient and TREK-1-overexpressing human A549 cells by confocal microscopy and western blotting, and measured IL-6 and MCP-1 levels using real-time PCR and ELISA. Results Cytochalasin D decreased the F-actin content of control cells. Jasplakinolide increased the F-actin content of TREK-1 deficient cells, similar to the effect of TREK-1 overexpression in control cells. Treatment of control and TREK-1 deficient cells with TNF-α, a strong stimulus for IL-6 and MCP-1 secretion, had no effect on F-actin structures. The combination of TNF-α+cytochalasin D or TNF-α+jasplakinolide had no additional effect on the F-actin content or architecture when compared to cytochalasin D or jasplakinolide alone. Although TREK-1 deficient AECs contained less F-actin at baseline, quantified biochemically, they contained more α-tubulin. Exposure to nocodazole disrupted α-tubulin filaments in control and TREK-1 deficient cells, but left the overall amount of α-tubulin unchanged. Although TNF-α had no effect on the F-actin or α-tubulin contents, it increased IL-6 and MCP-1 production and secretion from control and TREK-1 deficient cells. IL-6 and MCP-1 secretions from control and TREK-1 deficient cells after TNF-α+jasplakinolide or TNF-α+nocodazole treatment was similar to the effect of TNF-α alone. Interestingly, cytochalasin D decreased TNF-α-induced IL-6 but not MCP-1 secretion from control but not TREK-1 deficient cells. Conclusion Although cytochalasin D, jasplakinolide and nocodazole altered the F-actin and α-tubulin structures of control and TREK-1 deficient AEC, the changes in cytokine secretion from TREK-1 deficient cells cannot be explained by cytoskeletal rearrangements in these cells.


Scientific Reports | 2017

A heteromeric molecular complex regulates the migration of lung alveolar epithelial cells during wound healing

Manik C. Ghosh; Patrudu S. Makena; Joseph Kennedy; Bin Teng; Charlean L. Luellen; Scott E. Sinclair; Christopher M. Waters

Alveolar type II epithelial cells (ATII) are instrumental in early wound healing in response to lung injury, restoring epithelial integrity through spreading and migration. We previously reported in separate studies that focal adhesion kinase-1 (FAK) and the chemokine receptor CXCR4 promote epithelial repair mechanisms. However, potential interactions between these two pathways were not previously considered. In the present study, we found that wounding of rat ATII cells promoted increased association between FAK and CXCR4. In addition, protein phosphatase-5 (PP5) increased its association with this heteromeric complex, while apoptosis signal regulating kinase-1 (ASK1) dissociated from the complex. Cell migration following wounding was decreased when PP5 expression was decreased using shRNA, but migration was increased in ATII cells isolated from ASK1 knockout mice. Interactions between FAK and CXCR4 were increased upon depletion of ASK1 using shRNA in MLE-12 cells, but unaffected when PP5 was depleted. Furthermore, we found that wounded rat ATII cells exhibited decreased ASK1 phosphorylation at Serine-966, decreased serine phosphorylation of FAK, and decreased association of phosphorylated ASK1 with FAK. These changes in phosphorylation were dependent upon expression of PP5. These results demonstrate a unique molecular complex comprising CXCR4, FAK, ASK1, and PP5 in ATII cells during wound healing.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2017

Hyperoxia treatment of TREK-1/TREK-2/TRAAK-deficient mice is associated with a reduction in surfactant proteins

Andreas Schwingshackl; Benjamin Lopez; Bin Teng; Charlean L. Luellen; Florian Lesage; John A. Belperio; Riccardo Olcese; Christopher M. Waters

We previously proposed a role for the two-pore domain potassium (K2P) channel TREK-1 in hyperoxia (HO)-induced lung injury. To determine whether redundancy among the three TREK isoforms (TREK-1, TREK-2, and TRAAK) could protect from HO-induced injury, we now examined the effect of deletion of all three TREK isoforms in a clinically relevant scenario of prolonged HO exposure and mechanical ventilation (MV). We exposed WT and TREK-1/TREK-2/TRAAK-deficient [triple knockout (KO)] mice to either room air, 72-h HO, MV [high and low tidal volume (TV)], or a combination of HO + MV and measured quasistatic lung compliance, bronchoalveolar lavage (BAL) protein concentration, histologic lung injury scores (LIS), cellular apoptosis, and cytokine levels. We determined surfactant gene and protein expression and attempted to prevent HO-induced lung injury by prophylactically administering an exogenous surfactant (Curosurf). HO treatment increased lung injury in triple KO but not WT mice, including an elevated LIS, BAL protein concentration, and markers of apoptosis, decreased lung compliance, and a more proinflammatory cytokine phenotype. MV alone had no effect on lung injury markers. Exposure to HO + MV (low TV) further decreased lung compliance in triple KO but not WT mice, and HO + MV (high TV) was lethal for triple KO mice. In triple KO mice, the HO-induced lung injury was associated with decreased surfactant protein (SP) A and SPC but not SPB and SPD expression. However, these changes could not be explained by alterations in the transcription factors nuclear factor-1 (NF-1), NKX2.1/thyroid transcription factor-1 (TTF-1) or c-jun, or lamellar body levels. Prophylactic Curosurf administration did not improve lung injury scores or compliance in triple KO mice.


Critical Care Medicine | 2016

1057: THE K+ CHANNEL TREK-1 REGULATES CYTOKINE SECRETION FROM ALVEOLAR EPITHELIAL CELLS INDEPENDENTLY OF K

Andreas Schwingshackl; Bin Teng; Christopher M. Waters

Learning Objectives: Based on our previous data, the 2-pore domain potassium (K2P) channel TREK-1 appears to play an important role in the development of lung injury using in vitro and in vivo models of Acute Lung Injury/ARDS. In vitro, TREK-1 deficiency altered TNF-α-induced IL-6 and monocyte chemotactic protein-1 (MCP-1) but not IL-8 secretion from alveolar epithelial cells (AECs). In an in vivo model of ventilator-induced ARDS we found that TREK-1 deficiency led to increased lung damage but decreased broncho-alveolar lavage cytokine levels. Unfortunately, the mechanisms underlying TREK-1 regulation of inflammatory cytokine secretion remain poorly understood. Aim: In this study we determined whether the changes in cytokine secretion from TREK-1 deficient AECs are due to the absence of TREK-1 protein or rather the lack of TREK1-mediated potassium currents. Methods: We stimulated control and TREK-1 deficient human AECs with TNF-α and measured IL-6 and MCP-1 release by ELISA in the presence and absence of the specific TREK-1 blocker Spadin (0.5mM). Results: TREK-1 deficient AECs released less IL-6 and more MCP-1 protein upon TNF-α stimulation but these cytokine alterations were unaffected by Spadin. To determine whether IL-6 and MCP-1 secretion occurred independently of potassium currents in general, we raised the extracellular potassium concentration to 100mM thus minimizing the electrochemical driving force for potassium ions to exit the cell. In contrast to TREK-1 inhibition, we found that TNF-α-induced IL-6 and MCP-1 secretion was significantly impaired in the absence of a physiologic potassium gradient.Conclusions: In summary, TREK-1 protein, but not TREK-1 mediated potassium current, is required for IL-6 and MCP-1 secretion from AECs, although other potassium conductances are important for cytokine secretion. These data suggest that TREK-1 may alter cytokine secretion from AECs by functioning as a regulatory molecule rather than a potassium channel. 1058


American Journal of Translational Research | 2013

Regulation of Monocyte Chemotactic Protein-1 secretion by the Two-Pore-Domain Potassium (K2P) channel TREK-1 in human alveolar epithelial cells.

Andreas Schwingshackl; Bin Teng; Manik C. Ghosh; Christopher M. Waters

Collaboration


Dive into the Bin Teng's collaboration.

Top Co-Authors

Avatar

Andreas Schwingshackl

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Christopher M. Waters

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Manik C. Ghosh

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Charlean L. Luellen

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Patrudu S. Makena

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Alina West

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Cynthia Rovnaghi

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Scott E. Sinclair

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Dai Kimura

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge