1
|
Godbole NM, Chowdhury AA, Chataut N, Awasthi S. Tight Junctions, the Epithelial Barrier, and Toll-like Receptor-4 During Lung Injury. Inflammation 2022; 45:2142-2162. [PMID: 35779195 PMCID: PMC9649847 DOI: 10.1007/s10753-022-01708-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/31/2022] [Accepted: 06/13/2022] [Indexed: 11/25/2022]
Abstract
Lung epithelium is constantly exposed to the environment and is critically important for the orchestration of initial responses to infectious organisms, toxins, and allergic stimuli, and maintenance of normal gaseous exchange and pulmonary function. The integrity of lung epithelium, fluid balance, and transport of molecules is dictated by the tight junctions (TJs). The TJs are formed between adjacent cells. We have focused on the topic of the TJ structure and function in lung epithelial cells. This review includes a summary of the last twenty years of literature reports published on the disrupted TJs and epithelial barrier in various lung conditions and expression and regulation of specific TJ proteins against pathogenic stimuli. We discuss the molecular signaling and crosstalk among signaling pathways that control the TJ structure and function. The Toll-like receptor-4 (TLR4) recognizes the pathogen- and damage-associated molecular patterns released during lung injury and inflammation and coordinates cellular responses. The molecular aspects of TLR4 signaling in the context of TJs or the epithelial barrier are not fully known. We describe the current knowledge and possible networking of the TLR4-signaling with cellular and molecular mechanisms of TJs, lung epithelial barrier function, and resistance to treatment strategies.
Collapse
Affiliation(s)
- Nachiket M Godbole
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Asif Alam Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Neha Chataut
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA
| | - Shanjana Awasthi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
2
|
Zhang P, Liu L, Yao L, Song X. Improved Differentiation Ability and Therapeutic Effect of miR-23a-3p Expressing Bone Marrow-Derived Mesenchymal Stem Cells in Mice Model with Acute Lung Injury. Int J Stem Cells 2021; 14:229-239. [PMID: 33632989 PMCID: PMC8138660 DOI: 10.15283/ijsc20136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/16/2020] [Accepted: 01/20/2021] [Indexed: 12/27/2022] Open
Abstract
Background and Objectives Implantation of bone marrow-derived mesenchymal stem cells (BMSCs) has been recognized as an effective therapy for attenuating acute lung injury (ALI). This study aims to discover microRNA (miRNA)-mediated improvement of BMSCs-based therapeutic effects. Methods and Results Mice were treated with lipopolysaccharide (LPS) for induction of ALI. BMSCs with lentivirus-mediated expression of miR-23b-3p or fibroblast growth factor 2 (FGF2) were intratracheally injected into the mice with ALI. The expressions of miR-23b-3p, FGF2, Occludin, and surfactant protein C (SPC) in lung tissues were analyzed by immunoblot or quantitative reverse transcription polymerase chain reaction. Histopathological changes in lung tissues were observed via hematoxylin-eosin staining. Lung edema was assessed by the ratio of lung wet weight/body weight (LWW/BW). The levels of interleukin (IL)-1β, IL-6, IL-4, and IL-8 in bronchoalveolar lavage fluid (BALF) were assessed by ELISA. LPS injection downregulated the expressions of miR-23b-3p, SPC and Occludin in the lung tissues, increased the LWW/BW ratio and aggravated histopathological abnormalities, while upregulating IL-1β, IL-6, IL-4, and IL-8 in the BALF. Upregulated miR-23b-3p counteracted LPS-induced effects, whereas downregulated miR-23b-3p intensified LPS-induced effects. FGF2, which was downregulated by miR-23b-3p upregulation, was a target gene of miR-23b-3p. Overexpressing FGF2 downregulated the expressions of miR-23b-3p, SPC and Occludin, increased the LWW/BW ratio and aggravated histopathological abnormalities, while upregulating IL-1β, IL-6, IL-4, and IL-8, and it offset miR-23b-3p upregulation-caused effects on the ALI mice. Conclusions Overexpression of miR-23b-3p in BMSCs strengthened BMSC-mediated protection against LPS-induced mouse acute lung injury via targeting FGF2.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Intensive Care Medicine, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Linghua Liu
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Lei Yao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaoxue Song
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
3
|
Berhan A, Harris T, Jaffar J, Jativa F, Langenbach S, Lönnstedt I, Alhamdoosh M, Ng M, Lee P, Westall G, Wilson N, Wilson M, Stewart AG. Cellular Microenvironment Stiffness Regulates Eicosanoid Production and Signaling Pathways. Am J Respir Cell Mol Biol 2021; 63:819-830. [PMID: 32926636 DOI: 10.1165/rcmb.2020-0227oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pathological changes in the biomechanical environment are implicated in the progression of idiopathic pulmonary fibrosis (IPF). Stiffened matrix augments fibroblast proliferation and differentiation and activates TGF-β1 (transforming growth factor-β1). Stiffened matrix impairs the synthesis of the antifibrogenic lipid mediator prostaglandin E2 (PGE2) and reduces the expression of the rate-limiting prostanoid biosynthetic enzyme cyclooxygenase-2 (COX-2). We now show that prostaglandin E synthase (PTGES), the final enzyme in the PGE2 biosynthetic pathway, is expressed at lower levels in the lungs of patients with IPF. We also show substantial induction of COX-2, PTGES, prostaglandin E receptor 4 (EP4), and cytosolic phospholipase A2 (cPLA2) expression in human lung fibroblasts cultured in soft collagen hydrogels or in spheroids compared with conventional culture on stiff plastic culture plates. Induction of COX-2, cPLA2, and PTGES expression in spheroid cultures was moderately inhibited by the p38 mitogen-activated protein kinase inhibitor SB203580. The induction of prostanoid biosynthetic enzyme expression was accompanied by an increase in PGE2 levels only in non-IPF-derived fibroblast spheroids. Our study reveals an extensive dysregulation of prostanoid biosynthesis and signaling pathways in IPF-derived fibroblasts, which are only partially abrogated by culture in soft microenvironments.
Collapse
Affiliation(s)
- Asres Berhan
- Department of Pharmacology and Therapeutics, and
| | - Trudi Harris
- Department of Pharmacology and Therapeutics, and
| | - Jade Jaffar
- Department of Allergy, Immunology, Respiratory Medicine, The Alfred Hospital/Monash University, Melbourne, Victoria, Australia
| | - Fernando Jativa
- Department of Pharmacology and Therapeutics, and.,Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | - Milica Ng
- CSL Ltd., Melbourne, Victoria, Australia; and
| | - Peter Lee
- Department of Biomedical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Glen Westall
- Department of Allergy, Immunology, Respiratory Medicine, The Alfred Hospital/Monash University, Melbourne, Victoria, Australia
| | - Nick Wilson
- CSL Ltd., Melbourne, Victoria, Australia; and
| | | | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, and.,ARC Centre for Personalised Therapeutics Technologies, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Epithelial Dysfunction in Lung Diseases: Effects of Amino Acids and Potential Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:57-70. [PMID: 32761570 DOI: 10.1007/978-3-030-45328-2_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung diseases affect millions of individuals all over the world. Various environmental factors, such as toxins, chemical pollutants, detergents, viruses, bacteria, microbial dysbiosis, and allergens, contribute to the development of respiratory disorders. Exposure to these factors activates stress responses in host cells and disrupt lung homeostasis, therefore leading to dysfunctional epithelial barriers. Despite significant advances in therapeutic treatments for lung diseases in the last two decades, novel interventional targets are imperative, considering the side effects and limited efficacy in patients treated with currently available drugs. Nutrients, such as amino acids (e.g., arginine, glutamine, glycine, proline, taurine, and tryptophan), peptides, and bioactive molecules, have attracted more and more attention due to their abilities to reduce oxidative stress, inhibit apoptosis, and regulate immune responses, thereby improving epithelial barriers. In this review, we summarize recent advances in amino acid metabolism in the lungs, as well as multifaceted functions of amino acids in attenuating inflammatory lung diseases based on data from studies with both human patients and animal models. The underlying mechanisms for the effects of physiological amino acids are likely complex and involve cell signaling, gene expression, and anti-oxidative reactions. The beneficial effects of amino acids are expected to improve the respiratory health and well-being of humans and other animals. Because viruses (e.g., coronavirus) and environmental pollutants (e.g., PM2.5 particles) induce severe damage to the lungs, it is important to determine whether dietary supplementation or intravenous administration of individual functional amino acids (e.g., arginine-HCl, citrulline, N-acetylcysteine, glutamine, glycine, proline and tryptophan) or their combinations to affected subjects may alleviate injury and dysfunction in this vital organ.
Collapse
|
5
|
Lats2-Underexpressing Bone Marrow-Derived Mesenchymal Stem Cells Ameliorate LPS-Induced Acute Lung Injury in Mice. Mediators Inflamm 2019; 2019:4851431. [PMID: 31772503 PMCID: PMC6854183 DOI: 10.1155/2019/4851431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/18/2019] [Accepted: 09/01/2019] [Indexed: 12/22/2022] Open
Abstract
The pathophysiology of the acute lung injury (ALI) is characterized by the damage of alveolar epithelial cells, which can be repaired by exogenous bone marrow-derived mesenchymal stem cells (BMSCs). However, the migration and differentiation abilities of BMSCs are not sufficient for the purpose, and a new approach that could strengthen the repair effects of BMSCs in ALI still needs to be clarified. We have previously proved that in vitro large tumor suppressor kinase 2- (Lats2-) underexpressing BMSCs may enhance their tissue repair effects in ALI; thus, in the present study, we tried to explore whether Lats2-underexpressing BMSCs could rescue lipopolysaccharide- (LPS-) induced ALI in vivo. BMSCs from C57BL/6 mice transfected with Lats2-interfering lentivirus vector or lentivirus blank controls were transplanted intratracheally into LPS-induced ALI mice. The retention and differentiation of BMSCs in the lung were evaluated by in vivo imaging, immunofluorescence staining, and Western blotting. The lung edema and permeability were assessed by lung wet weight/body weight ratio (LWW/BW) and measurements of proteins in bronchoalveolar lavage fluid (BALF) using ELISA. Acute lung inflammation was measured by the cytokines in the lung homogenate and BALF using RT-qPCR and ELISA, respectively. Lung injury was evaluated by HE staining and lung injury scoring. Pulmonary fibrosis was evaluated by Picrosirius red staining, immunohistochemistry for α-SMA and TGF-β1, and hydroxyproline assay and RT-qPCR for Col1α1 and Col3α1. Lats2-mediated inhibition of the Hippo pathway increased the retention of BMSCs and their differentiation toward type II alveolar epithelial cells in the lung. Furthermore, Lats2-underexpressing BMSCs improved lung edema, permeability of the lung epithelium, and lung inflammation. Finally, Lats2-underexpressing BMSCs alleviated lung injury and early pulmonary fibrosis. Our studies suggest that underexpression of Lats2 could further enhance the repair effects of BMSCs against epithelial impair and the therapeutic potential of BMSCs in ALI mice.
Collapse
|
6
|
Yang L, Wang WC, Lung SCC, Sun Z, Chen C, Chen JK, Zou Q, Lin YH, Lin CH. Polycyclic aromatic hydrocarbons are associated with increased risk of chronic obstructive pulmonary disease during haze events in China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 574:1649-1658. [PMID: 27614859 DOI: 10.1016/j.scitotenv.2016.08.211] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/28/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Although exposure to particulate matter with a diameter of <2.5μm (PM2.5) is associated with chronic obstructive pulmonary disease (COPD), the major components of PM2.5 in COPD pathogenesis are controversial. Here we employed the human lung epithelial cell line BEAS-2B to elucidate the association between COPD and the organic and water-soluble components of PM2.5. We found that the PM2.5 organic extract was a potential major risk factor for pulmonary epithelial barrier dysfunction through the depletion of proteins from the zonula occludens. This extract induced severe oxidative stress that increased DNA damage and the production of proinflammatory cytokines by BEAS-2B cells as well as decreased α1-antitrypsin expression, suggesting a mechanism that increases the risk of COPD. These effects were mainly mediated by polycyclic aromatic hydrocarbons (PAHs) through the aryl hydrocarbon receptor pathway. PAHs with high benzo(a)pyrene (BaP)-equivalent concentrations, but not major PAH components, have an increased risk of causing COPD, suggesting that BaP-equivalent concentrations represent a PM2.5-induced COPD risk metric, which may contribute to provide a rationale for the remediation of air pollution.
Collapse
Affiliation(s)
- Lingyan Yang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Wen-Cheng Wang
- Research Center for Environmental Changes, Academia Sinica, Taipei 11529, Taiwan
| | | | - Zhelin Sun
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Chongjun Chen
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Qiang Zou
- Suzhou Environmental Monitor Center, Suzhou 215004, China
| | - Yu-Hsin Lin
- Department of Food and Beverage Management, Taipei College of Maritime Technology, Taipei 11174, Taiwan
| | - Chia-Hua Lin
- Department of Biotechnology, National Formosa University, Yunlin 63208, Taiwan.
| |
Collapse
|
7
|
Sun Z, Yang L, Chen KF, Chen GW, Peng YP, Chen JK, Suo G, Yu J, Wang WC, Lin CH. Nano zerovalent iron particles induce pulmonary and cardiovascular toxicity in an in vitro human co-culture model. Nanotoxicology 2016; 10:881-90. [DOI: 10.3109/17435390.2015.1133861] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Zhelin Sun
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
- School of Life Sciences, Shanghai University, Shanghai, China,
| | - Lingyan Yang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Ku-Fan Chen
- Department of Civil Engineering, National Chi Nan University, Nantou, Taiwan,
| | - Guan-Wen Chen
- Department of Food and Beverage Management, Taipei College of Maritime Technology, Taipei, Taiwan,
| | - Yen-Ping Peng
- Department of Environmental Science and Engineering, Tunghai University, Taichung, Taiwan,
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan,
| | - Guangli Suo
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Jiantao Yu
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China,
| | - Wen-Cheng Wang
- Research Center for Environmental Changes, Academia Sinica, Taipei, Taiwan, and
| | - Chia-Hua Lin
- Department of Biotechnology, National Formosa University, Yunlin, Taiwan
| |
Collapse
|
8
|
Cai SX, Liu AR, Chen S, He HL, Chen QH, Xu JY, Pan C, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. The Orphan Receptor Tyrosine Kinase ROR2 Facilitates MSCs to Repair Lung Injury in ARDS Animal Model. Cell Transplant 2015; 25:1561-74. [PMID: 26531175 DOI: 10.3727/096368915x689776] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
There are some limitations to the therapeutic effects of mesenchymal stem cells (MSCs) on acute respiratory distress syndrome (ARDS) due to their low engraftment and differentiation rates in lungs. We found previously that noncanonical Wnt5a signaling promoted the differentiation of mouse MSCs (mMSCs) into type II alveolar epithelial cells (AT II cells), conferred resistance to oxidative stress, and promoted migration of MSCs in vitro. As receptor tyrosine kinase-like orphan receptor 2 (ROR2) is an essential receptor for Wnt5a, it was reasonable to deduce that ROR2 might be one of the key molecules for the therapeutic effect of MSCs in ARDS. The mMSCs that stably overexpressed ROR2 or the green fluorescent protein (GFP) control were transplanted intratracheally into the ARDS mice [induced by intratracheal injection of lipopolysaccharide (LPS)]. The results showed that ROR2-overexpressing mMSCs led to more significant effects than the GFP controls, including the retention of the mMSCs in the lung, differentiation into AT II cells, improvement of alveolar epithelial permeability, improvement of acute LPS-induced pulmonary inflammation, and, finally, reduction of the pathological impairment of the lung tissue. In conclusion, MSCs that overexpress ROR2 could further improve MSC-mediated protection against epithelial impairment in ARDS.
Collapse
Affiliation(s)
- Shi-Xia Cai
- Department of Critical Care Medicine, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Cai SX, Liu AR, Chen S, He HL, Chen QH, Xu JY, Pan C, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. Activation of Wnt/β-catenin signalling promotes mesenchymal stem cells to repair injured alveolar epithelium induced by lipopolysaccharide in mice. Stem Cell Res Ther 2015; 6:65. [PMID: 25889393 PMCID: PMC4414385 DOI: 10.1186/s13287-015-0060-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/03/2015] [Accepted: 03/20/2015] [Indexed: 01/11/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have potential for re-epithelization and recovery in acute respiratory distress syndrome (ARDS). In a previous in vitro study, the results showed that the canonical Wnt/β-catenin pathway promoted the differentiation of MSCs into type II alveolar epithelial cells, conferred resistance to oxidative stress, and promoted their migration, suggesting that the Wnt/β-catenin pathway might be one of the key mechanisms underling the therapeutic effect of mouse MSCs in ARDS. Methods Mouse MSCs stable transfected with β-catenin or green fluorescent protein control were transplanted intratracheally into the ARDS mice induced by lipopolysaccharide. Lung tissue injury and repair assessment were examined using haematoxylin and eosin staining, lung injury scoring, Masson’s trichrome staining and fibrosis scoring. Homing and differentiation of mouse MSCs were assayed by labelling and tracing MSCs using NIR815 dye, immunofluorescent staining, and Western immunoblot analysis. The inflammation and permeability were evaluated by detecting the cytokine and protein measurements in bronchoalveolar lavage fluid using enzyme-linked immunosorbent assay. Results In this study, β-catenin-overexpressing MSC engraftment led to more significant effects than the GFP controls, including the retention of the MSCs in the lung, differentiation into type II alveolar epithelial cells, improvement in alveolar epithelial permeability, and the pathologic impairment of the lung tissue. Conclusion These results suggest that the activation of canonical Wnt/β-catenin pathway by mouse MSCs by overexpressing β-catenin could further improve the protection of mouse MSCs against epithelial impair and the therapeutic effects of mouse MSCs in ARDS mice.
Collapse
Affiliation(s)
- Shi-xia Cai
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China. .,Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, People's Republic of China.
| | - Ai-ran Liu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Song Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Hong-li He
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Qi-hong Chen
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Jing-yuan Xu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Chun Pan
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Yi Yang
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Feng-mei Guo
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Ying-zi Huang
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Ling Liu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Hai-bo Qiu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
10
|
Jiang X, Zhang D, Zhang H, Huang Y, Teng M. Role of Ran-regulated nuclear-cytoplasmic trafficking of pVHL in the regulation of microtubular stability-mediated HIF-1α in hypoxic cardiomyocytes. Sci Rep 2015; 5:9193. [PMID: 25779090 PMCID: PMC4361876 DOI: 10.1038/srep09193] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 02/25/2015] [Indexed: 12/30/2022] Open
Abstract
Our previous study suggested that microtubule network alteration affects the process of glycolysis in cardiomyocytes (CMs) via the regulation of hypoxia-inducible factor (HIF)-1α during the early stages of hypoxia. However, little is known regarding the underlying mechanisms of microtubule network alteration-induced changes of HIF-1α. The von Hippel–Lindau tumor suppressor protein (pVHL) has been shown to mediate the ubiquitination of HIF-1α in the nuclear compartment prior to HIF-1α exportation to the cytoplasm, and pVHL dynamic nuclear-cytoplasmic trafficking is indicated to be involved in the process of HIF-1α degradation. In this study, by administering different microtubule-stabilizing and -depolymerizing interventions, we demonstrated that microtubule stabilization promoted pVHL nuclear export and drove the translocation of pVHL to the cytoplasm, while microtubule disruption prevented pVHL nuclear export in hypoxic CMs. Moreover, the ratio between nuclear and cytoplasmic pVHL was associated with HIF-1α regulation. Importantly, microtubule network alteration also affected the subcellular localization of Ran, which was involved in the regulation of pVHL nuclear-cytoplasmic trafficking. The above results suggest that the subcellular translocation of pVHL plays an important role in microtubular structure alteration-induced HIF-1α regulation. Interestingly, Ran is involved in the process of pVHL nuclear-cytoplasmic trafficking following microtubule network alteration in hypoxic CMs.
Collapse
Affiliation(s)
- Xupin Jiang
- 1] Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China [2] Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Hengshu Zhang
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Miao Teng
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Hsieh SJ, Zhuo H, Benowitz NL, Thompson BT, Liu KD, Matthay MA, Calfee CS. Prevalence and impact of active and passive cigarette smoking in acute respiratory distress syndrome. Crit Care Med 2014; 42:2058-68. [PMID: 24942512 PMCID: PMC4134734 DOI: 10.1097/ccm.0000000000000418] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Cigarette smoke exposure has recently been found to be associated with increased susceptibility to trauma- and transfusion-associated acute respiratory distress syndrome. We sought to determine 1) the incidence of cigarette smoke exposure in a diverse multicenter sample of acute respiratory distress syndrome patients and 2) whether cigarette smoke exposure is associated with severity of lung injury and mortality in acute respiratory distress syndrome. DESIGN Analysis of the Albuterol for the Treatment of Acute Lung Injury and Omega Acute Respiratory Distress Syndrome Network studies. SETTING Acute Respiratory Distress Syndrome Network hospitals. PATIENTS Three hundred eighty-one patients with acute respiratory distress syndrome. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol, a validated tobacco-specific marker, was measured in urine samples from subjects enrolled in two National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome Network randomized controlled trials. Urine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol levels were consistent with active smoking in 36% of acute respiratory distress syndrome patients and with passive smoking in 41% of nonsmokers (vs 20% and 40% in general population, respectively). Patients with 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol levels in the active smoking range were younger and had a higher incidence of alcohol misuse, fewer comorbidities, lower severity of illness, and less septic shock at enrollment compared with patients with undetectable 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol levels. Despite this lower severity of illness, the severity of lung injury did not significantly differ based on biomarker-determined smoking status. Cigarette smoke exposure was not significantly associated with death after adjusting for differences in age, alcohol use, comorbidities, and severity of illness. CONCLUSIONS In this first multicenter study of biomarker-determined cigarette smoke exposure in acute respiratory distress syndrome patients, we found that active cigarette smoke exposure was significantly more prevalent among acute respiratory distress syndrome patients compared to population averages. Despite their younger age, better overall health, and lower severity of illness, smokers by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol had similar severity of lung injury as patients with undetectable 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol. These findings suggest that active cigarette smoking may increase susceptibility to acute respiratory distress syndrome in younger, healthier patients.
Collapse
Affiliation(s)
- S. Jean Hsieh
- Division of Critical Care Medicine, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York
| | - Hanjing Zhuo
- Cardiovascular Research Institute, San Francisco, CA
| | - Neal L. Benowitz
- Division of Clinical Pharmacology and Experimental Therapeutics, University of California, San Francisco, California
- Center for Tobacco Control Research and Education, University of California, San Francisco, San Francisco, California
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kathleen D. Liu
- Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, California
- Department of Anesthesia, University of California San Francisco, San Francisco, California
| | - Michael A. Matthay
- Cardiovascular Research Institute, San Francisco, CA
- Department of Anesthesia, University of California San Francisco, San Francisco, California
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco
| | - Carolyn S. Calfee
- Center for Tobacco Control Research and Education, University of California, San Francisco, San Francisco, California
- Department of Anesthesia, University of California San Francisco, San Francisco, California
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco
| | | |
Collapse
|
12
|
Al-Shmgani HS, Moate RM, Macnaughton PD, Sneyd JR, Moody AJ. Effects of hyperoxia on the permeability of 16HBE14o- cell monolayers--the protective role of antioxidant vitamins E and C. FEBS J 2013; 280:4512-21. [PMID: 23809212 DOI: 10.1111/febs.12413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/05/2013] [Accepted: 06/13/2013] [Indexed: 12/29/2022]
Abstract
The use of hyperoxia for critically ill patients is associated with adverse impacts resulting in lung injury accompanied by inflammation. The aim of this study was to evaluate aspects of mechanisms that contribute to hyperoxia-induced disruption of the epithelial permeability barrier, and also the protective effects of the antioxidants α-tocopherol and ascorbate. 16HBE14o- cells were cultured as monolayers at an air-liquid interface for 6 days, after which transepithelial electrical resistance reached 251.2 ± 4.1 Ω.cm(2) (mean ± standard error of the mean). They were then exposed for 24 h to normoxia (21% O2, 5% CO2), hyperoxia (95% O2, 5% CO2), hyperoxia with 10(-7) M α-tocopherol, hyperoxia with 10(-7) M ascorbate, hyperoxia with 10(-6) M ascorbate, and hyperoxia with a combination of α-tocopherol and ascorbate (10(-7) M and 10(-6) M, respectively). Significant reductions (P < 0.05) in transepithelial electrical resistance seen after hyperoxia (with or without antioxidants) were associated with reductions in the levels of zona occludens-1 (ZO-1) observed by immunohistochemistry, and downregulation of ZO-1 expression (P < 0.01) as compared with normoxia. In contrast, the expression levels of interleukin (IL)-8, IL-6 and tumour necrosis factor-α (TNF-α) were increased after hyperoxia (P < 0.01), and marked increases in the levels of these cytokines (ELISA) were seen in the medium (P < 0.001) as compared with normoxia. The antioxidant vitamins E and C had a partial protective effect against the hyperoxia-induced reduction in ZO-1 levels and the increase in levels of the proinflammatory cytokines IL-8, IL-6, and TNF-α. In conclusion, hyperoxia-induced epithelial disruption is associated with tight junction weakening, and induction of a proinflammatory environment.
Collapse
|
13
|
Triggers and effectors of oxidative stress at blood-brain barrier level: relevance for brain ageing and neurodegeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297512. [PMID: 23533687 PMCID: PMC3606793 DOI: 10.1155/2013/297512] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/27/2013] [Accepted: 01/31/2013] [Indexed: 01/23/2023]
Abstract
As fundamental research advances, it is becoming increasingly clear that a clinically expressed disease implies a mixture of intertwining molecular disturbances. Oxidative stress is one of such pathogenic pathways involved in virtually all central nervous system pathologies, infectious, inflammatory, or degenerative in nature. Since brain homeostasis largely depends on integrity of blood-brain barrier (BBB), many studies focused lately on BBB alteration in a wide spectrum of brain diseases. The proper two-way molecular transfer through BBB depends on several factors, including the functional status of its tight junction (TJ) complexes of proteins sealing neighbour endothelial cells. Although there is abundant experimental work showing that oxidative stress associates BBB permeability alteration, less is known about its implications, at molecular level, in TJ protein expression or TJ-related cell signalling. In this paper, oxidative stress is presented as a common pathway for different brain pathogenic mechanisms which lead to BBB dysregulation. We revise here oxidative-induced molecular mechanisms of BBB disruption and TJ protein expression alteration, in relation to ageing and neurodegeneration.
Collapse
|
14
|
You K, Xu X, Fu J, Xu S, Yue X, Yu Z, Xue X. Hyperoxia disrupts pulmonary epithelial barrier in newborn rats via the deterioration of occludin and ZO-1. Respir Res 2012; 13:36. [PMID: 22559818 PMCID: PMC3424121 DOI: 10.1186/1465-9921-13-36] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 05/04/2012] [Indexed: 12/20/2022] Open
Abstract
Background Prolonged exposure to hyperoxia in neonates can cause hyperoxic acute lung injury (HALI), which is characterized by increased pulmonary permeability and diffuse infiltration of various inflammatory cells. Disruption of the epithelial barrier may lead to altered pulmonary permeability and maintenance of barrier properties requires intact epithelial tight junctions (TJs). However, in neonatal animals, relatively little is known about how the TJ proteins are expressed in the pulmonary epithelium, including whether expression of TJ proteins is regulated in response to hyperoxia exposure. This study determines whether changes in tight junctions play an important role in disruption of the pulmonary epithelial barrier during hyperoxic acute lung injury. Methods Newborn rats, randomly divided into two groups, were exposed to hyperoxia (95% oxygen) or normoxia for 1–7 days, and the severity of lung injury was assessed; location and expression of key tight junction protein occludin and ZO-1 were examined by immunofluorescence staining and immunobloting; messenger RNA in lung tissue was studied by RT-PCR; transmission electron microscopy study was performed for the detection of tight junction morphology. Results We found that different durations of hyperoxia exposure caused different degrees of lung injury in newborn rats. Treatment with hyperoxia for prolonged duration contributed to more serious lung injury, which was characterized by increased wet-to-dry ratio, extravascular lung water content, and bronchoalveolar lavage fluid (BALF):serum FD4 ratio. Transmission electron microscopy study demonstrated that hyperoxia destroyed the structure of tight junctions and prolonged hyperoxia exposure, enhancing the structure destruction. The results were compatible with pathohistologic findings. We found that hyperoxia markedly disrupted the membrane localization and downregulated the cytoplasm expression of the key tight junction proteins occludin and ZO-1 in the alveolar epithelium by immunofluorescence. The changes of messenger RNA and protein expression of occludin and ZO-1 in lung tissue detected by RT-PCR and immunoblotting were consistent with the degree of lung injury. Conclusions These data suggest that the disruption of the pulmonary epithelial barrier induced by hyperoxia is, at least in part, due to massive deterioration in the expression and localization of key TJ proteins.
Collapse
Affiliation(s)
- Kai You
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Specific inhibition of AQP1 water channels in human pulmonary microvascular endothelial cells by small interfering RNAs. J Trauma Acute Care Surg 2012; 72:150-61. [PMID: 22310126 DOI: 10.1097/ta.0b013e318230e25d] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Aquaporin (AQP)-1 is expressed in most microvasculature endothelial cells forming water channels that play major roles in a variety of physiologic processes. Our aim was to investigate the regulatory functions of AQP1 on trancellular and paracellular permeability. METHODS We designed, synthesized, and used small interfering RNAs (siRNAs) selective for AQP1 and investigated their effectiveness in altering AQP1-mediated permeability in human pulmonary microvascular endothelial cells. RESULTS Twenty-four hours after transfection of ECs with siRNAs targeting two different regions of the AQP1 transcript, AQP1 protein was inhibited by 47.8% to 74.6%. siRNAs containing the same percent of base pairs as the AQP1-siRNAs but in random sequence (i.e., scrambled siRNAs) had no effect. Suppression of AQP1 expression in ECs resulted in decreases in epithelial Na+ channel (ENaC) and Na-K ATPase of ECs, and the suppression ENaC α, β, γ, and Na-K ATPase were 43.1% to 48.2%,70.0% to 76.0%, 52.6% to 55.0%, and 72.7% to 79.3%, respectively. The reduced AQP1expression also resulted in decreased cell-cell junction protein level of VE-cadherin, which was suppressed by 36.5% to 59.5% but had no effect on occludin protein. Tube formation assay and tranwell assay showed AQP1 siRNAs induced high permeability of human pulmonary microvascular endothelial cells. Rho-kinase (ROCK) I and ROCK II were increased by 46.0% to 50.0% and 59% to 81%, respectively, AQP1 siRNA treatment accelerated the formation of F-actin bundles, demonstrating the activation of Rho/ROCK signaling pathway, and decreased mitochondrial membrane potential after AQP1 siRNA treatment, showing an important event of apoptosis process. CONCLUSIONS The data demonstrate that AQP1 is a critical participate in regulating endothelial permeability and barrier function and provide direct evidence of the contribution of AQP1 to blood vessel formation.
Collapse
|
16
|
González-Mariscal L, Quirós M, Díaz-Coránguez M. ZO proteins and redox-dependent processes. Antioxid Redox Signal 2011; 15:1235-53. [PMID: 21294657 DOI: 10.1089/ars.2011.3913] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SIGNIFICANCE ZO-1, ZO-2, and ZO-3 are scaffold proteins of the tight junction (TJ) that belong to the MAGUK protein family characterized for exhibiting PDZ, SH3, and GuK domains. ZO proteins are present only in multicellular organisms, being the placozoa the first to have them. ZO proteins associate among themselves and with other integral and adaptor proteins of the TJ, of the ZA and of gap junctions, as with numerous signaling proteins and the actin cytoskeleton. ZO proteins are also present at the nucleus of proliferating cells. RECENT ADVANCES Oxidative stress disassembles the TJs of endothelial and epithelial cells. CRITICAL ISSUES Oxidative stress alters ZO proteins expression and localization, in conditions like hypoxia, bacterial and viral infections, vitamin deficiencies, age-related diseases, diabetes and inflammation, alcohol and tobacco consumption. FUTURE DIRECTIONS Molecules present in the signaling pathways triggered by oxidative stress can be targets for therapeutic intervention.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico DF, México.
| | | | | |
Collapse
|
17
|
Study on the relationship of cPLA2, CK-MB, and membrane phospholipid content in acute myocardial infarction. Heart Vessels 2010; 26:64-8. [DOI: 10.1007/s00380-010-0031-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 02/07/2010] [Indexed: 10/18/2022]
|
18
|
D'Souza El-Guindy NB, Kovacs EJ, De Witte P, Spies C, Littleton JM, de Villiers WJS, Lott AJ, Plackett TP, Lanzke N, Meadows GG. Laboratory models available to study alcohol-induced organ damage and immune variations: choosing the appropriate model. Alcohol Clin Exp Res 2010; 34:1489-511. [PMID: 20586763 PMCID: PMC2929290 DOI: 10.1111/j.1530-0277.2010.01234.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The morbidity and mortality resulting from alcohol-related diseases globally impose a substantive cost to society. To minimize the financial burden on society and improve the quality of life for individuals suffering from the ill effects of alcohol abuse, substantial research in the alcohol field is focused on understanding the mechanisms by which alcohol-related diseases develop and progress. Since ethical concerns and inherent difficulties limit the amount of alcohol abuse research that can be performed in humans, most studies are performed in laboratory animals. This article summarizes the various laboratory models of alcohol abuse that are currently available and are used to study the mechanisms by which alcohol abuse induces organ damage and immune defects. The strengths and weaknesses of each of the models are discussed. Integrated into the review are the presentations that were made in the symposium "Methods of Ethanol Application in Alcohol Model-How Long is Long Enough" at the joint 2008 Research Society on Alcoholism (RSA) and International Society for Biomedical Research on Alcoholism (ISBRA) meeting, Washington, DC, emphasizing the importance not only of selecting the most appropriate laboratory alcohol model to address the specific goals of a project but also of ensuring that the findings can be extrapolated to alcohol-induced diseases in humans.
Collapse
Affiliation(s)
- Nympha B D'Souza El-Guindy
- Department of Internal Medicine, Division of Digestive Diseases, University of Kentucky and Veterans Affairs Medical Center, Lexington, Kentucky, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Xia XM, Wang FY, Wang ZK, Wan HJ, Xu WA, Lu H. Emodin enhances alveolar epithelial barrier function in rats with experimental acute pancreatitis. World J Gastroenterol 2010; 16:2994-3001. [PMID: 20572302 PMCID: PMC2890939 DOI: 10.3748/wjg.v16.i24.2994] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of emodin on expression of claudin-4, claudin-5 and occludin, as well as the alveolar epithelial barrier in rats with pancreatitis induced by sodium taurocholate.
METHODS: Experimental pancreatitis was induced by retrograde injection of 5% sodium taurocholate into the biliopancreatic duct. Emodin was injected via the external jugular vein 3 h after induction of acute pancreatitis. Rats from sham operation group and acute pancreatitis group were injected with normal saline (an equivalent volume as emodin) at the same time point. Samples of lung and serum were obtained 6 h after drug administration. Pulmonary morphology was examined with HE staining. Pulmonary edema was estimated by measuring water content in lung tissue samples. Tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) level were measured by enzyme-linked immunospecific assay. Serum amylase and pulmonary myeloperoxidase (MPO) activity were detected by spectrophotometry. Alveolar epithelial barrier was assessed by pulmonary dye extravasation. Expression of claudin-4, claudin-5 and occludin in lung tissue samples was examined by immunohistology, quantitative real-time reverse transcription polymerase chain reaction and Western blotting analysis, respectively.
RESULTS: Pancreatitis-associated lung injury was characterized by pulmonary edema, leukocyte infiltration, alveolar collapse, and elevated serum amylase level. The pulmonary damage, pulmonary pathological scores, serum amylase and MPO activity, TNF-α and IL-6 levels, and wet/dry ratio were decreased in rats after treatment with emodin. Immunostaining of claudin-4, claudin-5 and occludin was detected in lung tissue samples from rats in sham operation group, which was distributed in alveolar epithelium, vascular endothelium, and bronchial epithelium, respectively. The mRNA and protein expression levels of claudin-4, claudin-5 and occludin in lung tissue samples were markedly decreased, the expression level of claudin-4, claudin-5 and occluding was increased, and the pulmonary dye extravasation was reduced in lung tissue samples from rats with acute pancreatitis after treatment with emodin.
CONCLUSION: Emodin attenuates pulmonary edema and inflammation, enhances alveolar epithelial barrier function, and promotes expression of claudin-4, claudin-5 and occludin in lung tissue samples from rats with acute pancreatitis.
Collapse
|
20
|
Teng M, Dang YM, Zhang JP, Zhang Q, Fang YD, Ren J, Huang YS. Microtubular stability affects cardiomyocyte glycolysis by HIF-1alpha expression and endonuclear aggregation during early stages of hypoxia. Am J Physiol Heart Circ Physiol 2010; 298:H1919-31. [PMID: 20228255 DOI: 10.1152/ajpheart.01039.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypoxia-inducible factor (HIF)-1alpha is a key regulator of anaerobic energy metabolism. We asked the following question: Does the breakdown of microtubular structures influence glycolysis in hypoxic cardiomyocytes by regulating HIF-1alpha? Neonatal rat cardiomyocytes were cultured under hypoxic conditions, while microtubule-stabilizing (paclitaxel) and -depolymerizing (colchicine) agents were used to change microtubular structure. Models of high microtubule-associated protein 4 (MAP4) expression and RNA interference of microtubulin expression were established. Microtubular structural changes and intracellular HIF-1alpha protein distribution were observed with laser confocal scanning microscopy. Content of key glycolytic enzymes, viability, and energy content of cardiomyocytes were determined by colorimetry and high-performance liquid chromatography. HIF-1alpha protein content and mRNA expression were determined by Western blotting and real-time PCR, respectively. Low doses of microtubule-stabilizing agent (10 mumol/l paclitaxel) and enhanced expression of MAP4 stabilized the reticular microtubular structures in hypoxic cardiomyocytes, increased the content of key glycolytic enzymes, ameliorated energy supply and enhanced cell viability, and upregulated HIF-1alpha protein expression and endonuclear aggregation. In contrast, the microtubule-depolymerizing agent (10 mumol/l colchicine) or reduced microtubulin expression had adverse affects on the same parameters, in particular, HIF-1alpha protein content and endonuclear aggregation. We conclude that microtubular structural changes influence glycolysis in the early stages of hypoxia in cardiomyocytes by regulating HIF-1alpha content. Stabilizing microtubular structures increases endonuclear and total HIF-1alpha expression, content of key glycolytic enzymes, and energy supply. These findings provide potential therapeutic targets for ameliorating cell energy metabolism during early myocardial hypoxia.
Collapse
Affiliation(s)
- Miao Teng
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical Univ., Chongqing 400038, China
| | | | | | | | | | | | | |
Collapse
|
21
|
McCall IC, Betanzos A, Weber DA, Nava P, Miller GW, Parkos CA. Effects of phenol on barrier function of a human intestinal epithelial cell line correlate with altered tight junction protein localization. Toxicol Appl Pharmacol 2009; 241:61-70. [PMID: 19679145 DOI: 10.1016/j.taap.2009.08.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 07/14/2009] [Accepted: 08/03/2009] [Indexed: 01/11/2023]
Abstract
Phenol contamination of soil and water has raised concerns among people living near phenol-producing factories and hazardous waste sites containing the chemical. Phenol, particularly in high concentrations, is an irritating and corrosive substance, making mucosal membranes targets of toxicity in humans. However, few data on the effects of phenol after oral exposure exist. We used an in vitro model employing human intestinal epithelial cells (SK-CO15) cultured on permeable supports to examine effects of phenol on epithelial barrier function. We hypothesized that phenol disrupts epithelial barrier by altering tight junction (TJ) protein expression. The dose-response effect of phenol on epithelial barrier function was determined using transepithelial electrical resistance (TER) and FITC-dextran permeability measurements. We studied phenol-induced changes in cell morphology and expression of several tight junction proteins by immunofluorescence and Western blot analysis. Effects on cell viability were assessed by MTT, Trypan blue, propidium iodide and TUNEL staining. Exposure to phenol resulted in decreased TER and increased paracellular flux of FITC-dextran in a dose-dependent manner. Delocalization of claudin-1 and ZO-1 from TJs to cytosol correlated with the observed increase in permeability after phenol treatment. Additionally, the decrease in TER correlated with changes in the distribution of a membrane raft marker, suggesting phenol-mediated effects on membrane fluidity. Such observations were independent of effects of phenol on cell viability as enhanced permeability occurred at doses of phenol that did not cause cell death. Overall, these findings suggest that phenol may affect transiently the lipid bilayer of the cell membrane, thus destabilizing TJ-containing microdomains.
Collapse
Affiliation(s)
- Ingrid C McCall
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Glutamine attenuates lipopolysaccharide-induced acute lung injury. Nutrition 2009; 25:692-8. [PMID: 19286350 DOI: 10.1016/j.nut.2008.11.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 11/24/2008] [Accepted: 11/25/2008] [Indexed: 11/21/2022]
Abstract
OBJECTIVES It has been reported that glutamine (GLN) can attenuate acute lung injury after sepsis. GLN is also thought to be a precursor of glutathione (GSH) synthesis. Using the GSH synthesis blocker, L-buthionine-(S,R)-sulfoximine (BSO), we investigated the role of GSH synthesis in the protective effect of GLN on acute lung injury. METHODS In this study, we used an acute lung injury model induced by intratracheal injection of lipopolysaccharide (1 mg mL(-1) kg(-1)). GLN (0.75 g/kg, intravenous) and BSO (2 mmol/kg, intraperitoneal) were administrated simultaneously. At 2 and 18 h after the injections, the rats were sacrificed by right ventricular puncture and bronchoalveolar lavage was done. The lower right lung was excised for histologic examination. Total protein concentration and total cell and neutrophil counts in the bronchoalveolar lavage fluid were determined. CD11b expression in the blood was determined by flow cytometry. We also analyzed myeloperoxidase activity, and GSH and interleukin-8 levels in lung tissues. RESULTS GLN supplementation reduced the total protein concentration and total cell and neutrophils counts in bronchoalveolar lavage fluid after lipopolysaccharide challenge. GLN enhanced GSH synthesis and attenuated interleukin-8 release and myeloperoxidase activity in lung tissues. GLN also decreased CD11b expression in blood neutrophils and prevented lung histologic changes. BSO abolished the effects of GLN and attenuated its protection on acute lung injury. CONCLUSION These results indicate that GLN could prevent neutrophil recruitment and infiltration, protect the alveolar barrier, and attenuate inflammatory injury during sepsis. This effect may be related to enhanced GSH synthesis.
Collapse
|
23
|
WHAT'S NEW IN SHOCK, AUGUST 2007? Shock 2007. [DOI: 10.1097/shk.0b013e3181238792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|