251
|
Bchir S, Nasr HB, Bouchet S, Benzarti M, Garrouch A, Tabka Z, Susin S, Chahed K, Bauvois B. Concomitant elevations of MMP-9, NGAL, proMMP-9/NGAL and neutrophil elastase in serum of smokers with chronic obstructive pulmonary disease. J Cell Mol Med 2016; 21:1280-1291. [PMID: 28004483 PMCID: PMC5487915 DOI: 10.1111/jcmm.13057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022] Open
Abstract
A growing body of evidence points towards smoking‐related phenotypic differences in chronic obstructive pulmonary disease (COPD). As COPD is associated with systemic inflammation, we determined whether smoking status is related to serum levels of matrix metalloproteinase‐9 (pro‐ and active MMP‐9), neutrophil gelatinase‐associated lipocalin (NGAL) and the proMMP‐9/NGAL complex in patients with COPD. Serum samples were collected in 100 stable‐phase COPD patients (82 smokers, 18 never‐smokers) and 28 healthy adults (21 smokers, 7 never‐smokers). Serum levels of studied factors were measured in ELISA. Our data provide the first evidence of simultaneously elevated serum levels of MMP‐9, NGAL and proMMP‐9/NGAL in COPD smokers. While the triad discriminated between smokers and non‐smokers in the COPD group, MMP‐9 and proMMP‐9/NGAL (but not NGAL) discriminated between smokers with and without COPD. Adjustment for age and smoking pack‐years did not alter the findings. Serum MMP‐9, NGAL and proMMP‐9/NGAL levels were not correlated with the GOLD stage or FEV1 decline. Furthermore, serum levels of neutrophil elastase (NE) and MMP‐3 (but not of IL‐6 and MMP‐12) were also higher in COPD smokers than in healthy smokers before and after adjustment for age and pack‐years. Among COPD smokers, levels of MMP‐9, NGAL and proMMP‐9/NGAL were positively correlated with NE (P < 0.0001) but not with the remaining factors. Gelatin zymography detected proMMP‐9 in serum samples of healthy and COPD smoking groups. Our results suggest that associated serum levels of proMMP‐9, NGAL, proMMP‐9/NGAL and NE may reflect the state of systemic inflammation in COPD related to cigarette smoking.
Collapse
Affiliation(s)
- Sarra Bchir
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia.,Institut Supérieur de Biotechnologie de Monastir, Université de Monastir, Monastir, Tunisia.,Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Hela Ben Nasr
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Sandrine Bouchet
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France.,Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Mohamed Benzarti
- Service de Pneumo-Allergologie, CHU Farhat Hached, Sousse, Tunisia
| | | | - Zouhair Tabka
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia
| | - Santos Susin
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Karim Chahed
- Unité de recherche UR12ES06, Physiologie de l'Exercice et Physiopathologie de l'Intégré au Moléculaire, Biologie, Médecine et Santé, Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisia.,Faculté des Sciences de Sfax, Université de Sfax, Sfax, Tunisia
| | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Universités UPMC Paris 06, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| |
Collapse
|
252
|
New Role of Adult Lung c-kit + Cells in a Mouse Model of Airway Hyperresponsiveness. Mediators Inflamm 2016; 2016:3917471. [PMID: 28090152 PMCID: PMC5206449 DOI: 10.1155/2016/3917471] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
Structural changes contribute to airway hyperresponsiveness and airflow obstruction in asthma. Emerging evidence points to the involvement of c-kit+ cells in lung homeostasis, although their potential role in asthma is unknown. Our aim was to isolate c-kit+ cells from normal mouse lungs and to test whether these cells can interfere with hallmarks of asthma in an animal model. Adult mouse GFP-tagged c-kit+ cells, intratracheally delivered in the ovalbumin-induced airway hyperresponsiveness, positively affected airway remodeling and improved airway function. In bronchoalveolar lavage fluid of cell-treated animals, a reduction in the number of inflammatory cells and in IL-4, IL-5, and IL-13 release, along with an increase of IL-10, was observed. In MSC-treated mice, the macrophage polarization to M2-like subset may explain, at least in part, the increment in the level of anti-inflammatory cytokine IL-10. After in vitro stimulation of c-kit+ cells with proinflammatory cytokines, the indoleamine 2,3-dioxygenase and TGFβ were upregulated. These data, together with the increased apoptosis of inflammatory cells in vivo, indicate that c-kit+ cells downregulate immune response in asthma by influencing local environment, possibly by cell-to-cell contact combined to paracrine action. In conclusion, intratracheally administered c-kit+ cells reduce inflammation, positively modulate airway remodeling, and improve function. These data document previously unrecognized properties of c-kit+ cells, able to impede pathophysiological features of experimental airway hyperresponsiveness.
Collapse
|
253
|
Gohy ST, Hupin C, Pilette C, Ladjemi MZ. Chronic inflammatory airway diseases: the central role of the epithelium revisited. Clin Exp Allergy 2016; 46:529-42. [PMID: 27021118 DOI: 10.1111/cea.12712] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The respiratory epithelium plays a critical role for the maintenance of airway integrity and defense against inhaled particles. Physical barrier provided by apical junctions and mucociliary clearance clears inhaled pathogens, allergens or toxics, to prevent continuous stimulation of adaptive immune responses. The "chemical barrier", consisting of several anti-microbial factors such as lysozyme and lactoferrin, constitutes another protective mechanism of the mucosae against external aggressions before adaptive immune response starts. The reconstruction of damaged respiratory epithelium is crucial to restore this barrier. This review examines the role of the airway epithelium through recent advances in health and chronic inflammatory diseases in the lower conducting airways (in asthma and chronic obstructive pulmonary disease). Better understanding of normal and altered epithelial functions continuously provides new insights into the physiopathology of chronic airway diseases and should help to identify new epithelial-targeted therapies.
Collapse
Affiliation(s)
- S T Gohy
- Université catholique de Louvain (UCL), Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Brussels, Belgium.,Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| | - C Hupin
- Université catholique de Louvain (UCL), Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Brussels, Belgium
| | - C Pilette
- Université catholique de Louvain (UCL), Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Brussels, Belgium.,Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium.,Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Brussels, Belgium
| | - M Z Ladjemi
- Université catholique de Louvain (UCL), Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Brussels, Belgium.,Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Brussels, Belgium
| |
Collapse
|
254
|
Bruzauskaite I, Raudoniute J, Denkovskij J, Bagdonas E, Meidute-Abaraviciene S, Simonyte V, Bironaite D, Siaurys A, Bernotiene E, Aldonyte R. Native matrix-based human lung alveolar tissue model in vitro: studies of the reparatory actions of mesenchymal stem cells. Cytotechnology 2016; 69:1-17. [PMID: 27905026 DOI: 10.1007/s10616-016-0021-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/22/2016] [Indexed: 12/22/2022] Open
Abstract
Studies of lung diseases in vitro often rely on flat, plastic-based monocultures, due to short lifespan of primary cells, complicated anatomy, lack of explants, etc. We hereby present a native 3D model with cues for repopulating epithelial cells. Abilities of mesenchymal stem cells (MSC) to modulate bacterial lipopolysaccharide (LPS) and cigarette smoke-induced injury to pulmonary epithelium were tested in our model. Post-mortem human lung tissue was sliced, cut and decellularized. Resulting matrix pads were reseeded with pulmonary epithelium (A549 line). Markers of the layer integrity and certain secreted proteins in the presence of cigarette smoke extract (CSE) and LPS were assessed via Western blot, ELISA and RT-PCR assays. In parallel, the effects of MSC paracrine factors on exposed epithelial cells were also investigated at gene and protein levels. When cultured on native 3D matrix, A549 cells obtain dual, type I- and II-like morphology. Exposure to CSE and LPS leads to downregulation of several epithelial proteins and suppressed proliferation rate. MSC medium added to the model restores proliferation rate and some of the epithelial proteins, i.e. e-cadherin and beta-catenin. CSE also increases secretion of pro-inflammatory cytokines by epithelial cells and upregulates transcription factor NFκB. Some of these effects might be counteracted by MSC in our model. We introduce repopulated decellularized lung matrix that highly resembles in vivo situation and is convenient for studies of disease pathogenesis, cytotoxicology and for exploring therapeutic strategies in the human lung context in vitro. MSC paracrine products have produced protecting effects in our model.
Collapse
Affiliation(s)
- Ieva Bruzauskaite
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Jovile Raudoniute
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Jaroslav Denkovskij
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Edvardas Bagdonas
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | | | - Vaida Simonyte
- Department of Pathology, Vilnius City Hospital, Antakalnio 57, Vilnius, Lithuania
| | - Daiva Bironaite
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Almantas Siaurys
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Eiva Bernotiene
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania
| | - Ruta Aldonyte
- State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08046, Vilnius, Lithuania.
| |
Collapse
|
255
|
Wang X, Wang Y, Zhang J, Guan X, Chen M, Li Y, Zhang L. Galectin-3 contributes to vascular fibrosis in monocrotaline-induced pulmonary arterial hypertension rat model. J Biochem Mol Toxicol 2016; 31. [PMID: 27870162 DOI: 10.1002/jbt.21879] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/13/2016] [Accepted: 10/20/2016] [Indexed: 01/31/2023]
Abstract
Galectin-3 (Gal-3) plays a critical role in vascular inflammation and fibrosis. The role of TGF-β1 in mediating pulmonary vascular fibrosis is well documented; thus, we suspected that Gal-3 could be an important factor in TGF-β1-induced fibrosis in pulmonary adventitial fibroblasts (PAFs). We treated rats with monocrotaline (MCT) and cultured PAFs with TGF-β1 to stimulate fibrosis. We found that MCT injection induced vessel thickening and extracellular matrix deposition in vivo. TGF-β1 stimulated the production of collagen and fibronectin (Fn) protein in vitro. TGF-β1 promoted the expression of Gal-3 and its translocation, while silencing Gal-3 reduced Col-1a deposition. Blockage of STAT3 decreased the expression of Gal-3 induced by TGF-β1. Gal-3 increased Col-1a accumulation and downregulated matrix metallopeptidase 9 (MMP-9) expression in PAFs, but it did not affect Fn expression. These findings demonstrate that Gal-3 is required for TGF-β1-stimulated vascular fibrosis via a STAT3 signaling cascade and that MMP-9 is also involved in TGF-β1/Gal-3-induced vascular fibrosis.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Physiology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, People's Republic of China
| | - Yanli Wang
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei Province, 063000, People's Republic of China
| | - Jinbo Zhang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Xue Guan
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Minggang Chen
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China
| | - Yumei Li
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, China
| | - Li Zhang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, People's Republic of China.,Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang Province, 163319, China
| |
Collapse
|
256
|
Lung remodeling associated with recovery from acute lung injury. Cell Tissue Res 2016; 367:495-509. [DOI: 10.1007/s00441-016-2521-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
|
257
|
Ota C, Baarsma HA, Wagner DE, Hilgendorff A, Königshoff M. Linking bronchopulmonary dysplasia to adult chronic lung diseases: role of WNT signaling. Mol Cell Pediatr 2016; 3:34. [PMID: 27718180 PMCID: PMC5055515 DOI: 10.1186/s40348-016-0062-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in infants caused by pre- and/or postnatal lung injury. BPD is characterized by arrested alveolarization and vascularization due to extracellular matrix remodeling, inflammation, and impaired growth factor signaling. WNT signaling is a critical pathway for normal lung development, and its altered signaling has been shown to be involved in the onset and progression of incurable chronic lung diseases in adulthood, such as chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF). In this review, we summarize the impact of WNT signaling on different stages of lung development and its potential contribution to developmental lung diseases, especially BPD, and chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Chiharu Ota
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.,The Perinatal Center, Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
258
|
Paris AJ, Liu Y, Mei J, Dai N, Guo L, Spruce LA, Hudock KM, Brenner JS, Zacharias WJ, Mei HD, Slamowitz AR, Bhamidipati K, Beers MF, Seeholzer SH, Morrisey EE, Worthen GS. Neutrophils promote alveolar epithelial regeneration by enhancing type II pneumocyte proliferation in a model of acid-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1062-L1075. [PMID: 27694472 PMCID: PMC5206401 DOI: 10.1152/ajplung.00327.2016] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/25/2016] [Indexed: 12/18/2022] Open
Abstract
Alveolar epithelial regeneration is essential for resolution of the acute respiratory distress syndrome (ARDS). Although neutrophils have traditionally been considered mediators of epithelial damage, recent studies suggest they promote type II pneumocyte (AT2) proliferation, which is essential for regenerating alveolar epithelium. These studies did not, however, evaluate this relationship in an in vivo model of alveolar epithelial repair following injury. To determine whether neutrophils influence alveolar epithelial repair in vivo, we developed a unilateral acid injury model that creates a severe yet survivable injury with features similar to ARDS. Mice that received injections of the neutrophil-depleting Ly6G antibody had impaired AT2 proliferation 24 and 72 h after acid instillation, which was associated with decreased reepithelialization and increased alveolar protein concentration 72 h after injury. As neutrophil depletion itself may alter the cytokine response, we questioned the contribution of neutrophils to alveolar epithelial repair in neutropenic granulocyte-colony stimulating factor (G-CSF)-/- mice. We found that the loss of G-CSF recapitulated the neutrophil response of Ly6G-treated mice and was associated with defective alveolar epithelial repair, similar to neutrophil-depleted mice, and was reversed by administration of exogenous G-CSF. To approach the mechanisms, we employed an unbiased protein analysis of bronchoalveolar lavage fluid from neutrophil-depleted and neutrophil-replete mice 12 h after inducing lung injury. Pathway analysis identified significant differences in multiple signaling pathways that may explain the differences in epithelial repair. These data emphasize an important link between the innate immune response and tissue repair in which neutrophils promote alveolar epithelial regeneration.
Collapse
Affiliation(s)
- Andrew J Paris
- Division of Pulmonary, Allergy, and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yuhong Liu
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Junjie Mei
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ning Dai
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lei Guo
- Department of Viral Immunology, Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Kunming, China
| | - Lynn A Spruce
- Department of Pathology and Laboratory Medicine, Cell Pathology Division, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kristin M Hudock
- Division of Pulmonary, Allergy, and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Division of Pulmonary, Critical Care and Sleep Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Jacob S Brenner
- Division of Pulmonary, Allergy, and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - William J Zacharias
- Division of Pulmonary, Allergy, and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hankun D Mei
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Kartik Bhamidipati
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael F Beers
- Division of Pulmonary, Allergy, and Critical Care Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,The Penn Center for Pulmonary Biology, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Steven H Seeholzer
- Department of Pathology and Laboratory Medicine, Cell Pathology Division, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Edward E Morrisey
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,The Penn Center for Pulmonary Biology, Perelman School of Medicine, Philadelphia, Pennsylvania.,Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; and.,Penn Institute of Regenerative Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - G Scott Worthen
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; .,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,The Penn Center for Pulmonary Biology, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
259
|
Hu Y, Lou J, Mao YY, Lai TW, Liu LY, Zhu C, Zhang C, Liu J, Li YY, Zhang F, Li W, Ying SM, Chen ZH, Shen HH. Activation of MTOR in pulmonary epithelium promotes LPS-induced acute lung injury. Autophagy 2016; 12:2286-2299. [PMID: 27658023 DOI: 10.1080/15548627.2016.1230584] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MTOR (mechanistic target of rapamycin [serine/threonine kinase]) plays a crucial role in many major cellular processes including metabolism, proliferation and macroautophagy/autophagy induction, and is also implicated in a growing number of proliferative and metabolic diseases. Both MTOR and autophagy have been suggested to be involved in lung disorders, however, little is known about the role of MTOR and autophagy in pulmonary epithelium in the context of acute lung injury (ALI). In the present study, we observed that lipopolysaccharide (LPS) stimulation induced MTOR phosphorylation and decreased the expression of MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 β)-II, a hallmark of autophagy, in mouse lung epithelium and in human bronchial epithelial (HBE) cells. The activation of MTOR in HBE cells was mediated by TLR4 (toll-like receptor 4) signaling. Genetic knockdown of MTOR or overexpression of autophagy-related proteins significantly attenuated, whereas inhibition of autophagy further augmented, LPS-induced expression of IL6 (interleukin 6) and IL8, through NFKB signaling in HBE cells. Mice with specific knockdown of Mtor in bronchial or alveolar epithelial cells exhibited significantly attenuated airway inflammation, barrier disruption, and lung edema, and displayed prolonged survival in response to LPS exposure. Taken together, our results demonstrate that activation of MTOR in the epithelium promotes LPS-induced ALI, likely through downregulation of autophagy and the subsequent activation of NFKB. Thus, inhibition of MTOR in pulmonary epithelial cells may represent a novel therapeutic strategy for preventing ALI induced by certain bacteria.
Collapse
Affiliation(s)
- Yue Hu
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Jian Lou
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Yuan-Yuan Mao
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Tian-Wen Lai
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Li-Yao Liu
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Chen Zhu
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Chao Zhang
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Juan Liu
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Yu-Yan Li
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Fan Zhang
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Wen Li
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Song-Min Ying
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Zhi-Hua Chen
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China
| | - Hua-Hao Shen
- a Department of Respiratory and Critical Care Medicine , Second Affiliated Hospital of Zhejiang University School of Medicine , Hangzhou , China.,b State Key Lab of Respiratory Diseases , Guangzhou , China
| |
Collapse
|
260
|
Zhang J, Shao Y, He D, Zhang L, Xu G, Shen J. Evidence that bone marrow-derived mesenchymal stem cells reduce epithelial permeability following phosgene-induced acute lung injury via activation of wnt3a protein-induced canonical wnt/β-catenin signaling. Inhal Toxicol 2016; 28:572-579. [PMID: 27644345 DOI: 10.1080/08958378.2016.1228720] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Jing Zhang
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China, and
| | - Yiru Shao
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China, and
| | - Daikun He
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China, and
| | - Lin Zhang
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China, and
| | - Guoxiong Xu
- Department of Center Laboratory, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China,
- Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China, and
| |
Collapse
|
261
|
Scheraga RG, Thompson C, Tulapurkar ME, Nagarsekar AC, Cowan M, Potla R, Sun J, Cai R, Logun C, Shelhamer J, Todd NW, Singh IS, Luzina IG, Atamas SP, Hasday JD. Activation of heat shock response augments fibroblast growth factor-1 expression in wounded lung epithelium. Am J Physiol Lung Cell Mol Physiol 2016; 311:L941-L955. [PMID: 27638903 DOI: 10.1152/ajplung.00262.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/08/2016] [Indexed: 12/29/2022] Open
Abstract
We previously showed that coincident exposure to heat shock (HS; 42°C for 2 h) and TNF-α synergistically induces apoptosis in mouse lung epithelium. We extended this work by analyzing HS effects on human lung epithelial responses to clinically relevant injury. Cotreatment with TNF-α and HS induced little caspase-3 and poly(ADP-ribose) polymerase cleavage in human small airway epithelial cells, A549 cells, and BEAS2B cells. Scratch wound closure rates almost doubled when A549 and BEAS2B cells and air-liquid interface cultures of human bronchial epithelial cells were heat shocked immediately after wounding. Microarray, qRT-PCR, and immunoblotting showed fibroblast growth factor 1 (FGF1) to be synergistically induced by HS and wounding. Enhanced FGF1 expression in HS/wounded A549 was blocked by inhibitors of p38 MAPK (SB203580) or HS factor (HSF)-1 (KNK-437) and in HSF1 knockout BEAS2B cells. PCR demonstrated FGF1 to be expressed from the two most distal promoters in wounded/HS cells. Wound closure in HS A549 and BEAS2B cells was reduced by FGF receptor-1/3 inhibition (SU-5402) or FGF1 depletion. Exogenous FGF1 accelerated A549 wound closure in the absence but not presence of HS. In the presence of exogenous FGF1, HS slowed wound closure, suggesting that it increases FGF1 expression but impairs FGF1-stimulated wound closure. Frozen sections from normal and idiopathic pulmonary fibrosis (IPF) lung were analyzed for FGF1 and HSP70 by immunofluorescence confocal microscopy and qRT-PCR. FGF1 and HSP70 mRNA levels were 7.5- and 5.9-fold higher in IPF than normal lung, and the proteins colocalized to fibroblastic foci in IPF lung. We conclude that HS signaling may have an important impact on gene expression contributing to lung injury, healing, and fibrosis.
Collapse
Affiliation(s)
- Rachel G Scheraga
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | | | - Mohan E Tulapurkar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashish C Nagarsekar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mark Cowan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ratnakar Potla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Junfeng Sun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Rongman Cai
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Carolea Logun
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - James Shelhamer
- Critical Care Section, National Heart, Lung, Blood Institute, Bethesda, Maryland
| | - Nevins W Todd
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Ishwar S Singh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Irina G Luzina
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Sergei P Atamas
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| | - Jeffrey D Hasday
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; .,Medicine and Research Services, Baltimore Veterans Affairs Medical Care System, Baltimore, Maryland
| |
Collapse
|
262
|
Morin F, Kavian N, Nicco C, Cerles O, Chéreau C, Batteux F. Niclosamide Prevents Systemic Sclerosis in a Reactive Oxygen Species–Induced Mouse Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:3018-3028. [DOI: 10.4049/jimmunol.1502482] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 08/14/2016] [Indexed: 12/27/2022]
|
263
|
Gilpin SE, Charest JM, Ren X, Tapias LF, Wu T, Evangelista-Leite D, Mathisen DJ, Ott HC. Regenerative potential of human airway stem cells in lung epithelial engineering. Biomaterials 2016; 108:111-9. [PMID: 27622532 DOI: 10.1016/j.biomaterials.2016.08.055] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/25/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022]
Abstract
Bio-engineered organs for transplantation may ultimately provide a personalized solution for end-stage organ failure, without the risk of rejection. Building upon the process of whole organ perfusion decellularization, we aimed to develop novel, translational methods for the recellularization and regeneration of transplantable lung constructs. We first isolated a proliferative KRT5(+)TP63(+) basal epithelial stem cell population from human lung tissue and demonstrated expansion capacity in conventional 2D culture. We then repopulated acellular rat scaffolds in ex vivo whole organ culture and observed continued cell proliferation, in combination with primary pulmonary endothelial cells. To show clinical scalability, and to test the regenerative capacity of the basal cell population in a human context, we then recellularized and cultured isolated human lung scaffolds under biomimetic conditions. Analysis of the regenerated tissue constructs confirmed cell viability and sustained metabolic activity over 7 days of culture. Tissue analysis revealed extensive recellularization with organized tissue architecture and morphology, and preserved basal epithelial cell phenotype. The recellularized lung constructs displayed dynamic compliance and rudimentary gas exchange capacity. Our results underline the regenerative potential of patient-derived human airway stem cells in lung tissue engineering. We anticipate these advances to have clinically relevant implications for whole lung bioengineering and ex vivo organ repair.
Collapse
Affiliation(s)
- Sarah E Gilpin
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Jonathan M Charest
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Xi Ren
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Luis F Tapias
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Tong Wu
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Daniele Evangelista-Leite
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| | - Douglas J Mathisen
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States
| | - Harald C Ott
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Medical School, United States; Center for Regenerative Medicine, Massachusetts General Hospital, United States
| |
Collapse
|
264
|
Tsukamoto T, Yamamoto H, Okada S, Matano T. Recursion-based depletion of human immunodeficiency virus-specific naive CD4(+) T cells may facilitate persistent viral replication and chronic viraemia leading to acquired immunodeficiency syndrome. Med Hypotheses 2016; 94:81-85. [PMID: 27515208 DOI: 10.1016/j.mehy.2016.06.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 06/23/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
Although antiretroviral therapy has made human immunodeficiency virus (HIV) infection a controllable disease, it is still unclear how viral replication persists in untreated patients and causes CD4(+) T-cell depletion leading to acquired immunodeficiency syndrome (AIDS) in several years. Theorists tried to explain it with the diversity threshold theory in which accumulated mutations in the HIV genome make the virus so diverse that the immune system will no longer be able to recognize all the variants and fail to control the viraemia. Although the theory could apply to a number of cases, macaque AIDS models using simian immunodeficiency virus (SIV) have shown that failed viral control at the set point is not always associated with T-cell escape mutations. Moreover, even monkeys without a protective major histocompatibility complex (MHC) allele can contain replication of a super infected SIV following immunization with a live-attenuated SIV vaccine, while those animals are not capable of fighting primary SIV infection. Here we propose a recursion-based virus-specific naive CD4(+) T-cell depletion hypothesis through thinking on what may happen in individuals experiencing primary immunodeficiency virus infection. This could explain the mechanism for impairment of virus-specific immune response in the course of HIV infection.
Collapse
Affiliation(s)
| | - Hiroyuki Yamamoto
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Seiji Okada
- Center for AIDS Research, Kumamoto University, Kumamoto, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan; The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
265
|
Abstract
Innate lymphoid cells (ILCs), a newly identified member of the lymphoid population, play a critical role in the transition from innate to adaptive immunity in host defense. ILCs are important in mucosal barrier immunity, tissue homeostasis, and immune regulation throughout the body. Significant alterations in ILC responses in lung diseases have been observed and reported. Emerging evidence has shown that ILCs are importantly involved in the pathogenesis and development of a variety of lung diseases, i.e., helminth infections, allergic airway inflammation, and airway hyper-responsiveness. However, as a tissue-resident cell population, the role of ILCs in the lung remains poorly characterized. In this review, we discuss the role of ILCs in lung diseases, the mechanisms underlying the ILC-mediated regulation of immunity, and the therapeutic potential of modulating ILC responses.
Collapse
Affiliation(s)
- Deng-Ming Lai
- Department of Cardiovascular Surgery, the Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052 China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Qiang Shu
- Department of Cardiovascular Surgery, the Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310052 China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240 USA
| |
Collapse
|
266
|
Vladar EK, Nayak JV, Milla CE, Axelrod JD. Airway epithelial homeostasis and planar cell polarity signaling depend on multiciliated cell differentiation. JCI Insight 2016; 1. [PMID: 27570836 DOI: 10.1172/jci.insight.88027] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Motile airway cilia that propel contaminants out of the lung are oriented in a common direction by planar cell polarity (PCP) signaling, which localizes PCP protein complexes to opposite cell sides throughout the epithelium to orient cytoskeletal remodeling. In airway epithelia, PCP is determined in a 2-phase process. First, cell-cell communication via PCP complexes polarizes all cells with respect to the proximal-distal tissue axis. Second, during ciliogenesis, multiciliated cells (MCCs) undergo cytoskeletal remodeling to orient their cilia in the proximal direction. The second phase not only directs cilium polarization, but also consolidates polarization across the epithelium. Here, we demonstrate that in airway epithelia, PCP depends on MCC differentiation. PCP mutant epithelia have misaligned cilia, and also display defective barrier function and regeneration, indicating that PCP regulates multiple aspects of airway epithelial homeostasis. In humans, MCCs are often sparse in chronic inflammatory diseases, and these airways exhibit PCP dysfunction. The presence of insufficient MCCs impairs mucociliary clearance in part by disrupting PCP-driven polarization of the epithelium. Consistent with defective PCP, barrier function and regeneration are also disrupted. Pharmacological stimulation of MCC differentiation restores PCP and reverses these defects, suggesting its potential for broad therapeutic benefit in chronic inflammatory disease.
Collapse
Affiliation(s)
- Eszter K Vladar
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Jayakar V Nayak
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Carlos E Milla
- Division of Pulmonary Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Jeffrey D Axelrod
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
267
|
Yi NY, Newman DR, Zhang H, Morales Johansson H, Sannes PL. Heparin and LPS-induced COX-2 expression in airway cells: a link between its anti-inflammatory effects and GAG sulfation. Exp Lung Res 2016; 41:499-513. [PMID: 26495958 DOI: 10.3109/01902148.2015.1091053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE/AIM Previous studies have indicated that the sulfated polysaccharide heparin has anti-inflammatory effects. However, the mechanistic basis for these effects has not been fully elucidated. MATERIALS AND METHODS NCI-H292 (mucoepidermoid) and HBE-1 (normal) human bronchial epithelial cells were treated with LPS alone or in the presence of high-molecular-weight (HMW) fully sulfated heparin or desulfated HMW heparin. Cells were harvested to examine the phosphorylation levels of ERK1/2, p38, and NF-kB p65 and COX-2 protein expression by Western blot and gene expression of both COX-2 and CXCL-8 by TaqMan qRT-PCR. RESULTS Heparin is known to exert an influence on receptor-mediated signaling through its ability to both potentiate and inhibit the receptor-ligand interaction, depending upon its concentration. In H292 cells, fully-sulfated HMW heparin significantly reduced LPS-induced gene expression of both COX-2 and CXCL-8 for up to 48 hours, while desulfated heparin had little to no significant suppressive effect on signaling or on COX-2 gene or protein expression. Desulfated heparin, initially ineffective at preventing LPS-induced CXCL8 up-regulation, reduced CXCL8 transcription at 24 hours. In contrast, in normal HBE-1 cells, fully sulfated heparin significantly suppressed only ERK signaling, COX-2 gene expression at 12 hours, and CXCL-8 gene expression at 6 and 12 hours, while desulfated heparin had no significant effects on LPS-stimulated signaling or on gene or protein expression. Sulfation determines heparin's influence and may reflect the moderating role of GAG sulfation in lung injury and health. CONCLUSIONS Heparin's anti-inflammatory effects result from its nonspecific suppression of signaling and gene expression and are determined by its sulfation.
Collapse
Affiliation(s)
- Na Young Yi
- a Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh , North Carolina , USA
| | - Donna R Newman
- a Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh , North Carolina , USA
| | - Huiying Zhang
- a Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh , North Carolina , USA
| | - Helena Morales Johansson
- a Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh , North Carolina , USA
| | - Philip L Sannes
- a Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University , Raleigh , North Carolina , USA
| |
Collapse
|
268
|
Heijink IH, de Bruin HG, Dennebos R, Jonker MR, Noordhoek JA, Brandsma CA, van den Berge M, Postma DS. Cigarette smoke-induced epithelial expression of WNT-5B: implications for COPD. Eur Respir J 2016; 48:504-15. [PMID: 27126693 DOI: 10.1183/13993003.01541-2015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/10/2016] [Indexed: 02/04/2023]
Abstract
Wingless/integrase-1 (WNT) signalling is associated with lung inflammation and repair, but its role in chronic obstructive pulmonary disease (COPD) pathogenesis is unclear. We investigated whether cigarette smoke-induced dysregulation of WNT-5B contributes to airway remodelling in COPD.We analysed WNT-5B protein expression in the lung tissue of COPD patients and (non)smoking controls, and investigated the effects of cigarette smoke exposure on WNT-5B expression in COPD and control-derived primary bronchial epithelial cells (PBECs). Additionally, we studied downstream effects of WNT-5B on remodelling related genes fibronectin, matrix metalloproteinase (MMP)-2, MMP-9 and SnaiI in BEAS-2B and air-liquid interface (ALI)-cultured PBECs.We observed that airway epithelial WNT-5B expression is significantly higher in lung tissue from COPD patients than controls. Cigarette smoke extract significantly increased mRNA expression of WNT-5B in COPD, but not control-derived PBECs. Exogenously added WNT-5B augmented the expression of remodelling related genes in BEAS-2B cells, which was mediated by transforming growth factor (TGF)-β/Smad3 signalling. In addition, WNT-5B upregulated the expression of these genes in ALI-cultured PBECs, particularly PBECs from COPD patients.Together, our results provide evidence that exaggerated WNT-5B expression upon cigarette smoke exposure in the bronchial epithelium of COPD patients leads to TGF-β/Smad3-dependent expression of genes related to airway remodelling.
Collapse
Affiliation(s)
- Irene H Heijink
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| | - Harold G de Bruin
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Robin Dennebos
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Marnix R Jonker
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Jacobien A Noordhoek
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| | - Dirkje S Postma
- University of Groningen, University Medical Center Groningen, Dept of Pulmonology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen GRIAC Research Institute, Groningen, The Netherlands
| |
Collapse
|
269
|
Stone RC, Pastar I, Ojeh N, Chen V, Liu S, Garzon KI, Tomic-Canic M. Epithelial-mesenchymal transition in tissue repair and fibrosis. Cell Tissue Res 2016; 365:495-506. [PMID: 27461257 DOI: 10.1007/s00441-016-2464-0] [Citation(s) in RCA: 422] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/24/2016] [Indexed: 12/28/2022]
Abstract
The epithelial-mesenchymal transition (EMT) describes the global process by which stationary epithelial cells undergo phenotypic changes, including the loss of cell-cell adhesion and apical-basal polarity, and acquire mesenchymal characteristics that confer migratory capacity. EMT and its converse, MET (mesenchymal-epithelial transition), are integral stages of many physiologic processes and, as such, are tightly coordinated by a host of molecular regulators. Converging lines of evidence have identified EMT as a component of cutaneous wound healing, during which otherwise stationary keratinocytes (the resident skin epithelial cells) migrate across the wound bed to restore the epidermal barrier. Moreover, EMT plays a role in the development of scarring and fibrosis, as the matrix-producing myofibroblasts arise from cells of the epithelial lineage in response to injury but are pathologically sustained instead of undergoing MET or apoptosis. In this review, we summarize the role of EMT in physiologic repair and pathologic fibrosis of tissues and organs. We conclude that further investigation into the contribution of EMT to the faulty repair of fibrotic wounds might identify components of EMT signaling as common therapeutic targets for impaired healing in many tissues. Graphical Abstract Model for injury-triggered EMT activation in physiologic wound repair (left) and fibrotic wound healing (right).
Collapse
Affiliation(s)
- Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
- The Research Residency Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Fla., USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Nkemcho Ojeh
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
- Faculty of Medical Sciences, The University of the West Indies, Bridgetown, Barbados
| | - Vivien Chen
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Sophia Liu
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Karen I Garzon
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA.
| |
Collapse
|
270
|
Smirnova NF, Schamberger AC, Nayakanti S, Hatz R, Behr J, Eickelberg O. Detection and quantification of epithelial progenitor cell populations in human healthy and IPF lungs. Respir Res 2016; 17:83. [PMID: 27423691 PMCID: PMC4947297 DOI: 10.1186/s12931-016-0404-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/10/2016] [Indexed: 12/17/2022] Open
Abstract
Background In the human lung, epithelial progenitor cells in the airways give rise to the differentiated pseudostratified airway epithelium. In mice, emerging evidence confers a progenitor function to cytokeratin 5 (KRT5+) or cytokeratin 14 (KRT14+)-positive basal cells of the airway epithelium. Little is known, however, about the distribution of progenitor subpopulations in the human lung, particularly about aberrant epithelial differentiation in lung disease, such as idiopathic pulmonary fibrosis (IPF). Methods Here, we used multi-color immunofluorescence analysis to detect and quantify the distribution of airway epithelial progenitor subpopulations in human lungs obtained from healthy donors or IPF patients. Results In lungs from both, healthy donors and IPF patients, we detected KRT5+KRT14-, KRT5-KRT14+ and KRT5+KRT14+ populations in the proximal airways. KRT14+ cells, however, were absent in the distal airways of healthy lungs. In IPF, we detected a dramatic increase in the amount of KRT5+ cells and the emergence of a frequent KRT5+KRT14+ epithelial population, in particular in distal airways and alveolar regions. While the KRT14- progenitor population exhibited signs of proper epithelial differentiation, as evidenced by co-staining with pro-SPC, aquaporin 5, CC10, or MUC5B, the KRT14+ cell population did not co-stain with bronchial/alveolar differentiation markers in IPF. Conclusions We provide, for the first time, a quantitative profile of the distribution of epithelial progenitor populations in human lungs. We show compelling evidence for dysregulation and aberrant differentiation of these populations in IPF.
Collapse
Affiliation(s)
- N F Smirnova
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - A C Schamberger
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - S Nayakanti
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany
| | - R Hatz
- Thoraxchirurgisches Zentrum, Klinik für Allgemeine, Viszeral, Transplantations, Gefäß- und Thoraxchirurgie, Klinikum Großhadern, Ludwig-Maximilians-Universität, Munich, Germany.,Asklepios Fachkliniken München-Gauting, Munich, Germany.,German Center of Lung Research (DZL), Hannover, Germany
| | - J Behr
- Asklepios Fachkliniken München-Gauting, Munich, Germany.,Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians-Universität, Munich, Germany.,German Center of Lung Research (DZL), Hannover, Germany
| | - O Eickelberg
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, University Hospital, Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, 81377, Germany. .,German Center of Lung Research (DZL), Hannover, Germany. .,Comprehensive Pneumology Center (CPC), Ludwig-Maximilians-University and Helmholtz Zentrum München, Max-Lebsche-Platz 31, Munich, D-81377, Germany.
| |
Collapse
|
271
|
Jiang Z, Lao T, Qiu W, Polverino F, Gupta K, Guo F, Mancini JD, Naing ZZC, Cho MH, Castaldi PJ, Sun Y, Yu J, Laucho-Contreras ME, Kobzik L, Raby BA, Choi AMK, Perrella MA, Owen CA, Silverman EK, Zhou X. A Chronic Obstructive Pulmonary Disease Susceptibility Gene, FAM13A, Regulates Protein Stability of β-Catenin. Am J Respir Crit Care Med 2016; 194:185-97. [PMID: 26862784 PMCID: PMC5003213 DOI: 10.1164/rccm.201505-0999oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 01/21/2016] [Indexed: 12/28/2022] Open
Abstract
RATIONALE A genetic locus within the FAM13A gene has been consistently associated with chronic obstructive pulmonary disease (COPD) in genome-wide association studies. However, the mechanisms by which FAM13A contributes to COPD susceptibility are unknown. OBJECTIVES To determine the biologic function of FAM13A in human COPD and murine COPD models and discover the molecular mechanism by which FAM13A influences COPD susceptibility. METHODS Fam13a null mice (Fam13a(-/-)) were generated and exposed to cigarette smoke. The lung inflammatory response and airspace size were assessed in Fam13a(-/-) and Fam13a(+/+) littermate control mice. Cellular localization of FAM13A protein and mRNA levels of FAM13A in COPD lungs were assessed using immunofluorescence, Western blotting, and reverse transcriptase-polymerase chain reaction, respectively. Immunoprecipitation followed by mass spectrometry identified cellular proteins that interact with FAM13A to reveal insights on FAM13A's function. MEASUREMENTS AND MAIN RESULTS In murine and human lungs, FAM13A is expressed in airway and alveolar type II epithelial cells and macrophages. Fam13a null mice (Fam13a(-/-)) were resistant to chronic cigarette smoke-induced emphysema compared with Fam13a(+/+) mice. In vitro, FAM13A interacts with protein phosphatase 2A and recruits protein phosphatase 2A with glycogen synthase kinase 3β and β-catenin, inducing β-catenin degradation. Fam13a(-/-) mice were also resistant to elastase-induced emphysema, and this resistance was reversed by coadministration of a β-catenin inhibitor, suggesting that FAM13A could increase the susceptibility of mice to emphysema development by inhibiting β-catenin signaling. Moreover, human COPD lungs had decreased protein levels of β-catenin and increased protein levels of FAM13A. CONCLUSIONS We show that FAM13A may influence COPD susceptibility by promoting β-catenin degradation.
Collapse
Affiliation(s)
- Zhiqiang Jiang
- Channing Division of Network Medicine, Department of Medicine
| | - Taotao Lao
- Channing Division of Network Medicine, Department of Medicine
| | - Weiliang Qiu
- Channing Division of Network Medicine, Department of Medicine
| | - Francesca Polverino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Kushagra Gupta
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Feng Guo
- Channing Division of Network Medicine, Department of Medicine
| | - John D. Mancini
- Channing Division of Network Medicine, Department of Medicine
| | | | - Michael H. Cho
- Channing Division of Network Medicine, Department of Medicine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Peter J. Castaldi
- Channing Division of Network Medicine, Department of Medicine
- Division of General Internal Medicine, Department of Medicine, and
| | - Yang Sun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Jane Yu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Lester Kobzik
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | - Benjamin A. Raby
- Channing Division of Network Medicine, Department of Medicine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Mark A. Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Department of Medicine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| |
Collapse
|
272
|
Higuita-Castro N, Shukla VC, Mihai C, Ghadiali SN. Simvastatin Treatment Modulates Mechanically-Induced Injury and Inflammation in Respiratory Epithelial Cells. Ann Biomed Eng 2016; 44:3632-3644. [PMID: 27411707 DOI: 10.1007/s10439-016-1693-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/04/2016] [Indexed: 12/21/2022]
Abstract
Mechanical forces in the respiratory system, including surface tension forces during airway reopening and high transmural pressures, can result in epithelial cell injury, barrier disruption and inflammation. In this study, we investigated if a clinically relevant pharmaceutical agent, Simvastatin, could mitigate mechanically induced injury and inflammation in respiratory epithelia. Pulmonary alveolar epithelial cells (A549) were exposed to either cyclic airway reopening forces or oscillatory transmural pressure in vitro and treated with a wide range of Simvastatin concentrations. Simvastatin induced reversible depolymerization of the actin cytoskeleton and a statistically significant reduction the cell's elastic modulus. However, Simvastatin treatment did not result in an appreciable change in the cell's viscoelastic properties. Simvastatin treated cells did exhibit a reduced height-to-width aspect ratio and these changes in cell morphology resulted in a significant decrease in epithelial cell injury during airway reopening. Interestingly, although very high concentrations (25-50 µM) of Simvastatin resulted in dramatically less IL-6 and IL-8 pro-inflammatory cytokine secretion, 2.5 µM Simvastatin did not reduce the total amount of pro-inflammatory cytokines secreted during mechanical stimulation. These results indicate that although Simvastatin treatment may be useful in reducing cell injury during airway reopening, elevated local concentrations of Simvastatin might be needed to reduce mechanically-induced injury and inflammation in respiratory epithelia.
Collapse
Affiliation(s)
- N Higuita-Castro
- Biomedical Engineering Department, The Ohio State University, 270 Bevis Hall, 1080 Carmack Rd., Columbus, OH, 43221, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - V C Shukla
- Biomedical Engineering Department, The Ohio State University, 270 Bevis Hall, 1080 Carmack Rd., Columbus, OH, 43221, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - C Mihai
- Biomedical Engineering Department, The Ohio State University, 270 Bevis Hall, 1080 Carmack Rd., Columbus, OH, 43221, USA
| | - S N Ghadiali
- Biomedical Engineering Department, The Ohio State University, 270 Bevis Hall, 1080 Carmack Rd., Columbus, OH, 43221, USA. .,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
273
|
Warsinske HC, Wheaton AK, Kim KK, Linderman JJ, Moore BB, Kirschner DE. Computational Modeling Predicts Simultaneous Targeting of Fibroblasts and Epithelial Cells Is Necessary for Treatment of Pulmonary Fibrosis. Front Pharmacol 2016; 7:183. [PMID: 27445819 PMCID: PMC4917547 DOI: 10.3389/fphar.2016.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is pathologic remodeling of lung tissue that can result in difficulty breathing, reduced quality of life, and a poor prognosis for patients. Fibrosis occurs as a result of insult to lung tissue, though mechanisms of this response are not well-characterized. The disease is driven in part by dysregulation of fibroblast proliferation and differentiation into myofibroblast cells, as well as pro-fibrotic mediator-driven epithelial cell apoptosis. The most well-characterized pro-fibrotic mediator associated with pulmonary fibrosis is TGF-β1. Excessive synthesis of, and sensitivity to, pro-fibrotic mediators as well as insufficient production of and sensitivity to anti-fibrotic mediators has been credited with enabling fibroblast accumulation. Available treatments neither halt nor reverse lung damage. In this study we have two aims: to identify molecular and cellular scale mechanisms driving fibroblast proliferation and differentiation as well as epithelial cell survival in the context of fibrosis, and to predict therapeutic targets and strategies. We combine in vitro studies with a multi-scale hybrid agent-based computational model that describes fibroblasts and epithelial cells in co-culture. Within this model TGF-β1 represents a pro-fibrotic mediator and we include detailed dynamics of TGF-β1 receptor ligand signaling in fibroblasts. PGE2 represents an anti-fibrotic mediator. Using uncertainty and sensitivity analysis we identify TGF-β1 synthesis, TGF-β1 activation, and PGE2 synthesis among the key mechanisms contributing to fibrotic outcomes. We further demonstrate that intervention strategies combining potential therapeutics targeting both fibroblast regulation and epithelial cell survival can promote healthy tissue repair better than individual strategies. Combinations of existing drugs and compounds may provide significant improvements to the current standard of care for pulmonary fibrosis. Thus, a two-hit therapeutic intervention strategy may prove necessary to halt and reverse disease dynamics.
Collapse
Affiliation(s)
- Hayley C. Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Amanda K. Wheaton
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Kevin K. Kim
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | | | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|
274
|
Zhao H, Bian H, Bu X, Zhang S, Zhang P, Yu J, Lai X, Li D, Zhu C, Yao L, Su J. Targeting of Discoidin Domain Receptor 2 (DDR2) Prevents Myofibroblast Activation and Neovessel Formation During Pulmonary Fibrosis. Mol Ther 2016; 24:1734-1744. [PMID: 27350126 DOI: 10.1038/mt.2016.109] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 05/12/2016] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lethal human disease with short survival time and few treatment options. Herein, we demonstrated that discoidin domain receptor 2 (DDR2), a receptor tyrosine kinase that predominantly transduces signals from fibrillar collagens, plays a critical role in the induction of fibrosis and angiogenesis in the lung. In vitro cell studies showed that DDR2 can synergize the actions of both transforming growth factor (TGF)-β and fibrillar collagen to stimulate lung fibroblasts to undergo myofibroblastic changes and vascular endothelial growth factor (VEGF) expression. In addition, we confirmed that late treatment of the injured mice with specific siRNA against DDR2 or its kinase inhibitor exhibited therapeutic efficacy against lung fibrosis. Thus, this study not only elucidated novel mechanisms by which DDR2 controls the development of pulmonary fibrosis, but also provided candidate target for the intervention of this stubborn disease.
Collapse
Affiliation(s)
- Hu Zhao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Huan Bian
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xin Bu
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Shuya Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Pan Zhang
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiangtian Yu
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiaofeng Lai
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Di Li
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chuchao Zhu
- Department of Human Anatomy, Histology, and Embryology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Libo Yao
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jin Su
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, The Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
275
|
Schirm S, Ahnert P, Wienhold S, Mueller-Redetzky H, Nouailles-Kursar G, Loeffler M, Witzenrath M, Scholz M. A Biomathematical Model of Pneumococcal Lung Infection and Antibiotic Treatment in Mice. PLoS One 2016; 11:e0156047. [PMID: 27196107 PMCID: PMC4873198 DOI: 10.1371/journal.pone.0156047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/09/2016] [Indexed: 11/18/2022] Open
Abstract
Pneumonia is considered to be one of the leading causes of death worldwide. The outcome depends on both, proper antibiotic treatment and the effectivity of the immune response of the host. However, due to the complexity of the immunologic cascade initiated during infection, the latter cannot be predicted easily. We construct a biomathematical model of the murine immune response during infection with pneumococcus aiming at predicting the outcome of antibiotic treatment. The model consists of a number of non-linear ordinary differential equations describing dynamics of pneumococcal population, the inflammatory cytokine IL-6, neutrophils and macrophages fighting the infection and destruction of alveolar tissue due to pneumococcus. Equations were derived by translating known biological mechanisms and assuming certain response kinetics. Antibiotic therapy is modelled by a transient depletion of bacteria. Unknown model parameters were determined by fitting the predictions of the model to data sets derived from mice experiments of pneumococcal lung infection with and without antibiotic treatment. Time series of pneumococcal population, debris, neutrophils, activated epithelial cells, macrophages, monocytes and IL-6 serum concentrations were available for this purpose. The antibiotics Ampicillin and Moxifloxacin were considered. Parameter fittings resulted in a good agreement of model and data for all experimental scenarios. Identifiability of parameters is also estimated. The model can be used to predict the performance of alternative schedules of antibiotic treatment. We conclude that we established a biomathematical model of pneumococcal lung infection in mice allowing predictions regarding the outcome of different schedules of antibiotic treatment. We aim at translating the model to the human situation in the near future.
Collapse
Affiliation(s)
- Sibylle Schirm
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Sandra Wienhold
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Holger Mueller-Redetzky
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Geraldine Nouailles-Kursar
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Martin Witzenrath
- Department of Internal Medicine/Infectious Diseases and Respiratory Medicine Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Center of Civilization Diseases, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
276
|
Ruffin M, Bilodeau C, Maillé É, LaFayette SL, McKay GA, Trinh NTN, Beaudoin T, Desrosiers MY, Rousseau S, Nguyen D, Brochiero E. Quorum-sensing inhibition abrogates the deleterious impact of Pseudomonas aeruginosa on airway epithelial repair. FASEB J 2016; 30:3011-25. [PMID: 27178322 DOI: 10.1096/fj.201500166r] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/02/2016] [Indexed: 12/19/2022]
Abstract
Chronic Pseudomonas aeruginosa lung infections are associated with progressive epithelial damage and lung function decline. In addition to its role in tissue injury, the persistent presence of P. aeruginosa-secreted products may also affect epithelial repair ability, raising the need for new antivirulence therapies. The purpose of our study was to better understand the outcomes of P. aeruginosa exoproducts exposure on airway epithelial repair processes to identify a strategy to counteract their deleterious effect. We found that P. aeruginosa exoproducts significantly decreased wound healing, migration, and proliferation rates, and impaired the ability of directional migration of primary non-cystic fibrosis (CF) human airway epithelial cells. Impact of exoproducts was inhibited after mutations in P. aeruginosa genes that encoded for the quorum-sensing (QS) transcriptional regulator, LasR, and the elastase, LasB, whereas impact was restored by LasB induction in ΔlasR mutants. P. aeruginosa purified elastase also induced a significant decrease in non-CF epithelial repair, whereas protease inhibition with phosphoramidon prevented the effect of P. aeruginosa exoproducts. Furthermore, treatment of P. aeruginosa cultures with 4-hydroxy-2,5-dimethyl-3(2H)-furanone, a QS inhibitor, abrogated the negative impact of P. aeruginosa exoproducts on airway epithelial repair. Finally, we confirmed our findings in human airway epithelial cells from patients with CF, a disease featuring P. aeruginosa chronic respiratory infection. These data demonstrate that secreted proteases under the control of the LasR QS system impair airway epithelial repair and that QS inhibitors could be of benefit to counteract the deleterious effect of P. aeruginosa in infected patients.-Ruffin, M., Bilodeau, C., Maillé, É., LaFayette, S. L., McKay, G. A., Trinh, N. T. N., Beaudoin, T., Desrosiers, M.-Y., Rousseau, S., Nguyen, D., Brochiero, E. Quorum-sensing inhibition abrogates the deleterious impact of Pseudomonas aeruginosa on airway epithelial repair.
Collapse
Affiliation(s)
- Manon Ruffin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada; Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Claudia Bilodeau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada; Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Émilie Maillé
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Shantelle L LaFayette
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Geoffrey A McKay
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Nguyen Thu Ngan Trinh
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada; Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Trevor Beaudoin
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Martin-Yvon Desrosiers
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Dao Nguyen
- The Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada; Département de Médecine, Université de Montréal, Montréal, Québec, Canada;
| |
Collapse
|
277
|
Colby JK, Abdulnour REE, Sham HP, Dalli J, Colas RA, Winkler JW, Hellmann J, Wong B, Cui Y, El-Chemaly S, Petasis NA, Spite M, Serhan CN, Levy BD. Resolvin D3 and Aspirin-Triggered Resolvin D3 Are Protective for Injured Epithelia. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1801-1813. [PMID: 27171898 DOI: 10.1016/j.ajpath.2016.03.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 01/21/2016] [Accepted: 03/01/2016] [Indexed: 12/26/2022]
Abstract
Acute lung injury is a life-threatening condition caused by disruption of the alveolar-capillary barrier leading to edema, influx of inflammatory leukocytes, and impaired gas exchange. Specialized proresolving mediators biosynthesized from essential fatty acids, such as docosahexaenoic acid, have tissue protective effects in acute inflammation. Herein, we found that the docosahexaenoic acid-derived mediator resolvin D3 (RvD3): 4S,11R,17S-trihydroxydocosa-5Z,7E,9E,13Z,15E,19Z-hexaenoic acid was present in uninjured lungs, and increased significantly 24 to 72 hours after hydrochloric acid-initiated injury. Because of its delayed enzymatic degradation, we used aspirin-triggered (AT)-RvD3: 4S,11R,17R-trihydroxydocosa-5Z,7E,9E,13Z,15E,19Z-hexaenoic acid, a 17R-epimer of RvD3, for in vivo experiments. Histopathological correlates of acid injury (alveolar wall thickening, edema, and leukocyte infiltration) were reduced in mice receiving AT-RvD3 1 hour after injury. AT-RvD3-treated mice had significantly reduced edema, as demonstrated by lower wet/dry weight ratios, increased epithelial sodium channel γ expression, and more lymphatic vessel endothelial hyaluronan receptor 1-positive vascular endothelial growth factor receptor 3-positive lymphatic vessels. Evidence for counterregulation of NF-κB by RvD3 and AT-RvD3 was seen in vitro and by AT-RvD3 in vivo. Increases in lung epithelial cell proliferation and bronchoalveolar lavage fluid levels of keratinocyte growth factor were observed with AT-RvD3, which also promoted cutaneous re-epithelialization. Together, these data demonstrate protective actions of RvD3 and AT-RvD3 for injured mucosa that accelerated restoration of epithelial barrier and function.
Collapse
Affiliation(s)
- Jennifer K Colby
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raja-Elie E Abdulnour
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ho Pan Sham
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jesmond Dalli
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Romain A Colas
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jeremy W Winkler
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jason Hellmann
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Blenda Wong
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ye Cui
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Souheil El-Chemaly
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nicos A Petasis
- Department of Chemistry, Loker Hydrocarbon Research Institute, University of Southern California, Los Angeles, California
| | - Matthew Spite
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Charles N Serhan
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
278
|
Deeb RS, Hajjar DP. Repair Mechanisms in Oxidant-Driven Chronic Inflammatory Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1736-1749. [PMID: 27171899 DOI: 10.1016/j.ajpath.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 02/24/2016] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
The interplay that governs chronic diseases through pathways specifically associated with chronic inflammation remains undefined. Many metabolic events have been identified during the injury and repair process. Nonetheless, the cellular events that control the pathogenesis of inflammation-induced disease have not been fully characterized. We and others reason that chronic inflammatory diseases associated with a cascade of complex network mediators, such as nitric oxide, arachidonic acid metabolites, cytokines, and reactive oxygen species, play a significant role in the governance of alterations in homeostasis, oxidative stress, and thromboatherosclerosis. In this context, we discuss lipid mediators associated with the maintenance of health, including the specialized proresolving mediators that help drive cellular repair. Emphasis is placed on the pathophysiology of chronic metabolic insults involving both the airways and the cardiovascular system during oxidant-driven inflammatory disease. In this review, we highlight new pathways of inquiry that show promise for the identification of those metabolic targets that can improve therapy for chronic inflammation.
Collapse
Affiliation(s)
- Ruba S Deeb
- Department of Bioengineering, University of Bridgeport, Bridgeport, Connecticut.
| | - David P Hajjar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, New York.
| |
Collapse
|
279
|
Regulation of FGF signaling: Recent insights from studying positive and negative modulators. Semin Cell Dev Biol 2016; 53:101-14. [DOI: 10.1016/j.semcdb.2016.01.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/19/2016] [Indexed: 11/19/2022]
|
280
|
Robb CT, Regan KH, Dorward DA, Rossi AG. Key mechanisms governing resolution of lung inflammation. Semin Immunopathol 2016; 38:425-48. [PMID: 27116944 PMCID: PMC4896979 DOI: 10.1007/s00281-016-0560-6] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 04/14/2016] [Indexed: 12/11/2022]
Abstract
Innate immunity normally provides excellent defence against invading microorganisms. Acute inflammation is a form of innate immune defence and represents one of the primary responses to injury, infection and irritation, largely mediated by granulocyte effector cells such as neutrophils and eosinophils. Failure to remove an inflammatory stimulus (often resulting in failed resolution of inflammation) can lead to chronic inflammation resulting in tissue injury caused by high numbers of infiltrating activated granulocytes. Successful resolution of inflammation is dependent upon the removal of these cells. Under normal physiological conditions, apoptosis (programmed cell death) precedes phagocytic recognition and clearance of these cells by, for example, macrophages, dendritic and epithelial cells (a process known as efferocytosis). Inflammation contributes to immune defence within the respiratory mucosa (responsible for gas exchange) because lung epithelia are continuously exposed to a multiplicity of airborne pathogens, allergens and foreign particles. Failure to resolve inflammation within the respiratory mucosa is a major contributor of numerous lung diseases. This review will summarise the major mechanisms regulating lung inflammation, including key cellular interplays such as apoptotic cell clearance by alveolar macrophages and macrophage/neutrophil/epithelial cell interactions. The different acute and chronic inflammatory disease states caused by dysregulated/impaired resolution of lung inflammation will be discussed. Furthermore, the resolution of lung inflammation during neutrophil/eosinophil-dominant lung injury or enhanced resolution driven via pharmacological manipulation will also be considered.
Collapse
Affiliation(s)
- C T Robb
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - K H Regan
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - D A Dorward
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - A G Rossi
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
281
|
Role of mitochondrial hydrogen peroxide induced by intermittent hypoxia in airway epithelial wound repair in vitro. Exp Cell Res 2016; 344:143-151. [PMID: 27093911 DOI: 10.1016/j.yexcr.2016.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 12/19/2022]
Abstract
The airway epithelium acts as a frontline barrier against various environmental insults and its repair process after airway injury is critical for the lung homeostasis restoration. Recently, the role of intracellular reactive oxygen species (ROS) as transcription-independent damage signaling has been highlighted in the wound repair process. Both conditions of continuous hypoxia and intermittent hypoxia (IH) induce ROS. Although IH is important in clinical settings, the roles of IH-induced ROS in the airway repair process have not been investigated. In this study, we firstly showed that IH induced mitochondrial hydrogen peroxide (H2O2) production and significantly decreased bronchial epithelial cell migration, prevented by catalase treatment in a wound scratch assay. RhoA activity was higher during repair process in the IH condition compared to in the normoxic condition, resulting in the cellular morphological changes shown by immunofluorescence staining: round cells, reduced central stress fiber numbers, pronounced cortical actin filament distributions, and punctate focal adhesions. These phenotypes were replicated by exogenous H2O2 treatment under the normoxic condition. Our findings confirmed the transcription-independent role of IH-induced intracellular ROS in the bronchial epithelial cell repair process and might have significant implications for impaired bronchial epithelial cell regeneration.
Collapse
|
282
|
Hamilton JR, Sachs D, Lim JK, Langlois RA, Palese P, Heaton NS. Club cells surviving influenza A virus infection induce temporary nonspecific antiviral immunity. Proc Natl Acad Sci U S A 2016; 113:3861-6. [PMID: 27001854 PMCID: PMC4833272 DOI: 10.1073/pnas.1522376113] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A brief window of antigen-nonspecific protection has been observed after influenza A virus (IAV) infection. Although this temporary immunity has been assumed to be the result of residual nonspecific inflammation, this period of induced immunity has not been fully studied. Because IAV has long been characterized as a cytopathic virus (based on its ability to rapidly lyse most cell types in culture), it has been a forgone conclusion that directly infected cells could not be contributing to this effect. Using a Cre recombinase-expressing IAV, we have previously shown that club cells can survive direct viral infection. We show here not only that these cells can eliminate all traces of the virus and survive but also that they acquire a heightened antiviral response phenotype after surviving. Moreover, we experimentally demonstrate temporary nonspecific viral immunity after IAV infection and show that surviving cells are required for this phenotype. This work characterizes a virally induced modulation of the innate immune response that may represent a new mechanism to prevent viral diseases.
Collapse
Affiliation(s)
- Jennifer R Hamilton
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - David Sachs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ryan A Langlois
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Department of Microbiology and Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Nicholas S Heaton
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710
| |
Collapse
|
283
|
Umazume T, Thomas WM, Campbell S, Aluri H, Thotakura S, Zoukhri D, Makarenkova HP. Lacrimal Gland Inflammation Deregulates Extracellular Matrix Remodeling and Alters Molecular Signature of Epithelial Stem/Progenitor Cells. Invest Ophthalmol Vis Sci 2016; 56:8392-402. [PMID: 26747770 DOI: 10.1167/iovs.15-17477] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE The adult lacrimal gland (LG) is highly regenerative and is able to repair itself even after substantial damage; however, this ability to regenerate is lost with the development of dry eye conditions in chronically inflamed LGs.This study compares changes in the cell adhesion and cell matrix molecules and stem cell transcription factors in the LGs of healthy mice and of two mouse models of Sjögren's syndrome: nonobese diabetic (NOD) and MRL-lpr/lpr (MRL/lpr) mice during the early stage of inflammation. METHODS The LGs from 12- to 13-week-old female MRL/lpr and male NOD mice along with their respective control strains were harvested and divided into three pieces and processed for quantitative (q) RT-PCR and qRT-PCR Arrays, histology, immunohistochemistry, and Western blotting. RESULTS The extracellular matrix (ECM) and adhesion molecules RT2-PCR array combined with protein expression data revealed changes in the expression of integrins, matrix metalloproteinases, and other molecules, which are associated largely with invasion, attachment, and expansion of the lymphocytic cells, whereas changes in the stem cell transcription factors revealed substantial decrease in expression of transcription factors associated with epithelial stem/progenitor cell lineage. CONCLUSIONS We concluded that the expression of several important ECM components is significantly deregulated in the LG of two murine models of Sjögren's syndrome, suggesting an alteration of the epithelial stem/progenitor cell niche. This may result in profound effects on localization, activation, proliferation, and differentiation of the LG stem/progenitor cells and, therefore, LG regeneration.
Collapse
Affiliation(s)
- Takeshi Umazume
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| | - William M Thomas
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| | - Sabrina Campbell
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| | - Hema Aluri
- Department of Diagnosis and Health Promotion, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Suharika Thotakura
- Department of Diagnosis and Health Promotion, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Driss Zoukhri
- Department of Diagnosis and Health Promotion, Tufts University School of Dental Medicine, Boston, Massachusetts, United States
| | - Helen P Makarenkova
- Department of Cell and Molecular Biology The Scripps Research Institute, La Jolla, California, United States
| |
Collapse
|
284
|
RAGE and TGF-β1 Cross-Talk Regulate Extracellular Matrix Turnover and Cytokine Synthesis in AGEs Exposed Fibroblast Cells. PLoS One 2016; 11:e0152376. [PMID: 27015414 PMCID: PMC4807770 DOI: 10.1371/journal.pone.0152376] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
AGEs accumulation in the skin affects extracellular matrix (ECM) turnover and triggers diabetes associated skin conditions and accelerated skin aging. The receptor of AGEs (RAGE) has an essential contribution to cellular dysfunction driven by chronic inflammatory responses while TGF-β1 is critical in both dermal homeostasis and inflammation. We investigated the contribution of RAGE and TGF-β1 to the modulation of inflammatory response and ECM turnover in AGEs milieu, using a normal fibroblast cell line. RAGE, TGF-β1, collagen I and III gene and protein expression were upregulated after exposure to AGEs-BSA, and MMP-2 was activated. AGEs-RAGE was pivotal in NF-κB dependent collagen I expression and joined with TGF-β1 to stimulate collagen III expression, probably via ERK1/2 signaling. AGEs-RAGE axis induced upregulation of TGF-β1, TNF-α and IL-8 cytokines. TNF-α and IL-8 were subjected to TGF-β1 negative regulation. RAGE’s proinflammatory signaling also antagonized AGEs-TGF-β1 induced fibroblast contraction, suggesting the existence of an inhibitory cross-talk mechanism between TGF-β1 and RAGE signaling. RAGE and TGF-β1 stimulated anti-inflammatory cytokines IL-2 and IL-4 expression. GM-CSF and IL-6 expression appeared to be dependent only on TGF-β1 signaling. Our data also indicated that IFN-γ upregulated in AGEs-BSA milieu in a RAGE and TGF-β1 independent mechanism. Our findings raise the possibility that RAGE and TGF-β1 are both involved in fibrosis development in a complex cross-talk mechanism, while also acting on their own individual targets. This study contributes to the understanding of impaired wound healing associated with diabetes complications.
Collapse
|
285
|
Li J, Huang S, Zhang J, Feng C, Gao D, Yao B, Wu X, Fu X. Mesenchymal stem cells ameliorate inflammatory cytokine-induced impairment of AT-II cells through a keratinocyte growth factor-dependent PI3K/Akt/mTOR signaling pathway. Mol Med Rep 2016; 13:3755-62. [PMID: 27035760 PMCID: PMC4838139 DOI: 10.3892/mmr.2016.5004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 02/04/2016] [Indexed: 01/18/2023] Open
Abstract
Lung epithelium restoration subsequent to injury is of concern in association with the outcomes of diverse inflammatory lung diseases. Previous studies have demonstrated that mesenchymal stem cells (MSCs) may promote epithelial repair subsequent to inflammatory injury, however the mechanism that mediates this effect remains unclear. The current study examined the role of MSCs in alveolar type II epithelial cell (AT-II cell) restoration subsequent to an inflammatory insult. AT-II cells were firstly exposed to inflammatory cytokines including tumor necrosis factor-α, interleukin (IL)-6 and IL-1β, then were co-cultured with MSCs in Transwell for 72 h. Cell proliferation, expression of surfactant protein A (SP-A) and expression of the α1 subunit were evaluated respectively by the Cell Counting Kit-8 assay, western blotting and semiquantitative reverse transcription-polymerase chain reaction. Keratinocyte growth factor (KGF) small interfering RNA (siRNA) was applied to knockdown the main cytoprotective factors in the MSCs. Subsequent to an inflammatory insult, AT-II cells were observed to be impaired, exhibiting the characteristics of injured cell morphology, reduced cell proliferation and reduced expression of SP-A and the α1 subunit. Co-culture with MSCs significantly ameliorated these cell impairments, while these benefits were weakened by the application of KGF siRNA. Simultaneously, expression levels of phosphorylated (p-) protein kinase B (AKT) and p-mammalian target of rapamycin (mTOR) in AT-II cells were upregulated by MSCs, suggesting activation of the phosphoinositide 3-kinase (PI3K) pathway. These data demonstrate that administration of MSCs to the inflammation-insulted AT-II cells may ameliorate the impairments through a KGF-dependent PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Sha Huang
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| | - Junhua Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Changjiang Feng
- Department of Thoracic and Cardiovascular Surgery, Peking University People's Hospital, Peking University, Beijing 100044, P.R. China
| | - Dongyun Gao
- Department of Oncology, Dongtai People's Hospital, Dongtai, Jiangsu 224200, P.R. China
| | - Bin Yao
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| | - Xu Wu
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaobing Fu
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| |
Collapse
|
286
|
Altered Profile of Circulating Endothelial-Derived Microparticles in Ventilator-Induced Lung Injury. Crit Care Med 2016; 43:e551-9. [PMID: 26308427 DOI: 10.1097/ccm.0000000000001280] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Pulmonary endothelial cell injury is central to the pathophysiology of acute lung injury. Mechanical ventilation can cause endothelial disruption and injury, even in the absence of preexisting inflammation. Platelet-endothelial cell adhesion molecule-1 is a transmembrane protein connecting adjacent endothelial cells. We hypothesized that injurious mechanical ventilation will increase circulating lung endothelial-derived microparticles, defined as microparticles positive for platelet-endothelial cell adhesion molecule-1, which could serve as potential biomarkers and mediators of ventilator-induced lung injury. DESIGN Prospective randomized, controlled, animal investigation. SETTING A hospital preclinical animal laboratory. SUBJECTS Forty-eight Sprague-Dawley rats. INTERVENTIONS Animals were randomly allocated to one of the three following ventilatory protocols for 4 hours: spontaneous breathing (control group), mechanical ventilation with low tidal volume (6 mL/kg), and mechanical ventilation with high tidal volume (20 mL/kg). In both mechanical ventilation groups, positive end-expiratory pressure of 2 cm H2O was applied. MEASUREMENTS AND MAIN RESULTS We analyzed histologic lung damage, gas exchange, wet-to-dry lung weight ratio, serum cytokines levels, circulating endothelial-derived microparticles, platelet-endothelial cell adhesion molecule-1 lung protein content, and immunohistochemistry. When compared with low-tidal volume mechanical ventilation, high-tidal volume ventilation increased lung edema score and caused gas-exchange deterioration. These changes were associated with a marked increased of circulating endothelial-derived microparticles and a reduction of platelet-endothelial cell adhesion molecule-1 protein levels in the high-tidal volume lungs (p < 0.0001). CONCLUSIONS There is an endothelial-derived microparticle profile associated with disease-specific features of ventilator-induced lung injury. This profile could serve both as a biomarker of acute lung injury and, potentially, as a mediator of systemic propagation of pulmonary inflammatory response.
Collapse
|
287
|
Ma Y, Huang W, Liu C, Li Y, Xia Y, Yang X, Sun W, Bai H, Li Q, Peng Z. Immunization against TGF-β1 reduces collagen deposition but increases sustained inflammation in a murine asthma model. Hum Vaccin Immunother 2016; 12:1876-85. [PMID: 26901684 DOI: 10.1080/21645515.2016.1145849] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor (TGF)-β1 is involved in the processes of airway inflammation and remodeling; however, its reported roles in asthma pathogenesis are controversial. We sought both to investigate the effects of active immunization targeting TGF-β1 on allergen-induced airway inflammatory responses and to evaluate its possible application for asthma treatment. BALB/c mice were immunized with a virus-like-particle (VLP) vaccine presenting a TGF-β1 peptide. For the preventive intervention of acute allergic airway inflammation, immunization was conducted before sensitization and challenges with ovalbumin (OVA), and for the therapeutic treatment of chronic inflammatory responses, immunization was initiated after inflammatory responses were established. Preventive immunization with VLPs led to increased proinflammatory IL-4, IL-13, and IL-33 levels in the bronchoalveolar lavage fluids (BALF) with no significant effects on lung tissue inflammation and airway goblet cell hyperplasia. Therapeutic treatment showed that at 24 h after the fourth 2-day challenge with OVA following 2 intraperitoneal sensitizations, airway subepithelial collagen deposition was significantly ameliorated in vaccinated mice, whereas the lung histology and cytokine profile in the BALF were not changed. In contrast, after a 4-week recovery from the last OVA challenge, the vaccinated mice's collagen deposition remained reduced, but they sustained lung-tissue inflammation and goblet-cell hyperplasia; elevated IL-13, TNF, and IFN-γ levels in the BALF; and increased airway resistance, tissue resistance, and tissue elastance. In a conclusion, the role of TGF-β1 is complicated in allergic airway inflammatory responses. It is important to make a careful assessment in accordance with specific disease conditions when targeting TGF-β1 for a therapeutic purpose.
Collapse
Affiliation(s)
- Yanbing Ma
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Weiwei Huang
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Cunbao Liu
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Yang Li
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Ye Xia
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Xu Yang
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Wenjia Sun
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Hongmei Bai
- a Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Kunming , China
| | - Qihan Li
- b Department of Viral Immunology , Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Zhikang Peng
- c Department of Pediatrics and Child Health , University of Manitoba , Winnipeg , Canada
| |
Collapse
|
288
|
Grzela K, Litwiniuk M, Zagorska W, Grzela T. Airway Remodeling in Chronic Obstructive Pulmonary Disease and Asthma: the Role of Matrix Metalloproteinase-9. Arch Immunol Ther Exp (Warsz) 2016; 64:47-55. [PMID: 26123447 PMCID: PMC4713715 DOI: 10.1007/s00005-015-0345-y] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 02/23/2015] [Indexed: 01/06/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) and asthma are both associated with airflow restriction and progressive remodeling, which affect the respiratory tract. Among various biological factors involved in the pathomechanisms of both diseases, proteolytic enzymes--matrix metalloproteinases (MMPs)--play an important role, especially MMP-9. In this review, the authors discuss the current topics of research concerning the possible role of MMP-9 in both mentioned diseases. They include the analysis of protein levels, nucleotide polymorphisms of MMP-9 gene and their possible correlation with asthma and COPD. Finally, the authors refer to the studies on MMP-9 inhibition as a new perspective for increasing the effectiveness of treatment in asthma and COPD.
Collapse
Affiliation(s)
- Katarzyna Grzela
- Department of Paediatrics, Pneumonology and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Litwiniuk
- Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004, Warsaw, Poland
- Potgraduate School of Molecular Medicine, Warsaw, Poland
| | - Wioletta Zagorska
- Department of Paediatrics, Pneumonology and Allergology, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Grzela
- Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004, Warsaw, Poland.
| |
Collapse
|
289
|
Iosifidis T, Garratt LW, Coombe DR, Knight DA, Stick SM, Kicic A. Airway epithelial repair in health and disease: Orchestrator or simply a player? Respirology 2016; 21:438-48. [PMID: 26804630 DOI: 10.1111/resp.12731] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/01/2015] [Accepted: 12/03/2015] [Indexed: 12/21/2022]
Abstract
Epithelial cells represent the most important surface of contact in the body and form the first line of defence of the body to external environment. Consequently, epithelia have numerous roles in order to maintain a homeostatic defence barrier. Although the epithelium has been extensively studied over several decades, it remains the focus of new research, indicating a lack of understanding that continues to exist around these cells in specific disease settings. Importantly, evidence is emerging that airway epithelial cells in particular have varied complex functions rather than simple passive roles. One area of current interest is its role following injury. In particular, the epithelial-specific cellular mechanisms regulating their migration during wound repair remain poorly understood and remain an area that requires much needed investigation. A better understanding of the physiological, cellular and molecular wound repair mechanisms could assist in elucidating pathological processes that contribute to airway epithelial pathology. This review attempts to highlight migration-specific and cell-extracellular matrix (ECM) aspects of repair used by epithelial cells under normal and disease settings, in the context of human airways.
Collapse
Affiliation(s)
- Thomas Iosifidis
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | - Luke W Garratt
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Deirdre R Coombe
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,School of Biomedical Science and Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Stephen M Stick
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Anthony Kicic
- School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| |
Collapse
|
290
|
Broekman W, Amatngalim GD, de Mooij-Eijk Y, Oostendorp J, Roelofs H, Taube C, Stolk J, Hiemstra PS. TNF-α and IL-1β-activated human mesenchymal stromal cells increase airway epithelial wound healing in vitro via activation of the epidermal growth factor receptor. Respir Res 2016; 17:3. [PMID: 26753875 PMCID: PMC4710048 DOI: 10.1186/s12931-015-0316-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are investigated for their potential to reduce inflammation and to repair damaged tissue. Inflammation and tissue damage are hallmarks of chronic obstructive pulmonary disease (COPD) and MSC infusion is a promising new treatment for COPD. Inflammatory mediators attract MSCs to sites of inflammation and affect their immune-modulatory properties, but little is known about their effect on regenerative properties of MSCs. This study investigates the effect of the pro-inflammatory cytokines TNF-α and IL-1β on the regenerative potential of MSCs, using an in vitro wound healing model of airway epithelial cells. Methods Standardized circular wounds were created by scraping cultures of the airway epithelial cell line NCI-H292 and primary bronchial epithelial cells cultured at the air-liquid interface (ALI-PBEC), and subsequently incubated with MSC conditioned medium (MSC-CM) that was generated in presence or absence of TNF-α/IL-1β. Remaining wound size was measured up to 72 h. Phosphorylation of ERK1/2 by MSC-CM was assessed using Western blot. Inhibitors for EGFR and c-Met signaling were used to investigate the contribution of these receptors to wound closure and to ERK1/2 phosphorylation. Transactivation of EGFR by MSC-CM was investigated using a TACE inhibitor, and RT-PCR was used to quantify mRNA expression of several growth factors in MSCs and NCI-H292. Results Stimulation of MSCs with the pro-inflammatory cytokines TNF-α and IL-1β increased the mRNA expression of various growth factors by MCSs and enhanced the regenerative potential of MSCs in an in vitro model of airway epithelial injury using NCI-H292 airway epithelial cells. Conditioned medium from cytokine stimulated MSCs induced ERK1/2 phosphorylation in NCI-H292, predominantly via EGFR; it induced ADAM-mediated transactivation of EGFR, and it induced airway epithelial expression of several EGFR ligands. The contribution of activation of c-Met via HGF to increased repair could not be confirmed by inhibitor experiments. Conclusion Our data imply that at sites of tissue damage, when inflammatory mediators are present, for example in lungs of COPD patients, MSCs become more potent inducers of repair, in addition to their well-known immune-modulatory properties.
Collapse
Affiliation(s)
- Winifred Broekman
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Gimano D Amatngalim
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Yvonne de Mooij-Eijk
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jaap Oostendorp
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Helene Roelofs
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
291
|
Kim SH. Airway epithelial cells in airway inflammation and remodeling in asthma. ALLERGY ASTHMA & RESPIRATORY DISEASE 2016. [DOI: 10.4168/aard.2016.4.2.82] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
292
|
Im D, Shi W, Driscoll B. Pediatric Acute Respiratory Distress Syndrome: Fibrosis versus Repair. Front Pediatr 2016; 4:28. [PMID: 27066462 PMCID: PMC4811965 DOI: 10.3389/fped.2016.00028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/15/2016] [Indexed: 01/11/2023] Open
Abstract
Clinical and basic experimental approaches to pediatric acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), have historically focused on acute care and management of the patient. Additional efforts have focused on the etiology of pediatric ALI and ARDS, clinically defined as diffuse, bilateral diseases of the lung that compromise function leading to severe hypoxemia within 7 days of defined insult. Insults can include ancillary events related to prematurity, can follow trauma and/or transfusion, or can present as sequelae of pulmonary infections and cardiovascular disease and/or injury. Pediatric ALI/ARDS remains one of the leading causes of infant and childhood morbidity and mortality, particularly in the developing world. Though incidence is relatively low, ranging from 2.9 to 9.5 cases/100,000 patients/year, mortality remains high, approaching 35% in some studies. However, this is a significant decrease from the historical mortality rate of over 50%. Several decades of advances in acute management and treatment, as well as better understanding of approaches to ventilation, oxygenation, and surfactant regulation have contributed to improvements in patient recovery. As such, there is a burgeoning interest in the long-term impact of pediatric ALI/ARDS. Chronic pulmonary deficiencies in survivors appear to be caused by inappropriate injury repair, with fibrosis and predisposition to emphysema arising as irreversible secondary events that can severely compromise pulmonary development and function, as well as the overall health of the patient. In this chapter, the long-term effectiveness of current treatments will be examined, as will the potential efficacy of novel, acute, and long-term therapies that support repair and delay or even impede the onset of secondary events, including fibrosis.
Collapse
Affiliation(s)
- Daniel Im
- Pediatric Critical Care Medicine, Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California , Los Angeles, CA , USA
| | - Wei Shi
- Developmental Biology and Regenerative Medicine Program, Department of Surgery, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California , Los Angeles, CA , USA
| | - Barbara Driscoll
- Developmental Biology and Regenerative Medicine Program, Department of Surgery, The Saban Research Institute, Children's Hospital Los Angeles, University of Southern California , Los Angeles, CA , USA
| |
Collapse
|
293
|
Chen YB, Lan YW, Chen LG, Huang TT, Choo KB, Cheng WTK, Lee HS, Chong KY. Mesenchymal stem cell-based HSP70 promoter-driven VEGFA induction by resveratrol alleviates elastase-induced emphysema in a mouse model. Cell Stress Chaperones 2015; 20:979-89. [PMID: 26243699 PMCID: PMC4595438 DOI: 10.1007/s12192-015-0627-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/01/2015] [Accepted: 07/19/2015] [Indexed: 01/03/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a sustained blockage of the airways due to lung inflammation occurring with chronic bronchitis and/or emphysema. Progression of emphysema may be slowed by vascular endothelial growth factor A (VEGFA), which reduces apoptotic tissue depletion. Previously, authors of the present report demonstrated that cis-resveratrol (c-RSV)-induced heat-shock protein 70 (HSP70) promoter-regulated VEGFA expression promoted neovascularization of genetically modified mesenchymal stem cells (HSP-VEGFA-MSC) in a mouse model of ischemic disease. Here, this same stem cell line was evaluated for its protective capacity to alleviate elastase-induced pulmonary emphysema in mice. Results of this study showed that c-RSV-treatment of HSP-VEGFA-MSC exhibited synergy between HSP70 transcription activity and induced expression of anti-oxidant-related genes when challenged by cigarette smoke extracts. Eight weeks after jugular vein injection of HSP-VEGFA-MSC into mice with elastase-induced pulmonary emphysema followed by c-RSV treatment to induce transgene expression, significant improvement was observed in respiratory functions. Expression of VEGFA, endogenous nuclear factor erythroid 2-related factor (Nrf 2), and manganese superoxide dismutase (MnSOD) was significantly increased in the lung tissues of the c-RSV-treated mice. Histopathologic examination of treated mice revealed gradual but significant abatement of emphysema and restoration of airspace volume. In conclusion, the present investigation demonstrates that c-RSV-regulated VEGFA expression in HSP-VEGFA-MSC significantly improved the therapeutic effects on the treatment of COPD in the mouse, possibly avoiding side effects associated with constitutive VEGFA expression.
Collapse
Affiliation(s)
- Young-Bin Chen
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Ying-Wei Lan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Lih-Geeng Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi, 600, Taiwan, Republic of China
| | - Tsung-Teng Huang
- Center for Molecular and Clinical Immunology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Kong-Bung Choo
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences and Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Winston T K Cheng
- Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan, Republic of China
| | - Hsuan-Shu Lee
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan, Republic of China.
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicne, Taipei, Taiwan, Republic of China.
| | - Kowit-Yu Chong
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Family Medicine, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan, Republic of China.
| |
Collapse
|
294
|
Peitzman ER, Zaidman NA, Maniak PJ, O'Grady SM. Agonist binding to β-adrenergic receptors on human airway epithelial cells inhibits migration and wound repair. Am J Physiol Cell Physiol 2015; 309:C847-55. [PMID: 26491049 DOI: 10.1152/ajpcell.00159.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/19/2015] [Indexed: 12/30/2022]
Abstract
Human airway epithelial cells express β-adrenergic receptors (β-ARs), which regulate mucociliary clearance by stimulating transepithelial anion transport and ciliary beat frequency. Previous studies using airway epithelial cells showed that stimulation with isoproterenol increased cell migration and wound repair by a cAMP-dependent mechanism. In the present study, impedance-sensing arrays were used to measure cell migration and epithelial restitution following wounding of confluent normal human bronchial epithelial (NHBE) and Calu-3 cells by electroporation. Stimulation with epinephrine or the β2-AR-selective agonist salbutamol significantly delayed wound closure and reduced the mean surface area of lamellipodia protruding into the wound. Treatment with the β-AR bias agonist carvedilol or isoetharine also produced a delay in epithelial restitution similar in magnitude to epinephrine and salbutamol. Measurements of extracellular signal-regulated kinase phosphorylation following salbutamol or carvedilol stimulation showed no significant change in the level of phosphorylation compared with untreated control cells. However, inhibition of protein phosphatase 2A activity completely blocked the delay in wound closure produced by β-AR agonists. In Calu-3 cells, where CFTR expression was inhibited by RNAi, salbutamol did not inhibit wound repair, suggesting that β-AR agonist stimulation and loss of CFTR function share a common pathway leading to inhibition of epithelial repair. Confocal images of the basal membrane of Calu-3 cells labeled with anti-β1-integrin (clone HUTS-4) antibody showed that treatment with epinephrine or carvedilol reduced the level of activated integrin in the membrane. These findings suggest that treatment with β-AR agonists delays airway epithelial repair by a G protein- and cAMP-independent mechanism involving protein phosphatase 2A and a reduction in β1-integrin activation in the basal membrane.
Collapse
Affiliation(s)
| | - Nathan A Zaidman
- Department of Integrative Biology and Physiology, University of Minnesota, St. Paul, Minnesota
| | - Peter J Maniak
- Department of Animal Science, University of Minnesota, St. Paul, Minnesota; and
| | - Scott M O'Grady
- Department of Animal Science, University of Minnesota, St. Paul, Minnesota; and Department of Integrative Biology and Physiology, University of Minnesota, St. Paul, Minnesota
| |
Collapse
|
295
|
Breakdown of Epithelial Barrier Integrity and Overdrive Activation of Alveolar Epithelial Cells in the Pathogenesis of Acute Respiratory Distress Syndrome and Lung Fibrosis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:573210. [PMID: 26523279 PMCID: PMC4615219 DOI: 10.1155/2015/573210] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/05/2015] [Accepted: 09/15/2015] [Indexed: 12/29/2022]
Abstract
Individual alveolar epithelial cells (AECs) collaboratively form a tight barrier between atmosphere and fluid-filled tissue to enable normal gas exchange. The tight junctions of AECs provide intercellular sealing and are integral to the maintenance of the AEC barrier integrity. Disruption and failure of reconstitution of AEC barrier result in catastrophic consequences, leading to alveolar flooding and subsequent devastating fibrotic scarring. Recent evidences reveal that many of the fibrotic lung diseases involve AECs both as a frequent target of injury and as a driver of ongoing pathological processes. Aberrantly activated AECs express most of the growth factors and chemokines responsible for the proliferation, migration, and activation of fibroblasts. Current evidences suggest that AECs may acquire overdrive activation in the initial step of fibrosis by several mechanisms, including abnormal recapitulation of the developmental pathway, defects of the molecules essential for epithelial integrity, and acceleration of aging-related properties. Among these initial triggering events, epithelial Pten, a multiple phosphatase that negatively regulates the PI3K/Akt pathway and is crucial for lung development, is essential for the prevention of alveolar flooding and lung fibrosis through the regulation of AEC barrier integrity after injury. Reestablishment of AEC barrier integrity also involves the deployment of specialized stem/progenitor cells.
Collapse
|
296
|
Furuya Y, Furuya AKM, Roberts S, Sanfilippo AM, Salmon SL, Metzger DW. Prevention of Influenza Virus-Induced Immunopathology by TGF-β Produced during Allergic Asthma. PLoS Pathog 2015; 11:e1005180. [PMID: 26407325 PMCID: PMC4583434 DOI: 10.1371/journal.ppat.1005180] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 09/01/2015] [Indexed: 12/14/2022] Open
Abstract
Asthma is believed to be a risk factor for influenza infection, however little experimental evidence exists to directly demonstrate the impact of asthma on susceptibility to influenza infection. Using a mouse model, we now report that asthmatic mice are actually significantly more resistant to a lethal influenza virus challenge. Notably, the observed increased resistance was not attributable to enhanced viral clearance, but instead, was due to reduced lung inflammation. Asthmatic mice exhibited a significantly reduced cytokine storm, as well as reduced total protein levels and cytotoxicity in the airways, indicators of decreased tissue injury. Further, asthmatic mice had significantly increased levels of TGF-β1 and the heightened resistance of asthmatic mice was abrogated in the absence of TGF-β receptor II. We conclude that a transient increase in TGF-β expression following acute asthma can induce protection against influenza-induced immunopathology. Influenza and asthma represent the two major lung diseases in humans. While most studies have focused on exacerbation of asthma symptoms by influenza virus infection, the effects of asthma on susceptibility to influenza virus infections has been far less studied. Using a novel mouse model of asthma and influenza infection, we show that asthmatic mice are highly resistant to primary challenge with the 2009 influenza pandemic strain (CA04) compared to non-asthmatic mice. The increased resistance of asthmatic mice is not due to the enhanced T or B cell immunity but rather, to a strong anti-inflammatory TGF-beta response triggered by asthma. This study is the first to provide a mechanistic explanation for asthma-mediated protection during the 2009 influenza pandemic.
Collapse
Affiliation(s)
- Yoichi Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Andrea K. M. Furuya
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Sean Roberts
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Alan M. Sanfilippo
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Sharon L. Salmon
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
| | - Dennis W. Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
297
|
Platelet-derived Wnt antagonist Dickkopf-1 is implicated in ICAM-1/VCAM-1-mediated neutrophilic acute lung inflammation. Blood 2015; 126:2220-9. [PMID: 26351298 DOI: 10.1182/blood-2015-02-622233] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/20/2015] [Indexed: 12/14/2022] Open
Abstract
Neutrophil infiltration represents the early acute inflammatory response in acute lung injury. The recruitment of neutrophils from the peripheral blood across the endothelial-epithelial barrier into the alveolar airspace is highly regulated by the adhesion molecules on alveolar epithelial cells (AECs). Wnt/β-catenin signaling is involved in the progression of inflammatory lung diseases including asthma, emphysema, and pulmonary fibrosis. However, the function of Wnt/β-catenin signaling in acute lung inflammation is unknown. Here, we identified platelet-derived Dickkopf-1 (Dkk1) as the major Wnt antagonist contributing to the suppression of Wnt/β-catenin signaling in AECs during acute lung inflammation. Intratracheal administration of Wnt3a or an antibody capable of neutralizing Dkk1 inhibited neutrophil influx into the alveolar airspace of injured lungs. Activation of Wnt/β-catenin signaling in AECs attenuated intercellular adhesion molecule 1 (ICAM-1)/vascular cell adhesion molecule 1 (VCAM-1)-mediated adhesion of both macrophages and neutrophils to AECs. Our results suggest a role for Wnt/β-catenin signaling in modulating the inflammatory response, and a functional communication between platelets and AECs during acute lung inflammation. Targeting Wnt/β-catenin signaling and the communication between platelets and AECs therefore represents potential therapeutic strategies to limit the damage of acute pulmonary inflammation.
Collapse
|
298
|
Girault A, Chebli J, Privé A, Trinh NTN, Maillé E, Grygorczyk R, Brochiero E. Complementary roles of KCa3.1 channels and β1-integrin during alveolar epithelial repair. Respir Res 2015; 16:100. [PMID: 26335442 PMCID: PMC4558634 DOI: 10.1186/s12931-015-0263-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 08/21/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Extensive alveolar epithelial injury and remodelling is a common feature of acute lung injury and acute respiratory distress syndrome (ARDS) and it has been established that epithelial regeneration, and secondary lung oedema resorption, is crucial for ARDS resolution. Much evidence indicates that K(+) channels are regulating epithelial repair processes; however, involvement of the KCa3.1 channels in alveolar repair has never been investigated before. RESULTS Wound-healing assays demonstrated that the repair rates were increased in primary rat alveolar cell monolayers grown on a fibronectin matrix compared to non-coated supports, whereas an anti-β1-integrin antibody reduced it. KCa3.1 inhibition/silencing impaired the fibronectin-stimulated wound-healing rates, as well as cell migration and proliferation, but had no effect in the absence of coating. We then evaluated a putative relationship between KCa3.1 channel and the migratory machinery protein β1-integrin, which is activated by fibronectin. Co-immunoprecipitation and immunofluorescence experiments indicated a link between the two proteins and revealed their cellular co-distribution. In addition, we demonstrated that KCa3.1 channel and β1-integrin membrane expressions were increased on a fibronectin matrix. We also showed increased intracellular calcium concentrations as well as enhanced expression of TRPC4, a voltage-independent calcium channel belonging to the large TRP channel family, on a fibronectin matrix. Finally, wound-healing assays showed additive effects of KCa3.1 and TRPC4 inhibitors on alveolar epithelial repair. CONCLUSION Taken together, our data demonstrate for the first time complementary roles of KCa3.1 and TRPC4 channels with extracellular matrix and β1-integrin in the regulation of alveolar repair processes.
Collapse
Affiliation(s)
- Alban Girault
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada. .,Département de médecine, Université de Montréal, CP6128, Succursale Centre-ville, Montréal, Québec, H3C3J7, Canada.
| | - Jasmine Chebli
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada. .,Département de médecine, Université de Montréal, CP6128, Succursale Centre-ville, Montréal, Québec, H3C3J7, Canada.
| | - Anik Privé
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada.
| | - Nguyen Thu Ngan Trinh
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada. .,Département de médecine, Université de Montréal, CP6128, Succursale Centre-ville, Montréal, Québec, H3C3J7, Canada.
| | - Emilie Maillé
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada.
| | - Ryszard Grygorczyk
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada. .,Département de médecine, Université de Montréal, CP6128, Succursale Centre-ville, Montréal, Québec, H3C3J7, Canada.
| | - Emmanuelle Brochiero
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Tour Viger, 900 rue Saint-Denis, Montréal, Québec, H2X0A9, Canada. .,Département de médecine, Université de Montréal, CP6128, Succursale Centre-ville, Montréal, Québec, H3C3J7, Canada.
| |
Collapse
|
299
|
Yucesoy B, Kashon ML, Johnson VJ, Lummus ZL, Fluharty K, Gautrin D, Cartier A, Boulet LP, Sastre J, Quirce S, Tarlo SM, Cruz MJ, Munoz X, Luster MI, Bernstein DI. Genetic variants in TNFα, TGFB1, PTGS1 and PTGS2 genes are associated with diisocyanate-induced asthma. J Immunotoxicol 2015; 13:119-26. [PMID: 25721048 DOI: 10.3109/1547691x.2015.1017061] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Diisocyanates are the most common cause of occupational asthma, but risk factors are not well defined. A case-control study was conducted to investigate whether genetic variants in inflammatory response genes (TNFα, IL1α, IL1β, IL1RN, IL10, TGFB1, ADAM33, ALOX-5, PTGS1, PTGS2 and NAG-1/GDF15) are associated with increased susceptibility to diisocyanate asthma (DA). These genes were selected based on their role in asthmatic inflammatory processes and previously reported associations with asthma phenotypes. The main study population consisted of 237 Caucasian French Canadians from among a larger sample of 280 diisocyanate-exposed workers in two groups: workers with specific inhalation challenge (SIC) confirmed DA (DA(+), n = 95) and asymptomatic exposed workers (AW, n = 142). Genotyping was performed on genomic DNA, using a 5' nuclease PCR assay. After adjusting for potentially confounding variables of age, smoking status and duration of exposure, the PTGS1 rs5788 and TGFB1 rs1800469 single nucleotide polymorphisms (SNP) showed a protective effect under a dominant model (OR = 0.38; 95% CI = 0.17, 0.89 and OR = 0.38; 95% CI = 0.18, 0.74, respectively) while the TNFα rs1800629 SNP was associated with an increased risk of DA (OR = 2.08; 95% CI = 1.03, 4.17). Additionally, the PTGS2 rs20417 variant showed an association with increased risk of DA in a recessive genetic model (OR = 6.40; 95% CI = 1.06, 38.75). These results suggest that genetic variations in TNFα, TGFB1, PTGS1 and PTGS2 genes contribute to DA susceptibility.
Collapse
Affiliation(s)
- Berran Yucesoy
- a Division of Immunology , Allergy and Rheumatology, University of Cincinnati College of Medicine , Cincinnati , OH , USA .,b CDC/National Institute for Occupational Safety and Health, Health Effects Laboratory Division , Morgantown , WV , USA
| | - Michael L Kashon
- b CDC/National Institute for Occupational Safety and Health, Health Effects Laboratory Division , Morgantown , WV , USA
| | | | - Zana L Lummus
- a Division of Immunology , Allergy and Rheumatology, University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Kara Fluharty
- b CDC/National Institute for Occupational Safety and Health, Health Effects Laboratory Division , Morgantown , WV , USA
| | - Denyse Gautrin
- d Université de Montréal, Hôpital du Sacré-Coeur de Montréal , Montreal , Quebec , Canada
| | - André Cartier
- d Université de Montréal, Hôpital du Sacré-Coeur de Montréal , Montreal , Quebec , Canada
| | | | - Joaquin Sastre
- f Department of Allergy , Fundación Jiménez Díaz and CIBER de Enfermedades Respiratorias CIBERES , Madrid , Spain
| | - Santiago Quirce
- g Department of Allergy , Hospital La Paz-IdiPAZ and CIBER de Enfermedades Respiratorias CIBERES , Madrid , Spain
| | - Susan M Tarlo
- h Department of Medicine , and.,i Dalla Lana School of Public Health, University of Toronto , Toronto , Ontario , Canada
| | - Maria-Jesus Cruz
- j Hospitals Vall D'Hebron, Barcelona and CIBER de Enfermedades Respiratorias CIBERES , Madrid , Spain , and
| | - Xavier Munoz
- j Hospitals Vall D'Hebron, Barcelona and CIBER de Enfermedades Respiratorias CIBERES , Madrid , Spain , and
| | - Michael I Luster
- k West Virginia University, School of Public Health , Morgantown , WV , USA
| | - David I Bernstein
- a Division of Immunology , Allergy and Rheumatology, University of Cincinnati College of Medicine , Cincinnati , OH , USA
| |
Collapse
|
300
|
Silasi-Mansat R, Zhu H, Georgescu C, Popescu N, Keshari RS, Peer G, Lupu C, Taylor FB, Pereira HA, Kinasewitz G, Lambris JD, Lupu F. Complement inhibition decreases early fibrogenic events in the lung of septic baboons. J Cell Mol Med 2015; 19:2549-63. [PMID: 26337158 PMCID: PMC4627561 DOI: 10.1111/jcmm.12667] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/03/2015] [Indexed: 01/09/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) induced by severe sepsis can trigger persistent inflammation and fibrosis. We have shown that experimental sepsis in baboons recapitulates ARDS progression in humans, including chronic inflammation and long-lasting fibrosis in the lung. Complement activation products may contribute to the fibroproliferative response, suggesting that complement inhibitors are potential therapeutic agents. We have been suggested that treatment of septic baboons with compstatin, a C3 convertase inhibitor protects against ARDS-induced fibroproliferation. Baboons challenged with 109 cfu/kg (LD50) live E. coli by intravenous infusion were treated or not with compstatin at the time of challenge or 5 hrs thereafter. Changes in the fibroproliferative response at 24 hrs post-challenge were analysed at both transcript and protein levels. Gene expression analysis showed that sepsis induced fibrotic responses in the lung as early as 24 hrs post-bacterial challenge. Immunochemical and biochemical analysis revealed enhanced collagen synthesis, induction of profibrotic factors and increased cell recruitment and proliferation. Specific inhibition of complement with compstatin down-regulated sepsis-induced fibrosis genes, including transforming growth factor-beta (TGF-β), connective tissue growth factor (CTGF), tissue inhibitor of metalloproteinase 1 (TIMP1), various collagens and chemokines responsible for fibrocyte recruitment (e.g. chemokine (C-C motif) ligand 2 (CCL2) and 12 (CCL12)). Compstatin decreased the accumulation of myofibroblasts and proliferating cells, reduced the production of fibrosis mediators (TGF-β, phospho-Smad-2 and CTGF) and inhibited collagen deposition. Our data demonstrate that complement inhibition effectively attenuates collagen deposition and fibrotic responses in the lung after severe sepsis. Inhibiting complement could prove an attractive strategy for preventing sepsis-induced fibrosis of the lung.
Collapse
Affiliation(s)
- Robert Silasi-Mansat
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hua Zhu
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Constantin Georgescu
- Programs in Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Narcis Popescu
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ravi S Keshari
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Glenn Peer
- Department of Medicine, Pulmonary and Critical Care Division, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Cristina Lupu
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Fletcher B Taylor
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Heloise Anne Pereira
- Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA.,Department of Pharmaceutical Sciences, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA.,Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Gary Kinasewitz
- Department of Medicine, Pulmonary and Critical Care Division, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Florea Lupu
- Programs in Cardiovascular Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.,Department of Pathology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA.,Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|