1
|
Portelli MA, Bhaker S, Pang V, Bates DO, Johnson SR, Mazar AP, Shaw D, Brightling C, Sayers I. Elevated PLAUR is observed in the airway epithelium of asthma patients and blocking improves barrier integrity. Clin Transl Allergy 2023; 13:e12293. [PMID: 37876037 PMCID: PMC10542610 DOI: 10.1002/clt2.12293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Expression of the urokinase plasminogen activator receptor (uPAR) is elevated in the airway epithelium in asthma; however, the contribution of uPAR to asthma pathogenesis and scope for therapeutic targeting remains unknown. OBJECTIVES To determine (i) the expression profile of uPAR in cultured human bronchial epithelial cells (HBEC) from asthma patients, (ii) the relationship between uPAR and the epithelial barrier, including blocking uPAR functions and (iii) the function of different uPAR isoforms. METHODS uPAR levels in HBECs isolated from asthma patients and cells at air liquid interface (ALI) during differentiation were quantified. Transepithelial electrical resistance or electrical cell impedance sensing was used to relate uPAR levels to barrier properties, including effects of uPAR blocking antibodies. The functional effects of gain of function was determined using transcriptomics, in cells over-expressing membrane (muPAR), soluble cleaved (scuPAR) or soluble spliced (ssuPAR) isoforms. RESULTS Elevated expression of uPAR was a feature of cultured HBECs from asthma patients, suggesting intrinsic alterations in asthma patient cells. Soluble uPAR levels inversely correlated with barrier properties of the HBEC layer in 2D and ALI. Blocking uPAR-integrin interactions enhanced barrier formation. The gain of function cells showed limited transcriptomic changes. CONCLUSION This study provides a significant advance in our understanding of the relationship between asthma, uPAR and the epithelial barrier, where elevated circulating uPAR results in a reduced cell barrier, a phenotype prevalent in asthma.
Collapse
Affiliation(s)
- Michael A. Portelli
- Centre for Respiratory ResearchNIHR Respiratory Biomedical Research CentreSchool of MedicineBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - Sangita Bhaker
- Centre for Respiratory ResearchNIHR Respiratory Biomedical Research CentreSchool of MedicineBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - Vincent Pang
- Tumour Vascular Biology GroupBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - David O. Bates
- Tumour Vascular Biology GroupBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - Simon R. Johnson
- Centre for Respiratory ResearchNIHR Respiratory Biomedical Research CentreSchool of MedicineBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - Andrew P. Mazar
- Department of PharmacologyFeinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| | - Dominick Shaw
- Centre for Respiratory ResearchNIHR Respiratory Biomedical Research CentreSchool of MedicineBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| | - Christopher Brightling
- Department of Respiratory MedicineUniversity of LeicesterUniversity Hospitals of Leicester NHS TrustLeicesterUK
| | - Ian Sayers
- Centre for Respiratory ResearchNIHR Respiratory Biomedical Research CentreSchool of MedicineBiodiscovery InstituteUniversity ParkUniversity of NottinghamNottinghamUK
| |
Collapse
|
2
|
Namba S, Konuma T, Wu KH, Zhou W, Okada Y. A practical guideline of genomics-driven drug discovery in the era of global biobank meta-analysis. CELL GENOMICS 2022; 2:100190. [PMID: 36778001 PMCID: PMC9903693 DOI: 10.1016/j.xgen.2022.100190] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/27/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
Genomics-driven drug discovery is indispensable for accelerating the development of novel therapeutic targets. However, the drug discovery framework based on evidence from genome-wide association studies (GWASs) has not been established, especially for cross-population GWAS meta-analysis. Here, we introduce a practical guideline for genomics-driven drug discovery for cross-population meta-analysis, as lessons from the Global Biobank Meta-analysis Initiative (GBMI). Our drug discovery framework encompassed three methodologies and was applied to the 13 common diseases targeted by GBMI (N mean = 1,329,242). Individual methodologies complementarily prioritized drugs and drug targets, which were systematically validated by referring previously known drug-disease relationships. Integration of the three methodologies provided a comprehensive catalog of candidate drugs for repositioning, nominating promising drug candidates targeting the genes involved in the coagulation process for venous thromboembolism and the interleukin-4 and interleukin-13 signaling pathway for gout. Our study highlighted key factors for successful genomics-driven drug discovery using cross-population meta-analyses.
Collapse
Affiliation(s)
- Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
| | - Takahiro Konuma
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
| | - Kuan-Han Wu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Global Biobank Meta-analysis Initiative
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., Takatsuki 569-1125, Japan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita 565-0871, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita 565-0871, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita 565-0871, Japan
| |
Collapse
|
3
|
Womble JT, McQuade VL, Ihrie MD, Ingram JL. Imbalanced Coagulation in the Airway of Type-2 High Asthma with Comorbid Obesity. J Asthma Allergy 2021; 14:967-980. [PMID: 34408442 PMCID: PMC8364356 DOI: 10.2147/jaa.s318017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Asthma is a common, chronic airway inflammatory disease marked by airway hyperresponsiveness, inflammation, and remodeling. Asthma incidence has increased rapidly in the past few decades and recent multicenter analyses have revealed several unique asthma endotypes. Of these, type-2 high asthma with comorbid obesity presents a unique clinical challenge marked by increased resistance to standard therapies and exacerbated disease development. The extrinsic coagulation pathway plays a significant role in both type-2 high asthma and obesity. The type-2 high asthma airway is marked by increased procoagulant potential, which is readily activated following damage to airway tissue. In this review, we summarize the current understanding of the role the extrinsic coagulation pathway plays in the airway of type-2 high asthma with comorbid obesity. We propose that asthma control is worsened in obesity as a result of a systemic and local airway shift towards a procoagulant and anti-fibrinolytic environment. Lastly, we hypothesize bariatric surgery as a treatment for improved asthma management in type-2 high asthma with comorbid obesity, facilitated by normalization of systemic procoagulant and pro-inflammatory mediators. A better understanding of attenuated coagulation parameters in the airway following bariatric surgery will advance our knowledge of biomolecular pathways driving asthma pathobiology in patients with obesity.
Collapse
Affiliation(s)
- Jack T Womble
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Victoria L McQuade
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Mark D Ihrie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Jennifer L Ingram
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| |
Collapse
|
4
|
A Synthetic Curcuminoid Analogue, 2,6-Bis-4-(Hydroxyl-3-Methoxybenzylidine)-Cyclohexanone (BHMC) Ameliorates Acute Airway Inflammation of Allergic Asthma in Ovalbumin-Sensitized Mice. Mediators Inflamm 2021; 2021:9725903. [PMID: 33883974 PMCID: PMC8041524 DOI: 10.1155/2021/9725903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 02/23/2021] [Accepted: 03/18/2021] [Indexed: 01/19/2023] Open
Abstract
2,6-Bis-(4-hydroxyl-3-methoxybenzylidine) cyclohexanone (BHMC), a synthetic curcuminoid analogue, has been shown to exhibit anti-inflammatory properties in cellular models of inflammation and improve the survival of mice from lethal sepsis. We further evaluated the therapeutic effect of BHMC on acute airway inflammation in a mouse model of allergic asthma. Mice were sensitized and challenged with ovalbumin (OVA), followed by intraperitoneal administration of 0.1, 1, and 10 mg/kg of BHMC. Bronchoalveolar lavage fluid, blood, and lung samples were collected, and the respiratory function was measured. OVA sensitization and challenge increased airway hyperresponsiveness (AHR) and pulmonary inflammation. All three doses of BHMC (0.1-10 mg/kg) significantly reduced the number of eosinophils, lymphocytes, macrophages, and neutrophils, as well as the levels of Th2 cytokines (IL-4, IL-5 and IL-13) in bronchoalveolar lavage fluid (BALF) as compared to OVA-challenged mice. However, serum level of IgE was not affected. All three doses of BHMC (0.1-10 mg/kg) were effective in suppressing the infiltration of inflammatory cells at the peribronchial and perivascular regions, with the greatest effect observed at 1 mg/kg which was comparable to dexamethasone. Goblet cell hyperplasia was inhibited by 1 and 10 mg/kg of BHMC, while the lowest dose (0.1 mg/kg) had no significant inhibitory effect. These findings demonstrate that BHMC, a synthetic nonsteroidal small molecule, ameliorates acute airway inflammation associated with allergic asthma, primarily by suppressing the release of inflammatory mediators and goblet cell hyperplasia to a lesser extent in acute airway inflammation of allergic asthma.
Collapse
|
5
|
Feng Z, Liu S, Chen Q, Tan Q, Xian J, Feng H, Chen Z, Li G. uPA alleviates kaolin-induced hydrocephalus by promoting the release and activation of hepatocyte growth factor in rats. Neurosci Lett 2020; 731:135011. [PMID: 32497735 DOI: 10.1016/j.neulet.2020.135011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 02/05/2023]
Abstract
Urokinase-type plasminogen activator (uPA) was demonstrated to alleviate kaolin-induced communicating hydrocephalus via inhibiting subarachnoid space fibrosis, but the exact mechanism remains elusive. Thus, this study was designed to investigate if hepatocyte growth factor (HGF), which plays a vital role in uPA-triggered inhibiting of fibrosis in multiple systems, is involved in this process in hydrocephalus. There were 2 parts in this study. First, hydrocephalus was induced in rats by basal cistern injection of kaolin. Then rats were treated with saline or uPA and brain tissue and CSF were collected for Western blot and enzyme-linked immuno sorbent assay (ELISA) four days later. Second, kaolin-induced hydrocephalus rats were treated with saline, uPA, uPA + PHA665752 (antagonist of HGF) or PHA665752. Some animals received MRI four weeks later and brains were used for immunofluorescence. The others were euthanized four days later for ELISA. Both levels of total and activated HGF in the CSF was increased after uPA injections, but related mRNA expression of HGF showed no statistical significance when compared with the control group. Further, the effects of uPA that alleviating ventricular enlargement, subarachnoid fibrosis and reactive astrocytosis were partially reversed by PHA665752. Moreover, PHA665752 partially abolished uPA-induced reduction of transforming growth factor- β1(TGF- β1) level in CSF. Our data suggest that uPA effectively inhibited subarachnoid fibrosis and restricted the development of communicating hydrocephalus in rats in part by promoting HGF release and activation, which may further regulate the TGF-β1 expression in CSF.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Shengyan Liu
- Chongqing Mental Health Center, Chongqing, 4001147, PR China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jishu Xian
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Gang Li
- Department of Cerebrovascular Disease, Affiliated Hospital of Zunyi Medical University, Zunyi Medical University, Guizhou, 563003, PR China.
| |
Collapse
|
6
|
Hiroyasu S, Turner CT, Richardson KC, Granville DJ. Proteases in Pemphigoid Diseases. Front Immunol 2019; 10:1454. [PMID: 31297118 PMCID: PMC6607946 DOI: 10.3389/fimmu.2019.01454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/10/2019] [Indexed: 12/28/2022] Open
Abstract
Pemphigoid diseases are a subgroup of autoimmune skin diseases characterized by widespread tense blisters. Standard of care typically involves immunosuppressive treatments, which may be insufficient and are often associated with significant adverse events. As such, a deeper understanding of the pathomechanism(s) of pemphigoid diseases is necessary in order to identify improved therapeutic approaches. A major initiator of pemphigoid diseases is the accumulation of autoantibodies against proteins at the dermal-epidermal junction (DEJ), followed by protease activation at the lesion. The contribution of proteases to pemphigoid disease pathogenesis has been investigated using a combination of in vitro and in vivo models. These studies suggest proteolytic degradation of anchoring proteins proximal to the DEJ is crucial for dermal-epidermal separation and blister formation. In addition, proteases can also augment inflammation, expose autoantigenic cryptic epitopes, and/or provoke autoantigen spreading, which are all important in pemphigoid disease pathology. The present review summarizes and critically evaluates the current understanding with respect to the role of proteases in pemphigoid diseases.
Collapse
Affiliation(s)
- Sho Hiroyasu
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Christopher T. Turner
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Katlyn C. Richardson
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
7
|
Hur J, Kang JY, Rhee CK, Kim YK, Lee SY. The leukotriene receptor antagonist pranlukast attenuates airway remodeling by suppressing TGF-β signaling. Pulm Pharmacol Ther 2017; 48:5-14. [PMID: 29031615 DOI: 10.1016/j.pupt.2017.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/27/2017] [Accepted: 10/12/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND/OBJECTIVE Asthma is a chronic airway disease characterized by airway eosinophilic inflammation and remodeling, which are associated with a loss in lung function. Although both contribute significantly to asthma pathogenesis, mechanistic studies and drug discovery have focused on inflammatory targets. In this study, we investigated the effect of the leukotriene receptor antagonist pranlukast on allergic airway inflammation and remodeling in vivo and in vitro. METHOD Four groups of female BALB/c mice (control; ovalbumin [OVA]-sensitized and -challenged; dimethyl sulfoxide [DMSO]-treated OVA; and pranlukast-treated OVA) were examined. Lung pathology, cytokine production, and airway hyperresponsiveness (AHR) measurements were compared among these groups. A human fetal lung fibroblast HFL-1 cell line was used in the peribranchial fibrosis analysis. RESULTS OVA-sensitized and -challenged mice exhibited allergic airway inflammation and significant increases in Th2 cytokines. Pranlukast-treated mice showed significant attenuation of allergic airway inflammation. The pranlukast treatment decreased AHR and attenuated airway remodeling to goblet cell hyperplasia, collagen deposition, α-smooth muscle actin expression, and pro-fibrotic gene expression. We further demonstrated that pranlukast not only inhibited transforming growth factor-beta 1 (TGF-β1)-induced Smad signaling in human fetal lung fibroblast cells but also simultaneously reduced collagen synthesis and pro-fibrotic gene expression. CONCLUSIONS The leukotriene receptor antagonist pranlukast can reduce airway inflammation and remodeling by inhibiting TGF-β/Smad signaling in an OVA-sensitized and -challenged asthma mouse model, thus suppressing AHR.
Collapse
Affiliation(s)
- Jung Hur
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Young Kang
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chin Kook Rhee
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Kyoon Kim
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sook Young Lee
- Division of Allergy and Pulmonary Medicine, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Feng Z, Tan Q, Tang J, Li L, Tao Y, Chen Y, Yang Y, Luo C, Feng H, Zhu G, Chen Q, Chen Z. Intraventricular administration of urokinase as a novel therapeutic approach for communicating hydrocephalus. Transl Res 2017; 180:77-90.e2. [PMID: 27614013 DOI: 10.1016/j.trsl.2016.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/18/2016] [Accepted: 08/13/2016] [Indexed: 01/10/2023]
Abstract
Fibrosis of the subarachnoid space (SAS) after infection, inflammation, or hemorrhage can impair cerebrospinal fluid absorption and circulation, causing diffuse ventricular dilatation. In the present study, we tested the hypothesis that urokinase (also known as urokinase-type plasminogen activator [uPA]), a fibrinolytic agent, attenuates fibrosis and ventriculomegaly in a rat model of kaolin-induced communicating hydrocephalus and thus may have potential as a therapy for these conditions. Thirty microliters of sterile 25% kaolin suspension was injected into the basal cisterns of adult Sprague-Dawley rats to induce hydrocephalus, and 2 intraventricular injections of either uPA or vehicle (saline) were administered immediately and 3 days thereafter. Ventricular volumes were measured by magnetic resonance imaging (MRI) on days 3, 14, and 28 after kaolin injection. Fibrosis and reactive astrogliosis were evaluated on day 28 by immunofluorescence and Western blotting. Neurocognitive features were tested using the Morris water maze from days 23 to 28. MRI analysis demonstrated that kaolin administration successfully induced hydrocephalus in rats and that uPA treatment significantly attenuated ventricular enlargement. In addition, uPA inhibited the deposition of laminin and fibronectin, extracellular matrix molecules, in the SAS, attenuated gliosis, and improved learning and memory in kaolin-treated rats. Therefore, we concluded that uPA prevents the development of kaolin-induced communicating hydrocephalus by preventing the development of subarachnoid fibrosis and by eliciting improvements in neurocognition. The results of this study indicate that uPA may be a novel clinical therapy for communicating hydrocephalus.
Collapse
Affiliation(s)
- Zhou Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Lin Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yihao Tao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Yunfeng Yang
- Department of Neurosurgery, Sichuan Provincial Corps Hospital, Chinese People's Armed Police Forces, Leshan, People's Republic of China
| | - Chunxia Luo
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
9
|
Terashita T, Kobayashi K, Nagano T, Kawa Y, Tamura D, Nakata K, Yamamoto M, Tachihara M, Kamiryo H, Nishimura Y. Administration of JTE013 abrogates experimental asthma by regulating proinflammatory cytokine production from bronchial epithelial cells. Respir Res 2016; 17:146. [PMID: 27829417 PMCID: PMC5103479 DOI: 10.1186/s12931-016-0465-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/01/2016] [Indexed: 12/18/2022] Open
Abstract
Background Sphingosine-1-phosphate (S1P) is a bioactive phospholipid that acts as a signal transducer by binding to S1P receptors (S1PR) 1 to 5. The S1P/S1PRs pathway has been associated with remodeling and allergic inflammation in asthma, but the expression pattern of S1PR and its effects on non-immune cells have not been completely clarified. The aim of this study was to examine the contribution of the signaling of S1P and S1PRs expressed in airway epithelial cells (ECs) to asthma responses in mice. Methods Bronchial asthma was experimentally induced in BALB/c mice by ovalbumin (OVA) sensitization followed by an OVA inhalation challenge. The effects of S1PR antagonists on the development of asthma were analyzed 24 h after the OVA challenge. Results Immunohistological analysis revealed S1PR1-3 expression on mouse airway ECs. Quantitative real-time polymerase chain reaction demonstrated that S1P greatly stimulated the induction of CCL3 and TIMP2 mRNA in human airway ECs, i.e., BEAS-2B cells, in a dose-dependent manner. Pretreatment with the S1PR2 antagonist JTE013 inhibited the CCL3 gene expression in BEAS-2B cells. Immunohistological analysis also showed that the expression level of CCL3 was attenuated by JTE013 in asthmatic mice. Furthermore, JTE013 as well as anti-CCL3 antibody attenuated allergic responses. Intratracheal administration of JTE013 also attenuated eosinophilic reactions in bronchoalveolar lavage fluids. S1P induced transcription factor NFκB activation, while JTE013 greatly reduced the NFκB activation. Conclusions JTE013 attenuated allergic airway reactions by regulating CCL3 production from bronchial ECs. The intratracheal administration of JTE013 may be a promising therapeutic strategy for bronchial asthma. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0465-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomomi Terashita
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshitaka Kawa
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Daisuke Tamura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kyosuke Nakata
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hiroshi Kamiryo
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
10
|
Elkhidir HS, Richards JB, Cromar KR, Bell CS, Price RE, Atkins CL, Spencer CY, Malik F, Alexander AL, Cockerill KJ, Haque IU, Johnston RA. Plasminogen activator inhibitor-1 does not contribute to the pulmonary pathology induced by acute exposure to ozone. Physiol Rep 2016; 4:4/18/e12983. [PMID: 27670409 PMCID: PMC5037925 DOI: 10.14814/phy2.12983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 11/24/2022] Open
Abstract
Expression of plasminogen activator inhibitor (PAI)-1, the major physiological inhibitor of fibrinolysis, is increased in the lung following inhalation of ozone (O3), a gaseous air pollutant. PAI-1 regulates expression of interleukin (IL)-6, keratinocyte chemoattractant (KC), and macrophage inflammatory protein (MIP)-2, which are cytokines that promote lung injury, pulmonary inflammation, and/or airway hyperresponsiveness following acute exposure to O3 Given these observations, we hypothesized that PAI-1 contributes to the severity of the aforementioned sequelae by regulating expression of IL-6, KC, and MIP-2 following acute exposure to O3 To test our hypothesis, wild-type mice and mice genetically deficient in PAI-1 (PAI-1-deficient mice) were acutely exposed to either filtered room air or O3 (2 ppm) for 3 h. Four and/or twenty-four hours following cessation of exposure, indices of lung injury [bronchoalveolar lavage fluid (BALF) protein and epithelial cells], pulmonary inflammation (BALF IL-6, KC, MIP-2, macrophages, and neutrophils), and airway responsiveness to aerosolized acetyl-β-methylcholine chloride (respiratory system resistance) were measured in wild-type and PAI-1-deficient mice. O3 significantly increased indices of lung injury, pulmonary inflammation, and airway responsiveness in wild-type and PAI-1-deficient mice. With the exception of MIP-2, which was significantly lower in PAI-1-deficient as compared to wild-type mice 24 h following cessation of exposure to O3, no other genotype-related differences occurred subsequent to O3 exposure. Thus, following acute exposure to O3, PAI-1 neither regulates pulmonary expression of IL-6 and KC nor functionally contributes to any of the pulmonary pathological sequelae that arise from the noxious effects of inhaled O3.
Collapse
Affiliation(s)
- Hamza S Elkhidir
- Division of Critical Care Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jeremy B Richards
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin R Cromar
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Cynthia S Bell
- Division of Nephrology, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Roger E Price
- Comparative Pathology Laboratory, Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas
| | - Constance L Atkins
- Division of Pulmonary Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Chantal Y Spencer
- Section of Pediatric Pulmonology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Farhan Malik
- Division of Critical Care Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Amy L Alexander
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Katherine J Cockerill
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Ikram U Haque
- Division of Critical Care Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Richard A Johnston
- Division of Critical Care Medicine, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas Pediatric Research Center, Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas Department of Integrative Biology and Pharmacology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
11
|
Matalon S, Bartoszewski R, Collawn JF. Role of epithelial sodium channels in the regulation of lung fluid homeostasis. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1229-38. [PMID: 26432872 DOI: 10.1152/ajplung.00319.2015] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/25/2015] [Indexed: 01/11/2023] Open
Abstract
In utero, fetal lung epithelial cells actively secrete Cl(-) ions into the lung air spaces while Na(+) ions follow passively to maintain electroneutrality. This process, driven by an electrochemical gradient generated by the Na(+)-K(+)-ATPase, is responsible for the secretion of fetal fluid that is essential for normal lung development. Shortly before birth, a significant upregulation of amiloride-sensitive epithelial channels (ENaCs) on the apical side of the lung epithelial cells results in upregulation of active Na(+) transport. This process is critical for the reabsorption of fetal lung fluid and the establishment of optimum gas exchange. In the adult lung, active Na(+) reabsorption across distal lung epithelial cells limits the degree of alveolar edema in patients with acute lung injury and cardiogenic edema. Cl(-) ions are transported either paracellularly or transcellularly to preserve electroneutrality. An increase in Cl(-) secretion across the distal lung epithelium has been reported following an acute increase in left atrial pressure and may result in pulmonary edema. In contrast, airway epithelial cells secrete Cl(-) through apical cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels and absorb Na(+). Thus the coordinated action of Cl(-) secretion and Na(+) absorption is essential for maintenance of the volume of epithelial lining fluid that, in turn, maximizes mucociliary clearance and facilitates clearance of bacteria and debris from the lungs. Any factor that interferes with Na(+) or Cl(-) transport or dramatically upregulates ENaC activity in airway epithelial cells has been associated with lung diseases such as cystic fibrosis or chronic obstructive lung disease. In this review we focus on the role of the ENaC, the mechanisms involved in ENaC regulation, and how ENaC dysregulation can lead to lung pathology.
Collapse
Affiliation(s)
- Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Pulmonary Injury and Repair Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; Gregory Fleming James Cystic Fibrosis Center, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
12
|
Wang Q, Wang Y, Zhang Y, Zhang Y, Xiao W. Involvement of urokinase in cigarette smoke extract-induced epithelial-mesenchymal transition in human small airway epithelial cells. J Transl Med 2015; 95:469-79. [PMID: 25706093 DOI: 10.1038/labinvest.2015.33] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/23/2014] [Accepted: 12/05/2014] [Indexed: 01/04/2023] Open
Abstract
Urokinase-type plasminogen activator (uPA) augments inflammation and tissue remodeling during lung injury and repair. The uPA expression in small airway epithelium of chronic obstructive pulmonary disease (COPD) increases. Epithelial-mesenchymal transition (EMT) is important in the small airway fibrosis of COPD. This study shows the uPA regulation in cigarette smoke extract (CSE)-induced EMT in human small airway epithelial cell lines (HSAEpiCs). uPA is overexpressed in the small airway epithelium of COPD patients and CSE-treated cell lines. Furthermore, uPA expression correlated with vimentin expression in the small airway epithelium of COPD patients. uPA inhibition blocks CSE-induced EMT by reversing E-cadherin and α-catenin expression and retarding the induction of N-cadherin and vimentin, resulting in reduction in migration. uPA overexpression in HSAEpiC cells also promotes EMT and migration. EMT is partly reversed in uPA-overexpressing HSAEpiC cells through the silencing expression of uPA receptor. In conclusion, this study provides new insights into the contribution of uPA upregulation to EMT associated with small airway remodeling in COPD.
Collapse
Affiliation(s)
- Qin Wang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Yunshan Wang
- 1] Department of Anatomy, Shandong University School of Medicine, Jinan, China [2] School of Ocean, Shandong University, Weihai, China
| | - Yi Zhang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Yuke Zhang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Wei Xiao
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
13
|
IMD-4690, a novel specific inhibitor for plasminogen activator inhibitor-1, reduces allergic airway remodeling in a mouse model of chronic asthma via regulating angiogenesis and remodeling-related mediators. PLoS One 2015; 10:e0121615. [PMID: 25785861 PMCID: PMC4364779 DOI: 10.1371/journal.pone.0121615] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/02/2015] [Indexed: 12/13/2022] Open
Abstract
Plasminogen activator inhibitor (PAI)-1 is the principal inhibitor of plasminogen activators, and is responsible for the degradation of fibrin and extracellular matrix. IMD-4690 is a newly synthesized inhibitor for PAI-1, whereas the effect on allergic airway inflammation and remodeling is still unclear. We examined the in vivo effects by using a chronic allergen exposure model of bronchial asthma in mice. The model was generated by an immune challenge for 8 weeks with house dust mite antigen, Dermatophagoides pteronyssinus (Dp). IMD-4690 was intraperitoneally administered during the challenge. Lung histopathology, hyperresponsiveness and the concentrations of mediators in lung homogenates were analyzed. The amount of active PAI-1 in the lungs was increased in mice treated with Dp. Administration with IMD-4690 reduced an active/total PAI-1 ratio. IMD-4690 also reduced the number of bronchial eosinophils in accordance with the decreased expressions of Th2 cytokines in the lung homogenates. Airway remodeling was inhibited by reducing subepithelial collagen deposition, smooth muscle hypertrophy, and angiogenesis. The effects of IMD-4690 were partly mediated by the regulation of TGF-β, HGF and matrix metalloproteinase. These results suggest that PAI-1 plays crucial roles in airway inflammation and remodeling, and IMD-4690, a specific PAI-1 inhibitor, may have therapeutic potential for patients with refractory asthma due to airway remodeling.
Collapse
|
14
|
Ji HL, Zhao R, Komissarov AA, Chang Y, Liu Y, Matthay MA. Proteolytic regulation of epithelial sodium channels by urokinase plasminogen activator: cutting edge and cleavage sites. J Biol Chem 2015; 290:5241-55. [PMID: 25555911 DOI: 10.1074/jbc.m114.623496] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Plasminogen activator inhibitor 1 (PAI-1) level is extremely elevated in the edematous fluid of acutely injured lungs and pleurae. Elevated PAI-1 specifically inactivates pulmonary urokinase-type (uPA) and tissue-type plasminogen activators (tPA). We hypothesized that plasminogen activation and fibrinolysis may alter epithelial sodium channel (ENaC) activity, a key player in clearing edematous fluid. Two-chain urokinase (tcuPA) has been found to strongly stimulate heterologous human αβγ ENaC activity in a dose- and time-dependent manner. This activity of tcuPA was completely ablated by PAI-1. Furthermore, a mutation (S195A) of the active site of the enzyme also prevented ENaC activation. By comparison, three truncation mutants of the amino-terminal fragment of tcuPA still activated ENaC. uPA enzymatic activity was positively correlated with ENaC current amplitude prior to reaching the maximal level. In sharp contrast to uPA, neither single-chain tPA nor derivatives, including two-chain tPA and tenecteplase, affected ENaC activity. Furthermore, γ but not α subunit of ENaC was proteolytically cleaved at ((177)GR↓KR(180)) by tcuPA. In summary, the underlying mechanisms of urokinase-mediated activation of ENaC include release of self-inhibition, proteolysis of γ ENaC, incremental increase in opening rate, and activation of closed (electrically "silent") channels. This study for the first time demonstrates multifaceted mechanisms for uPA-mediated up-regulation of ENaC, which form the cellular and molecular rationale for the beneficial effects of urokinase in mitigating mortal pulmonary edema and pleural effusions.
Collapse
Affiliation(s)
- Hong-Long Ji
- From the Department of Cellular and Molecular Biology and the Texas Lung Injury Institute, University of Texas Health Science Center, Tyler, Texas 75708,
| | - Runzhen Zhao
- From the Department of Cellular and Molecular Biology and
| | | | - Yongchang Chang
- the Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona 85013
| | - Yongfeng Liu
- the College of Public Health, Xinxiang Medical University, Xinxiang, Henan 453100, China, and
| | - Michael A Matthay
- the Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California 94143
| |
Collapse
|
15
|
Chen Z, Zhao R, Zhao M, Liang X, Bhattarai D, Dhiman R, Shetty S, Idell S, Ji HL. Regulation of epithelial sodium channels in urokinase plasminogen activator deficiency. Am J Physiol Lung Cell Mol Physiol 2014; 307:L609-17. [PMID: 25172911 DOI: 10.1152/ajplung.00126.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epithelial sodium channels (ENaC) govern transepithelial salt and fluid homeostasis. ENaC contributes to polarization, apoptosis, epithelial-mesenchymal transformation, etc. Fibrinolytic proteases play a crucial role in virtually all of these processes and are elaborated by the airway epithelium. We hypothesized that urokinase-like plasminogen activator (uPA) regulates ENaC function in airway epithelial cells and tested that possibility in primary murine tracheal epithelial cells (MTE). Both basal and cAMP-activated Na(+) flow through ENaC were significantly reduced in monolayers of uPA-deficient cells. The reduction in ENaC activity was further confirmed in basolateral membrane-permeabilized cells. A decrease in the Na(+)-K(+)-ATPase activity in the basolateral membrane could contribute to the attenuation of ENaC function in intact monolayer cells. Dysfunctional fluid resolution was seen in uPA-disrupted cells. Administration of uPA and plasmin partially restores ENaC activity and fluid reabsorption by MTEs. ERK1/2, but not Akt, phosphorylation was observed in the cells and lungs of uPA-deficient mice. On the other hand, cleavage of γ ENaC is significantly depressed in the lungs of uPA knockout mice vs. those of wild-type controls. Expression of caspase 8, however, did not differ between wild-type and uPA(-/-) mice. In addition, uPA deficiency did not alter transepithelial resistance. Taken together, the mechanisms for the regulation of ENaC by uPA in MTEs include augmentation of Na(+)-K(+)-ATPase, proteolysis, and restriction of ERK1/2 phosphorylation. We demonstrate for the first time that ENaC may serve as a downstream signaling target by which uPA controls the biophysical profiles of airway fluid and epithelial function.
Collapse
Affiliation(s)
- Zaixing Chen
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; School of Pharmacy, China Medical University, Liaoning Shenyang, China
| | - Runzhen Zhao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Meimi Zhao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; School of Pharmacy, China Medical University, Liaoning Shenyang, China
| | - Xinrong Liang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Deepa Bhattarai
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Rohan Dhiman
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Sreerama Shetty
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas; Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, Texas; and
| |
Collapse
|
16
|
Stewart AG, Xia YC, Harris T, Royce S, Hamilton JA, Schuliga M. Plasminogen-stimulated airway smooth muscle cell proliferation is mediated by urokinase and annexin A2, involving plasmin-activated cell signalling. Br J Pharmacol 2014; 170:1421-35. [PMID: 24111848 DOI: 10.1111/bph.12422] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/04/2013] [Accepted: 08/27/2013] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND AND PURPOSE The conversion of plasminogen into plasmin by interstitial urokinase plasminogen activator (uPA) is potentially important in asthma pathophysiology. In this study, the effect of uPA-mediated plasminogen activation on airway smooth muscle (ASM) cell proliferation was investigated. EXPERIMENTAL APPROACH Human ASM cells were incubated with plasminogen (0.5-50 μg·mL(-1) ) or plasmin (0.5-50 mU·mL(-1) ) in the presence of pharmacological inhibitors, including UK122, an inhibitor of uPA. Proliferation was assessed by increases in cell number or MTT reduction after 48 h incubation with plasmin(ogen), and by earlier increases in [(3) H]-thymidine incorporation and cyclin D1 expression. KEY RESULTS Plasminogen (5 μg·mL(-1) )-stimulated increases in cell proliferation were attenuated by UK122 (10 μM) or by transfection with uPA gene-specific siRNA. Exogenous plasmin (5 mU·mL(-1) ) also stimulated increases in cell proliferation. Inhibition of plasmin-stimulated ERK1/2 or PI3K/Akt signalling attenuated plasmin-stimulated increases in ASM proliferation. Furthermore, pharmacological inhibition of cell signalling mediated by the EGF receptor, a receptor trans-activated by plasmin, also reduced plasmin(ogen)-stimulated cell proliferation. Knock down of annexin A2, which has dual roles in both plasminogen activation and plasmin-signal transduction, also attenuated ASM cell proliferation following incubation with either plasminogen or plasmin. CONCLUSIONS AND IMPLICATIONS Plasminogen stimulates ASM cell proliferation in a manner mediated by uPA and involving multiple signalling pathways downstream of plasmin. Targeting mediators of plasminogen-evoked ASM responses, such as uPA or annexin A2, may be useful in the treatment of asthma.
Collapse
Affiliation(s)
- A G Stewart
- Department of Pharmacology, University of Melbourne, Parkville, VIC, Australia; Lung Health Research Centre, University of Melbourne, Parkville, VIC, Australia
| | | | | | | | | | | |
Collapse
|
17
|
Schuliga M, Westall G, Xia Y, Stewart AG. The plasminogen activation system: new targets in lung inflammation and remodeling. Curr Opin Pharmacol 2013; 13:386-93. [PMID: 23735578 DOI: 10.1016/j.coph.2013.05.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 11/26/2022]
Abstract
The plasminogen activation system (PAS) and the plasmin it forms have dual roles in chronic respiratory diseases including asthma, chronic obstructive pulmonary disease and interstitial lung disease. Whilst plasmin-mediated airspace fibrinolysis is beneficial, interstitial plasmin contributes to lung dysfunction because of its pro-inflammatory and tissue remodeling activities. Recent studies highlight the potential of fibrinolytic agents, including small molecule inhibitors of plasminogen activator inhibitor-1 (PAI-1), as treatments for chronic respiratory disease. Current data also suggest that interstitial urokinase plasminogen activator is an important mediator of lung inflammation and remodeling. However, further preclinical characterization of uPA as a drug target for lung disease is required. Here we review the concept of selectively targeting the contributions of PAS to treat chronic respiratory disease.
Collapse
Affiliation(s)
- Michael Schuliga
- Department of Pharmacol, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
18
|
Narumoto O, Matsuo Y, Sakaguchi M, Shoji S, Yamashita N, Schubert D, Abe K, Horiguchi K, Nagase T, Yamashita N. Suppressive effects of a pyrazole derivative of curcumin on airway inflammation and remodeling. Exp Mol Pathol 2012; 93:18-25. [PMID: 22542791 DOI: 10.1016/j.yexmp.2012.04.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 03/22/2012] [Accepted: 04/10/2012] [Indexed: 10/28/2022]
Abstract
To advance the control of airway epithelial cell function and asthma, we investigated the effects of a new curcumin derivative, CNB001, which possesses improved pharmacological properties. Normal human bronchial epithelial (NHBE) cells were stimulated with synthetic double-stranded RNA, Poly(I:C). CNB001 significantly suppressed IL-6, TNF-α, and GM-CSF production by NHBE cells, and did so more effectively than did curcumin or dexamethasone (DEX). CNB001 significantly inhibited the decrease of E-cadherin mRNA expression and increase of vimentin mRNA expression observed in NHBE cells induced by a combination of TGF-β1 and TNF-α, which are markers of airway remodeling. In NHBE cells stimulated by TGF-β1, CNB001 significantly downregulated the level of active serine peptidase inhibitor clade E member (SERPINE) 1, which is also reported to be related to airway remodeling. Whereas DEX alone significantly increased the active SERPINE1 level, the combination of DEX and CNB001 significantly suppressed active SERPINE1. In addition, CNB001 significantly suppressed neutrophil infiltration, IL-6, TNF-α, IL-13 and active SERPINE1 production in bronchoalveolar lavage fluid of the murine asthma model, which was not observed in the case of DEX. In conclusion, the curcumin derivative, CNB001, is a promising candidate to treat asthma associated with neutrophilic airway inflammation and remodeling.
Collapse
Affiliation(s)
- Osamu Narumoto
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences Musashino University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Asthma is a chronic airway disease characterized by paroxysmal airflow obstruction evoked by irritative stimuli on a background of allergic lung inflammation. Currently, there is no cure for asthma, only symptomatic treatment. In recent years, our understanding of the involvement of coagulation and anticoagulant pathways, the fibrinolytic system, and platelets in the pathophysiology of asthma has increased considerably. Asthma is associated with a procoagulant state in the bronchoalveolar space, further aggravated by impaired local activities of the anticoagulant protein C system and fibrinolysis. Protease-activated receptors have been implicated as the molecular link between coagulation and allergic inflammation in asthma. This review summarizes current knowledge of the impact of the disturbed hemostatic balance in the lungs on asthma severity and manifestations and identifies new possible targets for asthma treatment.
Collapse
|
20
|
Stewart CE, Nijmeh HS, Brightling CE, Sayers I. uPAR regulates bronchial epithelial repair in vitro and is elevated in asthmatic epithelium. Thorax 2011; 67:477-87. [PMID: 22139533 PMCID: PMC3358731 DOI: 10.1136/thoraxjnl-2011-200508] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background The asthma-associated gene urokinase plasminogen activator receptor (uPAR) may be involved in epithelial repair and airway remodelling. These processes are not adequately targeted by existing asthma therapies. A fuller understanding of the pathways involved in remodelling may lead to development of new therapeutic opportunities. uPAR expression in the lung epithelium of normal subjects and patients with asthma was investigated and the contribution of uPAR to epithelial wound repair in vitro was studied using primary bronchial epithelial cells (NHBECs). Methods Bronchial biopsy sections from normal subjects and patients with asthma were immunostained for uPAR. NHBECs were used in a scratch wound model to investigate the contribution of the plasminogen pathway to repair. The pathway was targeted via blocking of the interaction between urokinase plasminogen activator (uPA) and uPAR and overexpression of uPAR. The rate of wound closure and activation of intracellular signalling pathways and matrix metalloproteinases (MMPs) were measured. Results uPAR expression was significantly increased in the bronchial epithelium of patients with asthma compared with controls. uPAR expression was increased during wound repair in monolayer and air-liquid interface-differentiated NHBEC models. Blocking the uPA–uPAR interaction led to attenuated wound repair via changes in Erk1/2, Akt and p38MAPK signalling. Cells engineered to have raised levels of uPAR showed attenuated repair via sequestration of uPA by soluble uPAR. Conclusions The uPAR pathway is required for efficient epithelial wound repair. Increased uPAR expression, as seen in the bronchial epithelium of patients with asthma, leads to attenuated wound repair which may contribute to the development and progression of airway remodelling in asthma. This pathway may therefore represent a potential novel therapeutic target for the treatment of asthma.
Collapse
Affiliation(s)
- Ceri E Stewart
- Division of Therapeutics and Molecular Medicine, Nottingham Respiratory Biomedical Research Unit, University Hospital of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | |
Collapse
|
21
|
Miyamoto S, Hattori N, Senoo T, Onari Y, Iwamoto H, Kanehara M, Ishikawa N, Fujitaka K, Haruta Y, Murai H, Yokoyama A, Kohno N. Intra-airway administration of small interfering RNA targeting plasminogen activator inhibitor-1 attenuates allergic asthma in mice. Am J Physiol Lung Cell Mol Physiol 2011; 301:L908-16. [PMID: 21926267 DOI: 10.1152/ajplung.00115.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recent studies suggest that plasminogen activator inhibitor-1 (PAI-1), a major inhibitor of the fibrinolytic system, may promote the development of asthma. To further investigate the significance of PAI-1 in the pathogenesis of asthma and determine the possibility that PAI-1 could be a therapeutic target for asthma, this study was conducted. First, PAI-1 levels in induced sputum (IS) from asthmatic subjects and healthy controls were measured. In asthmatic subjects, IS PAI-1 levels were elevated, compared with that of healthy controls, and were significantly higher in patients with long-duration asthma compared with short-duration asthma. PAI-1 levels were also found to correlate with IS transforming growth factor-β levels. Then, acute and chronic asthma models induced by ovalbumin were established in PAI-1-deficient mice and wild-type mice that received intra-airway administrations of small interfering RNA against PAI-1 (PAI-1-siRNA). We could demonstrate that eosinophilic airway inflammation and airway hyperresponsiveness were reduced in an acute asthma model, and airway remodeling was suppressed in a chronic asthma model in both PAI-1-deficient mice and wild-type mice that received intra-airway administration of PAI-1-siRNA. These results indicate that PAI-1 is strongly involved in the pathogenesis of asthma, and intra-airway administration of PAI-1-siRNA may be able to become a new therapeutic approach for asthma.
Collapse
Affiliation(s)
- Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Concentrations of plasminogen activator inhibitor-1 (PAI-1) and urokinase plasminogen activator (uPA) in induced sputum of asthma patients after allergen challenge. Folia Histochem Cytobiol 2011; 48:518-23. [PMID: 21478092 DOI: 10.2478/v10042-010-0075-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Urokinase plasminogen activator (uPA) and its inhibitor (PAI-1) are involved in tiisue remodeling and repair processes associated with acute and chronic inflammation. The aim of the study was to evaluate the effect of allergen challenge on concentration of uPA and PAI-1 in induced sputum of house dust mite allergic asthmatics (HDM-AAs). Thirty HDM-AAs and ten healthy persons (HCs)were recruited for the study. In 24 HDM-AAs bronchial challenge with Dermatophagoides pteronyssinus (Dp) and in 6 HDM-AAs sham challenege with saline were performed. In HDM-AAs sputum was induced 24 hours before (T0) and 24 hours (T24) after the challenge. Concentration of uPA and PAI-1 in induced sputum were determined using immunoenzymatic assays. At T0 in HDM-AAs mean sputum uPA (151 ± 96 pg/ml) and PAI-1 (4341 ± 1262 pg/ml) concentrations were higher than in HC (18.8 ± 6.7 pg/ml; p=0.0002 and 596 ± 180 pg/ml; p<0.0001; for uPA and PAI-1 respectively). After allergen challenge further increase in sputum uPA (187 ± 144 pg/ml; p=0.03) and PAI-1 (6252 ± 2323 pg/ml; p<0.0001) concentrations were observed. Moreover, in Dp challenged, but not in saline challenged HDM-AAs the mean uPA/PAI-1 ratio decreased significantly at T24. No significant increase in the studied parameters were found in sham challenged patients. In HDM-AAs allergen exposure leads to activation of the plasmin system in the airways. Greater increase of the PAI-1 concentration than uPA concentration after allergen challenge may promote airway remodeling and play an important role in the development of bronchial hyperreactivity.
Collapse
|
23
|
Evidence of a genetic contribution to lung function decline in asthma. J Allergy Clin Immunol 2011; 128:479-84. [PMID: 21752436 DOI: 10.1016/j.jaci.2011.05.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/25/2011] [Accepted: 05/25/2011] [Indexed: 11/21/2022]
Abstract
There has been great progress in identifying new asthma susceptibility genes. In asthmatic subjects there is variable airway remodeling that includes features such as smooth muscle hypertrophy/hyperplasia, basement membrane thickening, and increased extracellular matrix deposition. Does airway remodeling have a genetic contribution in asthma? Data from different murine strains suggest there is a genetic contribution to the development and progression of airway remodeling. In human subjects it is important to consider what surrogate markers of remodeling have been used in genetic studies. Baseline FEV(1) and airway hyperresponsiveness are determined by a complex interplay of factors, including nonremodeling mechanisms; however, we consider a decline in FEV(1) as a robust marker of remodeling. To date, single nucleotide polymorphisms spanning ADAM33, ESR1, PLAUR, and VEGF have been associated with an excess decline in lung function in asthmatic subjects carrying the rare alleles (FEV(1), -13.0 to 55.2 mL/y excess). Interestingly these genes have overlapping functions in proteolytic pathways in the airways. There is accumulating evidence that genetic factors are important in the development of airway remodeling in asthmatic subjects, and further longitudinal studies with additional remodeling phenotypes and genome-wide association studies will identify novel susceptibility genes, leading to new approaches to target remodeling in asthmatic subjects.
Collapse
|
24
|
Chen X, Song CH, Feng BS, Li TL, Li P, Zheng PY, Chen XM, Xing Z, Yang PC. Intestinal epithelial cell-derived integrin αβ6 plays an important role in the induction of regulatory T cells and inhibits an antigen-specific Th2 response. J Leukoc Biol 2011; 90:751-9. [PMID: 21724807 DOI: 10.1189/jlb.1210696] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Toleroge nic DCs and Tregs are believed to play a critical role in oral tolerance. However, the mechanisms of the generation of tolerogenic DCs and activation of Tregs in the gut remain poorly understood. This study aims to dissect the molecular mechanisms by which IECs and protein antigen induce functional tolerogenic DCs and Tregs. Expression of αvβ6 by gut epithelial cell-derived exosomes, its coupling with food antigen, and their relationship with the development of functional tolerogenic DCs and Tregs were examined by using in vitro and in vivo approaches. The results show that IECs up-regulated the integrin αvβ6 upon uptake of antigens. The epithelial cell-derived exosomes entrapped and transported αvβ6 and antigens to the extracellular environment. The uptake of antigens alone induced DCs to produce LTGFβ, whereas exosomes carrying αvβ6/antigen resulted in the production of abundant, active TGF-β in DCs that conferred to DCs the tolerogenic properties. Furthermore, αvβ6/OVA-carrying, exosome-primed DCs were found to promote the production of active TGF-β in Tregs. Thus, in vivo administration of αvβ6/OVA-laden exosomes induced the generation of Tregs and suppressed skewed Th2 responses toward food antigen in the intestine. Our study provides important molecular insights into the molecular mechanisms of Treg development by demonstrating an important role of IEC-derived exosomes carrying αvβ6 and food antigen in the induction of tolerogenic DCs and antigen-specific Tregs.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Makarova AM, Lebedeva TV, Nassar T, Higazi AAR, Xue J, Carinato ME, Bdeir K, Cines DB, Stepanova V. Urokinase-type plasminogen activator (uPA) induces pulmonary microvascular endothelial permeability through low density lipoprotein receptor-related protein (LRP)-dependent activation of endothelial nitric-oxide synthase. J Biol Chem 2011; 286:23044-53. [PMID: 21540184 PMCID: PMC3123072 DOI: 10.1074/jbc.m110.210195] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2010] [Revised: 04/19/2011] [Indexed: 01/11/2023] Open
Abstract
Urokinase plasminogen activator (uPA) and PA inhibitor type 1 (PAI-1) are elevated in acute lung injury, which is characterized by a loss of endothelial barrier function and the development of pulmonary edema. Two-chain uPA and uPA-PAI-1 complexes (1-20 nM) increased the permeability of monolayers of human pulmonary microvascular endothelial cells (PMVECs) in vitro and lung permeability in vivo. The effects of uPA-PAI-1 were abrogated by the nitric-oxide synthase (NOS) inhibitor L-NAME (N(D)-nitro-L-arginine methyl ester). Two-chain uPA (1-20 nM) and uPA-PAI-1 induced phosphorylation of endothelial NOS-Ser(1177) in PMVECs, which was followed by generation of NO and the nitrosylation and dissociation of β-catenin from VE-cadherin. uPA-induced phosphorylation of eNOS was decreased by anti-low density lipoprotein receptor-related protein-1 (LRP) antibody and an LRP antagonist, receptor-associated protein (RAP), and when binding to the uPA receptor was blocked by the isolated growth factor-like domain of uPA. uPA-induced phosphorylation of eNOS was also inhibited by the protein kinase A (PKA) inhibitor, myristoylated PKI, but was not dependent on PI3K-Akt signaling. LRP blockade and inhibition of PKA prevented uPA- and uPA-PAI-1-induced permeability of PMVEC monolayers in vitro and uPA-induced lung permeability in vivo. These studies identify a novel pathway involved in regulating PMVEC permeability and suggest the utility of uPA-based approaches that attenuate untoward permeability following acute lung injury while preserving its salutary effects on fibrinolysis and airway remodeling.
Collapse
Affiliation(s)
- Anastasia M. Makarova
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tatiana V. Lebedeva
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Taher Nassar
- the Department of Clinical Biochemistry, Hebrew University-Hadassah Medical Center, Jerusalem 91120, Israel, and
| | - Abd Al-Roof Higazi
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- the Department of Clinical Biochemistry, Hebrew University-Hadassah Medical Center, Jerusalem 91120, Israel, and
| | - Jing Xue
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- the Department of Laboratory Medicine, Tianjin Huanhu Hospital, Tianjin 300060, China
| | - Maria E. Carinato
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Khalil Bdeir
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Douglas B. Cines
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Victoria Stepanova
- From the Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
26
|
Novak ML, Bryer SC, Cheng M, Nguyen MH, Conley KL, Cunningham AK, Xue B, Sisson TH, You JS, Hornberger TA, Koh TJ. Macrophage-specific expression of urokinase-type plasminogen activator promotes skeletal muscle regeneration. THE JOURNAL OF IMMUNOLOGY 2011; 187:1448-57. [PMID: 21709151 DOI: 10.4049/jimmunol.1004091] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Macrophages (Mp) and the plasminogen system play important roles in tissue repair following injury. We hypothesized that Mp-specific expression of urokinase-type plasminogen activator (uPA) is sufficient for Mp to migrate into damaged muscle and for efficient muscle regeneration. We generated transgenic mice expressing uPA only in Mp, and we assessed the ability of these mice to repair muscle injury. Mp-only uPA expression was sufficient to induce wild-type levels of Mp accumulation, angiogenesis, and new muscle fiber formation. In mice with wild-type uPA expression, Mp-specific overexpression further increased Mp accumulation and enhanced muscle fiber regeneration. Furthermore, Mp expression of uPA regulated the level of active hepatocyte growth factor, which is required for muscle fiber regeneration, in damaged muscle. In vitro studies demonstrated that uPA promotes Mp migration through proteolytic and nonproteolytic mechanisms, including proteolytic activation of hepatocyte growth factor. In summary, Mp-derived uPA promotes muscle regeneration by inducing Mp migration, angiogenesis, and myogenesis.
Collapse
Affiliation(s)
- Margaret L Novak
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Nassar T, Yarovoi S, Fanne RA, Waked O, Allen TC, Idell S, Cines DB, Higazi AAR. Urokinase plasminogen activator regulates pulmonary arterial contractility and vascular permeability in mice. Am J Respir Cell Mol Biol 2011; 45:1015-21. [PMID: 21617202 DOI: 10.1165/rcmb.2010-0302oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The concentration of urokinase plasminogen activator (uPA) is elevated in pathological settings such as acute lung injury, where pulmonary arterial contractility and permeability are disrupted. uPA limits the accretion of fibrin after injury. Here we investigated whether uPA also regulates pulmonary arterial contractility and permeability. Contractility was measured using isolated pulmonary arterial rings. Pulmonary blood flow was measured in vivo by Doppler and pulmonary vascular permeability, according to the extravasation of Evans blue. Our data show that uPA regulates the in vitro pulmonary arterial contractility induced by phenylephrine in a dose-dependent manner through two receptor-dependent pathways, and regulates vascular contractility and permeability in vivo. Physiological concentrations of uPA (≤1 nM) stimulate the contractility of pulmonary arterial rings induced by phenylephrine through the low-density lipoprotein receptor-related protein receptor. The procontractile effect of uPA is independent of its catalytic activity. At pathophysiological concentrations, uPA (20 nM) inhibits contractility and increases vascular permeability. The inhibition of vascular contractility and increase of vascular permeability is mediated through a two-step process that involves docking to N-methyl-d-aspartate receptor-1 (NMDA-R1) on pulmonary vascular smooth muscle cells, and requires catalytic activity. Peptides that specifically inhibit the docking of uPA to NMDA-R, or the uPA variant with a mutated receptor docking site, abolished both the effects of uPA on vascular contractility and permeability, without affecting its catalytic activity. These data show that uPA, at concentrations found under pathological conditions, reduces pulmonary arterial contractility and increases permeability though the activation of NMDA-R1. The selective inhibition of NMDAR-1 activation by uPA can be accomplished without a loss of fibrinolytic activity.
Collapse
Affiliation(s)
- Taher Nassar
- Department of Pathology, Laboratory Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kim DY, Kwon EY, Hong GU, Lee YS, Lee SH, Ro JY. Cigarette smoke exacerbates mouse allergic asthma through Smad proteins expressed in mast cells. Respir Res 2011; 12:49. [PMID: 21496353 PMCID: PMC3098800 DOI: 10.1186/1465-9921-12-49] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 04/18/2011] [Indexed: 11/10/2022] Open
Abstract
Background Many studies have found that smoking reduces lung function, but the relationship between cigarette smoke and allergic asthma has not been clearly elucidated, particularly the role of mast cells. This study aimed to investigate the effects of smoke exposure on allergic asthma and its association with mast cells. Methods BALB/c mice were sensitized and challenged by OVA to induce asthma, and bone marrow-derived mast cells (BMMCs) were stimulated with antigen/antibody reaction. Mice or BMMCs were exposed to cigarette smoke or CSE solution for 1 mo or 6 h, respectively. The recruitment of inflammatory cells into BAL fluid or lung tissues was determined by Diff-Quik or H&E staining, collagen deposition by Sircol assay, penh values by a whole-body plethysmography, co-localization of tryptase and Smad3 by immunohistochemistry, IgE and TGF-β level by ELISA, expressions of Smads proteins, activities of signaling molecules, or TGF-β mRNA by immunoblotting and RT-PCR. Results Cigarette smoke enhanced OVA-specific IgE levels, penh values, recruitment of inflammatory cells including mast cells, expressions of smad family, TGF-β mRNA and proteins, and cytokines, phosphorylations of Smad2 and 3, and MAP kinases, co-localization of tryptase and Smad3, and collagen deposition more than those of BAL cells and lung tissues of OVA-induced allergic mice. CSE solution pretreatment enhanced expressions of TGF-β, Smad3, activities of MAP kinases, NF-κB/AP-1 or PAI-1 more than those of activated-BMMCs. Conclusions The data suggest that smoke exposure enhances antigen-induced mast cell activation via TGF-β/Smad signaling pathways in mouse allergic asthma, and that it exacerbates airway inflammation and remodeling.
Collapse
Affiliation(s)
- Dae Yong Kim
- Department of Pharmacology and Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-726, Korea
| | | | | | | | | | | |
Collapse
|
29
|
Himes BE, Klanderman B, Ziniti J, Senter-Sylvia J, Soto-Quiros ME, Avila L, Celedón JC, Lange C, Mariani TJ, Lasky-Su J, Hersh CP, Raby BA, Silverman EK, Weiss ST, DeMeo DL. Association of SERPINE2 with asthma. Chest 2011; 140:667-674. [PMID: 21436250 DOI: 10.1378/chest.10-2973] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The "Dutch hypothesis" suggests that asthma and COPD have common genetic determinants. The serpin peptidase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 2 (SERPINE2) gene previously has been associated with COPD. We sought to determine whether SERPINE2 is associated with asthma and asthma-related phenotypes. METHODS We measured the association of 39 SERPINE2 single-nucleotide polymorphisms (SNPs) with asthma-related phenotypes in 655 parent-child trios from the Childhood Asthma Management Program (CAMP), and we measured the association of 19 SERPINE2 SNPs with asthma in a case-control design of 359 CAMP probands and 846 population control subjects. We attempted to replicate primary asthma-related phenotype findings in one independent population and primary asthma affection status findings in two independent populations. We compared association results with CAMP proband expression quantitative trait loci. RESULTS Nine of 39 SNPs had P < .05 for at least one phenotype in CAMP, and two of these replicated in an independent population of 426 people with childhood asthma. Six of 19 SNPs had P < .05 for association with asthma in CAMP/Illumina. None of these replicated in two independent populations. The expression quantitative trait loci revealed that five SNPs associated with asthma in CAMP/Illumina and one SNP associated with FEV(1) in CAMP are strongly correlated with SERPINE2 expression levels. Comparison of results to previous COPD studies identified five SNPs associated with both asthma- and COPD-related phenotypes. CONCLUSIONS Our results weakly support SERPINE2 as a Dutch hypothesis candidate gene through nominally significant associations with asthma and related traits. Further study of SERPINE2 is necessary to verify its involvement in asthma and COPD.
Collapse
Affiliation(s)
- Blanca E Himes
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA; Children's Hospital Informatics Program, Boston, MA.
| | - Barbara Klanderman
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - John Ziniti
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Jody Senter-Sylvia
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Manuel E Soto-Quiros
- Division of Pediatric Pulmonology, Hospital Nacional de Niños, San José, Costa Rica
| | - Lydiana Avila
- Division of Pediatric Pulmonology, Hospital Nacional de Niños, San José, Costa Rica
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, Allergy and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Christoph Lange
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Thomas J Mariani
- Division of Neonatology and Center for Pediatric Biomedical Research, University of Rochester, Rochester, NY
| | - Jessica Lasky-Su
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Craig P Hersh
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Benjamin A Raby
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Edwin K Silverman
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Scott T Weiss
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Dawn L DeMeo
- Channing Laboratory, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
30
|
Ma Z, Kwong KYC, Tovar JP, Paek D. Cyclic adenosine monophosphate induces plasminogen activator inhibitor-1 expression in human mast cells. Biochem Biophys Res Commun 2010; 400:569-74. [PMID: 20816667 DOI: 10.1016/j.bbrc.2010.08.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 08/25/2010] [Indexed: 02/02/2023]
Abstract
Plaminogen activator inhibitor-1 (PAI-1), the key physiological inhibitor of the plasmin fibrinolytic system, plays important roles in the pathogenesis of asthma. Mast cells (MCs) are crucial effector cells and a major source of PAI-1 for asthma. Cyclic adenosine monophosphate (cAMP) is the important regulator of MCs; however, its effects on PAI-1 expression in MCs remain unknown. We reported cAMP/protein kinase A pathway positively regulates PAI-1 expression through cAMP-response element binding protein binding to hypoxia response element-1 at -158 to -153bp of human PAI-1 promoter in human MCs. Moreover, cAMP synergistically augments PAI-1 expression with ionomycin- or IgE receptor cross-linking-mediated stimulation.
Collapse
Affiliation(s)
- Zhongcai Ma
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Torrance, CA 90502, USA.
| | | | | | | |
Collapse
|
31
|
Schuliga M, Harris T, Stewart AG. Plasminogen activation by airway smooth muscle is regulated by type I collagen. Am J Respir Cell Mol Biol 2010; 44:831-9. [PMID: 20693403 DOI: 10.1165/rcmb.2009-0469oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Plasmin, the activated protease product of plasminogen, is involved in collagen remodeling, and is strongly implicated in asthma pathophysiology by recent genome-wide association studies. This study examines plasminogen "activation" by airway smooth muscle cells, and its regulation in a fibrotic environment created by culture on type I collagen and incubation with transforming growth factor (TGF)-β. Urokinase plasminogen activator (uPA) activity was detected in the supernatants of human airway smooth muscle cell cultures maintained in serum-free conditions. Incubation with plasminogen (1.5-50.0 μg/ml, 24 h) increased plasmin activity in a concentration-dependent manner (P < 0.001). uPA activity was higher in cultures maintained on fibrillar type I collagen substrata than in those on plastic, as was plasmin activity after incubation with plasminogen (20 μg/ml). Pretreatment with TGF-β (100 pM) for 18 hours inhibited plasminogen activation by airway smooth muscle cells maintained on plastic, but not on collagen. TGF-β stimulated an increase in the level of uPA mRNA in airway smooth muscle cells grown on collagen, but not on plastic. Reducing the levels of β1-integrin collagen receptor, using interference RNA, attenuated plasmin formation by airway smooth muscle cells grown on collagen, and restored the inhibitory effect of TGF-β. This study shows that airway smooth muscle activation of plasminogen by uPA is accelerated in a collagen-rich environment in which the inhibitory effect of TGF-β is attenuated in association with greater uPA expression induced via β1-integrin signaling. These findings suggest that the plasminogen-activation system involving uPA has the potential to contribute to airway wall remodeling in asthma.
Collapse
Affiliation(s)
- Michael Schuliga
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia.
| | | | | |
Collapse
|
32
|
Nassar T, Yarovoi S, Fanne RA, Akkawi S, Jammal M, Allen TC, Idell S, Cines DB, Higazi AAR. Regulation of airway contractility by plasminogen activators through N-methyl-D-aspartate receptor-1. Am J Respir Cell Mol Biol 2010; 43:703-11. [PMID: 20097831 DOI: 10.1165/rcmb.2009-0257oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Reactive airway disease is mediated by smooth muscle contraction initiated through several agonist-dependent pathways. Activation of type 1 N-methyl-D-aspartate receptors (NMDA-R1s) by plasminogen activators (PAs) has been linked to control of vascular tone, but their effect on airway smooth muscle contractility has not previously been studied to our knowledge. We observed that NMDA-R1s are expressed by human airway smooth muscle cells and constitutively inhibit the contraction of isolated rat tracheal rings in response to acetylcholine (Ach). Both tissue-type PA (tPA) and urokinase-type PA (uPA) bind to NMDA-R1 and reverse this effect, thereby enhancing Ach-induced tracheal contractility. Tracheal contractility initiated by Ach is reduced in rings isolated from tPA(-/-) and uPA(-/-) mice compared with their wild-type counterparts. The procontractile effect of uPA or tPA was mimicked and augmented by the nitric oxide synthase inhibitor, l-NAME. uPA and tPA further enhanced the contractility of rings denuded of epithelium, an effect that was inhibited by the NMDA-R antagonist, MK-801. Binding of PAs to NMDA-R1 and the subsequent activation of the receptor were inhibited by PA inhibitor type 1, by a PA inhibitor type 1-derived hexapeptide that recognizes the tPA and uPA docking domains, as well as by specific mutations within the docking site of tPA. These studies identify involvement of PAs and NMDA-R1 in airway contractility, and define new loci that could lead to the development of novel interventions for reactive airway disease.
Collapse
Affiliation(s)
- Taher Nassar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 513A Stellar-Chance, 422 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|