51
|
Murtha LA, Schuliga MJ, Mabotuwana NS, Hardy SA, Waters DW, Burgess JK, Knight DA, Boyle AJ. The Processes and Mechanisms of Cardiac and Pulmonary Fibrosis. Front Physiol 2017; 8:777. [PMID: 29075197 PMCID: PMC5643461 DOI: 10.3389/fphys.2017.00777] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is the formation of fibrous connective tissue in response to injury. It is characterized by the accumulation of extracellular matrix components, particularly collagen, at the site of injury. Fibrosis is an adaptive response that is a vital component of wound healing and tissue repair. However, its continued activation is highly detrimental and a common final pathway of numerous disease states including cardiovascular and respiratory disease. Worldwide, fibrotic diseases cause over 800,000 deaths per year, accounting for ~45% of total deaths. With an aging population, the incidence of fibrotic disease and subsequently the number of fibrosis-related deaths will rise further. Although, fibrosis is a well-recognized cause of morbidity and mortality in a range of disease states, there are currently no viable therapies to reverse the effects of chronic fibrosis. Numerous predisposing factors contribute to the development of fibrosis. Biological aging in particular, interferes with repair of damaged tissue, accelerating the transition to pathological remodeling, rather than a process of resolution and regeneration. When fibrosis progresses in an uncontrolled manner, it results in the irreversible stiffening of the affected tissue, which can lead to organ malfunction and death. Further investigation into the mechanisms of fibrosis is necessary to elucidate novel, much needed, therapeutic targets. Fibrosis of the heart and lung make up a significant proportion of fibrosis-related deaths. It has long been established that the heart and lung are functionally and geographically linked when it comes to health and disease, and thus exploring the processes and mechanisms that contribute to fibrosis of each organ, the focus of this review, may help to highlight potential avenues of therapeutic investigation.
Collapse
Affiliation(s)
- Lucy A Murtha
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Michael J Schuliga
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Nishani S Mabotuwana
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sean A Hardy
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - David W Waters
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Janette K Burgess
- Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, W. J. Kolff Research Institute, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, Glebe, NSW, Australia.,Discipline of Pharmacology, University of Sydney, Sydney, NSW, Australia
| | - Darryl A Knight
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BS, Canada.,Department of Medicine, University of Western Australia, Perth, WA, Australia.,Research and Innovation Conjoint, Hunter New England Health, Newcastle, NSW, Australia
| | - Andrew J Boyle
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
52
|
Birring SS, Wijsenbeek MS, Agrawal S, van den Berg JWK, Stone H, Maher TM, Tutuncu A, Morice AH. A novel formulation of inhaled sodium cromoglicate (PA101) in idiopathic pulmonary fibrosis and chronic cough: a randomised, double-blind, proof-of-concept, phase 2 trial. THE LANCET RESPIRATORY MEDICINE 2017; 5:806-815. [PMID: 28923239 DOI: 10.1016/s2213-2600(17)30310-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/31/2017] [Accepted: 08/01/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cough can be a debilitating symptom of idiopathic pulmonary fibrosis (IPF) and is difficult to treat. PA101 is a novel formulation of sodium cromoglicate delivered via a high-efficiency eFlow nebuliser that achieves significantly higher drug deposition in the lung compared with the existing formulations. We aimed to test the efficacy and safety of inhaled PA101 in patients with IPF and chronic cough and, to explore the antitussive mechanism of PA101, patients with chronic idiopathic cough (CIC) were also studied. METHODS This pilot, proof-of-concept study consisted of a randomised, double-blind, placebo-controlled trial in patients with IPF and chronic cough and a parallel study of similar design in patients with CIC. Participants with IPF and chronic cough recruited from seven centres in the UK and the Netherlands were randomly assigned (1:1, using a computer-generated randomisation schedule) by site staff to receive PA101 (40 mg) or matching placebo three times a day via oral inhalation for 2 weeks, followed by a 2 week washout, and then crossed over to the other arm. Study participants, investigators, study staff, and the sponsor were masked to group assignment until all participants had completed the study. The primary efficacy endpoint was change from baseline in objective daytime cough frequency (from 24 h acoustic recording, Leicester Cough Monitor). The primary efficacy analysis included all participants who received at least one dose of study drug and had at least one post-baseline efficacy measurement. Safety analysis included all those who took at least one dose of study drug. In the second cohort, participants with CIC were randomly assigned in a study across four centres with similar design and endpoints. The study was registered with ClinicalTrials.gov (NCT02412020) and the EU Clinical Trials Register (EudraCT Number 2014-004025-40) and both cohorts are closed to new participants. FINDINGS Between Feb 13, 2015, and Feb 2, 2016, 24 participants with IPF were randomly assigned to treatment groups. 28 participants with CIC were enrolled during the same period and 27 received study treatment. In patients with IPF, PA101 reduced daytime cough frequency by 31·1% at day 14 compared with placebo; daytime cough frequency decreased from a mean 55 (SD 55) coughs per h at baseline to 39 (29) coughs per h at day 14 following treatment with PA101, versus 51 (37) coughs per h at baseline to 52 (40) cough per h following placebo treatment (ratio of least-squares [LS] means 0·67, 95% CI 0·48-0·94, p=0·0241). By contrast, no treatment benefit for PA101 was observed in the CIC cohort; mean reduction of daytime cough frequency at day 14 for PA101 adjusted for placebo was 6·2% (ratio of LS means 1·27, 0·78-2·06, p=0·31). PA101 was well tolerated in both cohorts. The incidence of adverse events was similar between PA101 and placebo treatments, most adverse events were mild in severity, and no severe adverse events or serious adverse events were reported. INTERPRETATION This study suggests that the mechanism of cough in IPF might be disease specific. Inhaled PA101 could be a treatment option for chronic cough in patients with IPF and warrants further investigation. FUNDING Patara Pharma.
Collapse
Affiliation(s)
- Surinder S Birring
- Division of Asthma, Allergy & Lung Biology, School of Transplantation, Immunology, Infection & Inflammation Sciences, Faculty of Life Sciences & Medicine, King's College London, King's Health Partners, London, UK.
| | - Marlies S Wijsenbeek
- Department of Respiratory Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sanjay Agrawal
- Department of Respiratory Medicine, Glenfield Hospital, Leicester, UK
| | | | - Helen Stone
- Department of Respiratory Medicine, Royal Stoke University Hospital, Stoke on Trent, UK
| | - Toby M Maher
- Royal Brompton Hospital, London, UK; Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | | | - Alyn H Morice
- Hull York Medical School, Castle Hill Hospital, Hull, UK
| |
Collapse
|
53
|
Zhang L, Wang T, Chang M, Kaiser C, Kim JD, Wu T, Cao X, Zhang X, Schwarz EM. Teriparatide Treatment Improves Bone Defect Healing Via Anabolic Effects on New Bone Formation and Non-Anabolic Effects on Inhibition of Mast Cells in a Murine Cranial Window Model. J Bone Miner Res 2017; 32:1870-1883. [PMID: 28556967 PMCID: PMC5555820 DOI: 10.1002/jbmr.3178] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/03/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
Investigations of teriparatide (recombinant parathyroid hormone [rPTH]) as a potential treatment for critical defects have demonstrated the predicted anabolic effects on bone formation, and significant non-anabolic effects on healing via undefined mechanisms. Specifically, studies in murine models of structural allograft healing demonstrated that rPTH treatment increased angiogenesis (vessels <30 μm), and decreased arteriogenesis (>30 μm) and mast cell numbers, which lead to decreased fibrosis and accelerated healing. To better understand these non-anabolic effects, we interrogated osteogenesis, vasculogenesis, and mast cell accumulation in mice randomized to placebo (saline), rPTH (20 μg/kg/2 days), or the mast cell inhibitor sodium cromolyn (SC) (24 μg/kg/ 2days), via longitudinal micro-computed tomography (μCT) and multiphoton laser scanning microscopy (MPLSM), in a critical calvaria defect model. μCT demonstrated that SC significantly increased defect window closure and new bone volume versus placebo (p < 0.05), although these effects were not as great as rPTH. Interestingly, both rPTH and SC have similar inhibitory effects on arteriogenesis versus placebo (p < 0.05) without affecting total vascular volume. MPLSM time-course studies in untreated mice revealed that large numbers of mast cells were detected 1 day postoperation (43 ± 17), peaked at 6 days (76 ± 6), and were still present in the critical defect at the end of the experiment on day 30 (20 ± 12). In contrast, angiogenesis was not observed until day 4, and functional vessels were first observed on 6 days, demonstrating that mast cell accumulation precedes vasculogenesis. To confirm a direct role of mast cells on osteogenesis and vasculogenesis, we demonstrated that specific diphtheria toxin-α deletion in Mcpt5-Cre-iDTR mice results in similar affects as SC treatment in WT mice. Collectively, these findings demonstrate that mast cells inhibit bone defect healing by stimulating arteriogenesis associated with fibrotic scaring, and that an efficacious non-anabolic effect of rPTH therapy on bone repair is suppression of arteriogenesis and fibrosis secondary to mast cell inhibition. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Longze Zhang
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Tao Wang
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Martin Chang
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Claire Kaiser
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Jason D Kim
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Tianyu Wu
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xiaoyi Cao
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
54
|
IL-33, IL-25, and TSLP induce a distinct phenotypic and activation profile in human type 2 innate lymphoid cells. Blood Adv 2017; 1:577-589. [PMID: 29296700 DOI: 10.1182/bloodadvances.2016002352] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/13/2017] [Indexed: 01/11/2023] Open
Abstract
Innate lymphoid cells (ILCs) represent a distinct branch of the lymphoid lineage composed of 3 major subpopulations: ILC1, ILC2, and ILC3. ILCs are mainly described as tissue-resident cells but can be detected at low levels in human blood. However, unlike mouse ILCs, there is still no consistent methodology to purify and culture these cells that enables in-depth analysis of their intrinsic biology. Here, we describe defined culture conditions for ILC2s, which allowed us to dissect the roles of interleukin 2 (IL-2), IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) individually, or in combination, in modulating ILC2 phenotype and function. We show that TSLP is important for ILC2 survival, while ILC2 activation is more dependent on IL-33, especially when in combination with IL-2 or TSLP. We found that activation of ILC2s by IL-33 and TSLP dramatically upregulated their surface expression of c-Kit and downregulated expression of the canonical markers IL-7Rα and CRTH2. IL-2 further amplified ILC2 production of IL-5, IL-13, and granulocyte-macrophage colony-stimulating factor but also induced a more natural killer (NK)-like phenotype in ILC2, with upregulation of granzyme B production by these cells. Furthermore, ILC2 plasticity was observed in serum-free SFEM II media in response to IL-33, IL-25, and TSLP stimulation and independently of IL-12 and IL-1β. This is the first comprehensive report of an in vitro culture system for human ILC2s, without the use of feeder layers, which additionally evaluates the impact of IL-25, IL-33, and TSLP alone or in combination on ILC2 surface phenotype and activation status.
Collapse
|
55
|
Tomos IP, Tzouvelekis A, Aidinis V, Manali ED, Bouros E, Bouros D, Papiris SA. Extracellular matrix remodeling in idiopathic pulmonary fibrosis. It is the 'bed' that counts and not 'the sleepers'. Expert Rev Respir Med 2017; 11:299-309. [PMID: 28274188 DOI: 10.1080/17476348.2017.1300533] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by irreversible fibrosis. Current disease pathogenesis assumes an aberrant wound healing process in response to repetitive injurious stimuli leading to apoptosis of epithelial cells, activation of fibroblasts and accumulation of extracellular matrix (ECM). Particularly, lung ECM is a highly dynamic structure that lies at the core of several physiological and developmental pathways. The scope of this review article is to summarize current knowledge on the role of ECM in the pathogenesis of IPF, unravel novel mechanistic data and identify future more effective therapeutic targets. Areas covered: The exact mechanisms through which lung microenvironment activates fibroblasts and inflammatory cells, regulates profibrotic signaling cascades through growth factors, integrins and degradation enzymes ultimately leading to excessive matrix deposition are discussed. Furthermore, the potential therapeutic usefulness of specific inhibitors of matrix deposition or activators of matrix degradation pathways are also presented. Expert commentary: With a gradually increasing worldwide incidence IPF still present a major challenge in clinical research due to its unknown etiopathogenesis and current ineffective treatment approaches. Today, there is an amenable need for more effective therapeutic targets and ECM components may represent one.
Collapse
Affiliation(s)
- Ioannis P Tomos
- a Respiratory Medicine Department , 'Attikon' University Hospital, Athens Medical School, National and Kapodistrian University of Athens , Athens , Greece
| | - Argyrios Tzouvelekis
- b Division of Immunology , Biomedical Sciences Research Center 'Alexander Fleming,' , Athens , Greece
| | - Vassilis Aidinis
- b Division of Immunology , Biomedical Sciences Research Center 'Alexander Fleming,' , Athens , Greece
| | - Effrosyni D Manali
- a Respiratory Medicine Department , 'Attikon' University Hospital, Athens Medical School, National and Kapodistrian University of Athens , Athens , Greece
| | - Evangelos Bouros
- c First Academic Department of Pneumonology, Hospital for Diseases of the Chest, 'Sotiria,' Medical School , National and Kapodistrian University of Athens , Athens , Greece
| | - Demosthenes Bouros
- c First Academic Department of Pneumonology, Hospital for Diseases of the Chest, 'Sotiria,' Medical School , National and Kapodistrian University of Athens , Athens , Greece
| | - Spyros A Papiris
- a Respiratory Medicine Department , 'Attikon' University Hospital, Athens Medical School, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
56
|
|
57
|
Clarke DL, Murray LA, Crestani B, Sleeman MA. Is personalised medicine the key to heterogeneity in idiopathic pulmonary fibrosis? Pharmacol Ther 2017; 169:35-46. [DOI: 10.1016/j.pharmthera.2016.09.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
58
|
Fukihara J, Kondoh Y. Nintedanib (OFEV) in the treatment of idiopathic pulmonary fibrosis. Expert Rev Respir Med 2016; 10:1247-1254. [DOI: 10.1080/17476348.2016.1249854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
59
|
Kanemaru A, Yamamoto K, Kawaguchi M, Fukushima T, Lin CY, Johnson MD, Camerer E, Kataoka H. Deregulated matriptase activity in oral squamous cell carcinoma promotes the infiltration of cancer-associated fibroblasts by paracrine activation of protease-activated receptor 2. Int J Cancer 2016; 140:130-141. [PMID: 27615543 DOI: 10.1002/ijc.30426] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/24/2016] [Accepted: 09/05/2016] [Indexed: 12/29/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are known to contribute to cancer progression. We have reported that cell surface expression of hepatocyte growth factor activator inhibitor 1 (HAI-1) is decreased in invasive oral squamous cell carcinoma (OSCC) cells. This study examined if HAI-1-insufficiency contributes to CAF recruitment in OSCC. Serum-free conditioned medium (SFCM) from a human OSCC line (SAS) stimulated the migration of 3 human fibroblast cell lines, NB1RGB, MRC5 and KD. SFCM from HAI-1-knockdown SAS showed an additive effect on the migration of NB1RGB and MRC5, but not KD. SAS SFCM induced protease-activated receptor-2 (PAR-2) expression in NB1RGB and MRC5, but not in KD, and a PAR-2 antagonist blocked the stimulatory effect of HAI-1 knockdown on migration of the PAR-2 expressing cell lines. Moreover, HAI-1-deficient SFCM showed additive stimulatory effects on the migration of wild-type but not PAR-2-deficient mouse fibroblasts. Therefore, the enhanced migration induced by HAI-1-insufficiency was mediated by PAR-2 activation in fibroblasts. This activation resulted from the deregulation of the activity of matriptase, a PAR-2 agonist protease. HAI-1 may thus prevent CAF recruitment to OSCC by controlling matriptase activity. When HAI-1 expression is reduced on OSCC, matriptase may contribute to CAF accumulation by paracrine activation of fibroblast PAR-2. Immunohistochemical analysis of resected OSCC revealed increased PAR2-positive CAFs in 35% (33/95) of the cases studied. The increased PAR-2 positive CAFs tended to correlate with infiltrative histology of the invasion front and shorter disease-free survival of the patients.
Collapse
Affiliation(s)
- Ai Kanemaru
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Koji Yamamoto
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Makiko Kawaguchi
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Tsuyoshi Fukushima
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| | - Chen-Yong Lin
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Michael D Johnson
- School of Medicine, Lambardi Comprehensive Cancer Centre, Georgetown University, Washington, DC
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Paris, France
| | - Hiroaki Kataoka
- Section of Oncopathology and Regenerative Biology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Japan
| |
Collapse
|
60
|
Virk H, Arthur G, Bradding P. Mast cells and their activation in lung disease. Transl Res 2016; 174:60-76. [PMID: 26845625 DOI: 10.1016/j.trsl.2016.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 12/14/2022]
Abstract
Mast cells and their activation contribute to lung health via innate and adaptive immune responses to respiratory pathogens. They are also involved in the normal response to tissue injury. However, mast cells are involved in disease processes characterized by inflammation and remodeling of tissue structure. In these diseases mast cells are often inappropriately and chronically activated. There is evidence for activation of mast cells contributing to the pathophysiology of asthma, pulmonary fibrosis, and pulmonary hypertension. They may also play a role in chronic obstructive pulmonary disease, acute respiratory distress syndrome, and lung cancer. The diverse mechanisms through which mast cells sense and interact with the external and internal microenvironment account for their role in these diseases. Newly discovered mechanisms of redistribution and interaction between mast cells, airway structural cells, and other inflammatory cells may offer novel therapeutic targets in these disease processes.
Collapse
Affiliation(s)
- Harvinder Virk
- Department of Infection, Immunity and Inflammation, Institute of Lung Health, University of Leicester, Leicester, United Kingdom
| | - Greer Arthur
- Department of Infection, Immunity and Inflammation, Institute of Lung Health, University of Leicester, Leicester, United Kingdom
| | - Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute of Lung Health, University of Leicester, Leicester, United Kingdom.
| |
Collapse
|
61
|
Arthur G, Bradding P. New Developments in Mast Cell Biology: Clinical Implications. Chest 2016; 150:680-93. [PMID: 27316557 DOI: 10.1016/j.chest.2016.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/26/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
Mast cells (MCs) are present in connective tissue and at mucosal surfaces in all classes of vertebrates. In health, they contribute to tissue homeostasis, host defense, and tissue repair via multiple receptors regulating the release of a vast stockpile of proinflammatory mediators, proteases, and cytokines. However, these potentially protective cells are a double-edged sword. When there is a repeated or long-term stimulus, MC activation leads to tissue damage and dysfunction. Accordingly, MCs are implicated in the pathophysiologic aspects of numerous diseases covering all organs. Understanding the biology of MCs, their heterogeneity, mechanisms of activation, and signaling cascades may lead to the development of novel therapies for many diseases for which current treatments are lacking or are of poor efficacy. This review will focus on updates and developments in MC biology and their clinical implications, with a particular focus on their role in respiratory diseases.
Collapse
Affiliation(s)
- Greer Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, England
| | - Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, England.
| |
Collapse
|
62
|
Kondoh H, Nishiyama T, Kikuchi Y, Fukayama M, Saito M, Kii I, Kudo A. Periostin Deficiency Causes Severe and Lethal Lung Injury in Mice With Bleomycin Administration. J Histochem Cytochem 2016; 64:441-53. [PMID: 27270966 DOI: 10.1369/0022155416652611] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/10/2016] [Indexed: 01/11/2023] Open
Abstract
Pulmonary capillary leakage followed by influx of blood fluid into the air space of lung alveoli is a crucial step in the progression of acute lung injury (ALI). This influx is due to increased permeability of the alveolar-capillary barrier. The extracellular matrix (ECM) between the capillary and the epithelium would be expected to be involved in prevention of the influx; however, the role of the ECM remains to be addressed. Here, we show that the ECM architecture organized by periostin, a matricellular protein, plays a pivotal role in the survival of bleomycin-exposed mice. Periostin was localized in the alveolar walls. Although periostin-null mice displayed no significant difference in lung histology and air-blood permeability, they exhibited early lethality in a model of bleomycin-induced lung injury, compared with their wild-type counterparts. This early lethality may have been due to increased pulmonary leakage of blood fluid into the air space in the bleomycin-exposed periostin-null mice. These results suggest that periostin in the ECM architecture prevents pulmonary leakage of blood fluid, thus increasing the survival rate in mice with ALI. Thus, this study provides an evidence for the protective role of the ECM architecture in the lung alveoli.
Collapse
Affiliation(s)
- Hirofumi Kondoh
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan (HK, TN, IK, AK)
| | - Takashi Nishiyama
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan (HK, TN, IK, AK)
| | - Yoshinao Kikuchi
- Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (YK, MF)
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan (YK, MF)
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, Jikei University School of Medicine, Tokyo, Japan (MS)
| | - Isao Kii
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan (HK, TN, IK, AK),Pathophysiological and Health Science Team, Imaging Application Group, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe, Japan (IK)
| | - Akira Kudo
- Department of Biological Information, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan (HK, TN, IK, AK)
| |
Collapse
|
63
|
Gao H, Deng H, Xu H, Yang Q, Zhou Y, Zhang J, Zhao D, Liu F. MicroRNA-223 promotes mast cell apoptosis by targeting the insulin-like growth factor 1 receptor. Exp Ther Med 2016; 11:2171-2176. [PMID: 27284298 PMCID: PMC4887759 DOI: 10.3892/etm.2016.3227] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/11/2016] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to examine the functional role of miR-223 in the regulation of mast cell apoptosis. Overexpressed miR-223 in mast cells transfected by Lipofectamine 2000 was used as a model, and miR-223 was found to promote mast cell apoptosis. To investigate the underlying mechanisms involved, the potential and putative target molecules of miR-223 were detected by bioinformatical analysis using predictive software, and western blotting. Insulin-like growth factor-1 receptor (IGF-1R) was found to be the functional target of miR-223 in the promotion of cell apoptosis. The downstream PI3K/protein kinase B (Akt) signaling pathway was also inhibited, and signaling was mediated by IGF-1R. Furthermore, the relative luciferase activity of the reporter containing the 3′-untranslated region (3′-UTR) of IGF-1R was significantly suppressed, while suppression of miR-223-inhibited IGF-1R protein expression was also observed. In conclusion, the results suggest that IGF-1R is the functional target for miR-223 promotion of cell apoptosis, and its downstream PI3K/Akt signaling pathway was suppressed by miR-223 through targeting of IGF-1R.
Collapse
Affiliation(s)
- Haiyan Gao
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China; Department of Pediatrics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Huan Deng
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Xu
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qianyuan Yang
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yao Zhou
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiamin Zhang
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Deyu Zhao
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Feng Liu
- Department of Respiratory Medicine, Nanjing Children's Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
64
|
|
65
|
Elevated protein arginine methyltransferase 1 expression regulates fibroblast motility in pulmonary fibrosis. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2678-88. [DOI: 10.1016/j.bbadis.2015.09.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/14/2015] [Accepted: 09/16/2015] [Indexed: 12/29/2022]
|
66
|
White MJV, Gomer RH. Trypsin, Tryptase, and Thrombin Polarize Macrophages towards a Pro-Fibrotic M2a Phenotype. PLoS One 2015; 10:e0138748. [PMID: 26407067 PMCID: PMC4583378 DOI: 10.1371/journal.pone.0138748] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023] Open
Abstract
For both wound healing and the formation of a fibrotic lesion, circulating monocytes enter the tissue and differentiate into fibroblast-like cells called fibrocytes and pro-fibrotic M2a macrophages, which together with fibroblasts form scar tissue. Monocytes can also differentiate into classically activated M1 macrophages and alternatively activated M2 macrophages. The proteases thrombin, which is activated during blood clotting, and tryptase, which is released by activated mast cells, potentiate fibroblast proliferation and fibrocyte differentiation, but their effect on macrophages is unknown. Here we report that thrombin, tryptase, and the protease trypsin bias human macrophage differentiation towards a pro-fibrotic M2a phenotype expressing high levels of galectin-3 from unpolarized monocytes, or from M1 and M2 macrophages, and that these effects appear to operate through protease-activated receptors. These results suggest that proteases can initiate scar tissue formation by affecting fibroblasts, fibrocytes, and macrophages.
Collapse
Affiliation(s)
- Michael J. V. White
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
67
|
Witkowski M, Katkiewicz M, Zając S, Kochan J. Effect of Long-term Hyperimmunization on the Presence of Mast Cells in the Endometrium of the Mare. J Equine Vet Sci 2015. [DOI: 10.1016/j.jevs.2015.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
68
|
Lin C, von der Thüsen J, Daalhuisen J, ten Brink M, Crestani B, van der Poll T, Borensztajn K, Spek CA. Pharmacological Targeting of Protease-Activated Receptor 2 Affords Protection from Bleomycin-Induced Pulmonary Fibrosis. Mol Med 2015; 21:576-83. [PMID: 26147947 DOI: 10.2119/molmed.2015.00094] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/29/2015] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis is the most devastating diffuse fibrosing lung disease that remains refractory to therapy. Despite increasing evidence that protease-activated receptor 2 (PAR-2) contributes to fibrosis, its importance in pulmonary fibrosis is under debate. We addressed whether PAR-2 deficiency persistently reduces bleomycin-induced pulmonary fibrosis or merely delays disease progression and whether pharmacological PAR-2 inhibition limits experimental pulmonary fibrosis. Bleomycin was instilled intranasally into wild-type or PAR-2-deficient mice in the presence/absence of a specific PAR-2 antagonist (P2pal-18S). Pulmonary fibrosis was consistently reduced in PAR-2-deficient mice throughout the fibrotic phase, as evident from reduced Ashcroft scores (29%) and hydroxyproline levels (26%) at d 28. Moreover, P2pal-18S inhibited PAR-2-induced profibrotic responses in both murine and primary human pulmonary fibroblasts (p < 0.05). Once daily treatment with P2pal-18S reduced the severity and extent of fibrotic lesions in lungs of bleomycin-treated wild-type mice but did not further reduce fibrosis in PAR-2-deficient mice. Importantly, P2pal-18S treatment starting even 7 d after the onset of fibrosis limits pulmonary fibrosis as effectively as when treatment was started together with bleomycin instillation. Overall, PAR-2 contributes to the progression of pulmonary fibrosis, and targeting PAR-2 may be a promising therapeutic strategy for treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Cong Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Joost Daalhuisen
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Marieke ten Brink
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Bruno Crestani
- Assistance Publique-Hôpitaux de Paris, Department of Pulmonology A, Reference Center for Rare Lung Diseases, Bichat-Claude Bernard University Hospital, Paris, France
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Inserm UMR1152, Medical School Xavier Bichat, Paris, France.,Département Hospitalo-universtaire FIRE (Fibrosis, Inflammation and Remodeling) and LabEx Inflamex, Paris, France
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
69
|
Kosanovic D, Luitel H, Dahal BK, Cornitescu T, Janssen W, Danser AHJ, Garrelds IM, De Mey JGR, Fazzi G, Schiffers P, Iglarz M, Fischli W, Ghofrani HA, Weissmann N, Grimminger F, Seeger W, Reiss I, Schermuly RT. Chymase: a multifunctional player in pulmonary hypertension associated with lung fibrosis. Eur Respir J 2015; 46:1084-94. [PMID: 26113671 DOI: 10.1183/09031936.00018215] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 04/09/2015] [Indexed: 12/22/2022]
Abstract
Limited literature sources implicate mast-cell mediator chymase in the pathologies of pulmonary hypertension and pulmonary fibrosis. However, there is no evidence on the contribution of chymase to the development of pulmonary hypertension associated with lung fibrosis, which is an important medical condition linked with increased mortality of patients who already suffer from a life-threatening interstitial lung disease.The aim of this study was to investigate the role of chymase in this particular pulmonary hypertension form, by using a bleomycin-induced pulmonary hypertension model.Chymase inhibition resulted in attenuation of pulmonary hypertension and pulmonary fibrosis, as evident from improved haemodynamics, decreased right ventricular remodelling/hypertrophy, pulmonary vascular remodelling and lung fibrosis. These beneficial effects were associated with a strong tendency of reduction in mast cell number and activity, and significantly diminished chymase expression levels. Mechanistically, chymase inhibition led to attenuation of transforming growth factor β1 and matrix-metalloproteinase-2 contents in the lungs. Furthermore, chymase inhibition prevented big endothelin-1-induced vasoconstriction of the pulmonary arteries.Therefore, chymase plays a role in the pathogenesis of pulmonary hypertension associated with pulmonary fibrosis and may represent a promising therapeutic target. In addition, this study may provide valuable insights on the contribution of chymase in the pulmonary hypertension context, in general, regardless of the pulmonary hypertension form.
Collapse
Affiliation(s)
- Djuro Kosanovic
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Division of Neonatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Himal Luitel
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Bhola Kumar Dahal
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Risk Factor Modification Centre (RFMC), St. Michael's Hospital, Toronto, ON, Canada
| | - Teodora Cornitescu
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Wiebke Janssen
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - A H Jan Danser
- Dept of Pharmacology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Ingrid M Garrelds
- Dept of Pharmacology, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Jo G R De Mey
- Dept of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Gregorio Fazzi
- Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Paul Schiffers
- Dept of Pharmacology, Maastricht University, Maastricht, The Netherlands
| | - Marc Iglarz
- Actelion Pharmaceuticals Ltd, Allschwill, Switzerland
| | | | - Hossein Ardeschir Ghofrani
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Norbert Weissmann
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Friedrich Grimminger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany
| | - Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Max-Planck-Institute for Heart and Lung Research, Member of the German Center for Lung Research, Bad Nauheim, Germany
| | - Irwin Reiss
- Division of Neonatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands Both authors contributed equally
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research, Giessen, Germany Both authors contributed equally
| |
Collapse
|
70
|
Mast cells in airway diseases and interstitial lung disease. Eur J Pharmacol 2015; 778:125-38. [PMID: 25959386 DOI: 10.1016/j.ejphar.2015.04.046] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 12/31/2022]
Abstract
Mast cells are major effector cells of inflammation and there is strong evidence that mast cells play a significant role in asthma pathophysiology. There is also a growing body of evidence that mast cells contribute to other inflammatory and fibrotic lung diseases such as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. This review discusses the role that mast cells play in airway diseases and highlights how mast cell microlocalisation within specific lung compartments and their cellular interactions are likely to be critical for their effector function in disease.
Collapse
|
71
|
Grimminger F, Günther A, Vancheri C. The role of tyrosine kinases in the pathogenesis of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45:1426-33. [PMID: 25745048 DOI: 10.1183/09031936.00149614] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 12/06/2014] [Indexed: 12/23/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with a median survival time from diagnosis of 2-3 years. Although the pathogenic pathways have not been fully elucidated, IPF is believed to be caused by persistent epithelial injury in genetically susceptible individuals. Tyrosine kinases are involved in a range of signalling pathways that are essential for cellular homeostasis. However, there is substantial evidence from in vitro studies and animal models that receptor tyrosine kinases, such as the platelet-derived growth factor receptor, vascular endothelial growth factor receptor and fibroblast growth factor receptor, and non-receptor tyrosine kinases, such as the Src family, play critical roles in the pathogenesis of pulmonary fibrosis. For example, the expression and release of tyrosine kinases are altered in patients with IPF, while specific tyrosine kinases stimulate the proliferation of lung fibroblasts in vitro. Agents that inhibit tyrosine kinases have shown anti-fibrotic and anti-inflammatory effects in animal models of pulmonary fibrosis. Recently, the tyrosine kinase inhibitor nintedanib has shown positive results in two phase III trials in patients with IPF. Here, we summarise the evidence for involvement of specific tyrosine kinases in the pathogenesis of IPF and the development of tyrosine kinase inhibitors as treatments for IPF.
Collapse
Affiliation(s)
- Friedrich Grimminger
- Dept of Hematology/Oncology, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Andreas Günther
- Dept of Pulmonary and Critical Care Medicine, ILD Program, University Hospital of Giessen and Marburg, Marburg, Germany
| | - Carlo Vancheri
- "Regional Centre for Rare Lung Diseases", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
72
|
Abstract
Interstitial lung disease (ILD) encompasses a large and diverse group of pathological conditions that share similar clinical, radiological and pathological manifestations, despite potentially having quite different aetiologies and comorbidities. Idiopathic pulmonary fibrosis (IPF) represents probably the most aggressive form of ILD and systemic sclerosis is a multiorgan fibrotic disease frequently associated with ILD. Although the aetiology of these disorders remains unknown, in this review we analyse the pathogenic mechanisms by cell of interest (fibroblast, fibrocyte, myofibroblast, endothelial and alveolar epithelial cells and immune competent cells). New insights into the complex cellular contributions and interactions will be provided, comparing the role of cell subsets in the pathogenesis of IPF and systemic sclerosis. Distinct cell populations contribute to the complex pathogenesis of IPF and systemic sclerosis-associated ILDhttp://ow.ly/AjFaz
Collapse
|
73
|
Landolina N, Gangwar RS, Levi-Schaffer F. Mast cells' integrated actions with eosinophils and fibroblasts in allergic inflammation: implications for therapy. Adv Immunol 2015; 125:41-85. [PMID: 25591464 DOI: 10.1016/bs.ai.2014.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mast cells (MCs) and eosinophils (Eos) are the key players in the development of allergic inflammation (AI). Their cross-talk, named the Allergic Effector Unit (AEU), takes place through an array of soluble mediators and ligands/receptors interactions that enhance the functions of both the cells. One of the salient features of the AEU is the CD48/2B4 receptor/ligand binding complex. Furthermore, MCs and Eos have been demonstrated to play a role not only in AI but also in the modulation of its consequence, i.e., fibrosis/tissue remodeling, by directly influencing fibroblasts (FBs), the main target cells of these processes. In turn, FBs can regulate the survival, activity, and phenotype of both MCs and Eos. Therefore, a complex three players, MCs/Eos/FBs interaction, can take place in various stages of AI. The characterization of the soluble and physical mediated cross talk among these three cells might lead to the identification of both better and novel targets for the treatment of allergy and its tissue remodeling consequences.
Collapse
Affiliation(s)
- Nadine Landolina
- Department of Pharmacology, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Roopesh Singh Gangwar
- Department of Pharmacology, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Francesca Levi-Schaffer
- Department of Pharmacology, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
74
|
The role of mouse mast cell proteases in the proliferative phase of wound healing in microdeformational wound therapy. Plast Reconstr Surg 2014; 134:459-467. [PMID: 24814421 DOI: 10.1097/prs.0000000000000432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Stored in the secretory granules of cutaneous mouse mast cells are mouse mast cell proteases (mMCP-4, -5, and -6). Using transgenic mouse lines that lacked these enzymes, it was shown that mMCP-4 and mMCP-5 modulate the outcome of burn-induced skin injury. Whether or not these proteases also play a role in the repair of surgically damaged skin, with or without microdeformational wound therapy, remains to be determined. METHODS Wild-type C57BL/6 mice and transgenic C57BL/6 mouse lines lacking mMCP-4, -5, or -6 were subjected to surgical wounding of their skin. Wounds were splinted with a stabilizing patch, and the mice received either microdeformational wound therapy (n = 5) or occlusive dressing (n = 5) for 7 days. Wound healing parameters were assessed in the proliferative phase. RESULTS Cell proliferation in the wounded wild-type mice receiving microdeformational wound therapy was 60 ± 3 percent. Cell proliferation was only 35 ± 5 percent, 25 ± 5 percent, and 45 ± 4 percent for the treated mMCP-4-, mMCP-5-, and mMCP-6-null mice, respectively (p = 0.005). Blood vessel sprouting was higher in the control mice with microdeformational wound therapy (170 ± 40 vessels/high-power field) compared with mouse mast cell protease 6-null mice with microdeformational wound therapy (70 ± 20 vessels/high-power field; p = 0.005), and higher in the control mice with occlusive dressing (110 ± 30 vessels/high-power field) compared with mMCP-4-null mice with occlusive dressing (50 ± 20 vessels/high-power field; p = 0.01). Qualitatively, the granulation tissue of all the protease-deficient groups receiving microdeformational wound therapy was disrupted. CONCLUSION Results suggest that mouse mast cell proteases 4, 5, and 6 are mediators of the critical role mast cells play in microdeformational wound therapy in the proliferative phase of healing.
Collapse
|
75
|
Increase in both angiogenic and angiostatic mediators in patients with idiopathic pulmonary fibrosis. ACTA ACUST UNITED AC 2014; 62:391-4. [DOI: 10.1016/j.patbio.2014.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 07/15/2014] [Indexed: 11/20/2022]
|
76
|
White MJV, Galvis-Carvajal E, Gomer RH. A brief exposure to tryptase or thrombin potentiates fibrocyte differentiation in the presence of serum or serum amyloid p. THE JOURNAL OF IMMUNOLOGY 2014; 194:142-50. [PMID: 25429068 DOI: 10.4049/jimmunol.1401777] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A key question in both wound healing and fibrosis is the trigger for the initial formation of scar tissue. To help form scar tissue, circulating monocytes enter the tissue and differentiate into fibroblast-like cells called fibrocytes, but fibrocyte differentiation is strongly inhibited by the plasma protein serum amyloid P (SAP), and healthy tissues contain very few fibrocytes. In wounds and fibrotic lesions, mast cells degranulate to release tryptase, and thrombin mediates blood clotting in early wounds. Tryptase and thrombin are upregulated in wound healing and fibrotic lesions, and inhibition of these proteases attenuates fibrosis. We report that tryptase and thrombin potentiate human fibrocyte differentiation at biologically relevant concentrations and exposure times, even in the presence of concentrations of serum and SAP that normally completely inhibit fibrocyte differentiation. Fibrocyte potentiation by thrombin and tryptase is mediated by protease-activated receptors 1 and 2, respectively. Together, these results suggest that tryptase and thrombin may be an initial trigger to override SAP inhibition of fibrocyte differentiation to initiate scar tissue formation.
Collapse
Affiliation(s)
- Michael J V White
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| | | | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843-3474
| |
Collapse
|
77
|
Behzad H, Sharma A, Mousavizadeh R, Lu A, Scott A. Mast cells exert pro-inflammatory effects of relevance to the pathophyisology of tendinopathy. Arthritis Res Ther 2014; 15:R184. [PMID: 24517261 PMCID: PMC3978883 DOI: 10.1186/ar4374] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/25/2013] [Indexed: 02/06/2023] Open
Abstract
Introduction We have previously found an increased mast cell density in tendon biopsies from patients with patellar tendinopathy compared to controls. This study examined the influence of mast cells on basic tenocyte functions, including production of the inflammatory mediator prostaglandin E2 (PGE2), extracellular matrix remodeling and matrix metalloproteinase (MMP) gene transcription, and collagen synthesis. Methods Primary human tenocytes were stimulated with an established human mast cell line (HMC-1). Extracellular matrix remodeling was studied by culturing tenocytes in a three-dimensional collagen lattice. Survival/proliferation was assessed with the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium salt (MTS) assay. Levels of mRNA for COX-2, COL1A1, MMP1, and MMP7 were determined by quantitative real-time polymerase chain reaction (qPCR). Cox-2 protein level was assessed by Western blot analysis and type I procollagen was detected by immunofluorescent staining. PGE2 levels were determined using an enzyme-linked immunosorbent assay (ELISA). Results Mast cells stimulated tenocytes to produce increased levels of COX-2 and the pro-inflammatory mediator PGE2, which in turn decreased COL1A1 mRNA expression. Additionally, mast cells reduced the type I procollagen protein levels produced by tenocytes. Transforming growth factor beta 1 (TGF-β1) was responsible for the induction of Cox-2 and PGE2 by tenocytes. Mast cells increased MMP1 and MMP7 transcription and increased the contraction of a three-dimensional collagen lattice by tenocytes, a phenomenon which was blocked by a pan-MMP inhibitor (Batimastat). Conclusion Our data demonstrate that mast cell-derived PGE2 reduces collagen synthesis and enhances expression and activities of MMPs in human tenocytes.
Collapse
|
78
|
Abstract
PURPOSE OF REVIEW To summarize the new knowledge on tissue remodeling in the context of lung diseases. Tissue remodeling includes changes in cells: differentiation; response to growths factors, hormones, or environmental factors; and composition of the extracellular matrix. So, can one trigger cause them all or are they independently regulated? RECENT FINDINGS New evidence from clinical and experimental studies strengthened the view that a susceptibility to remodeling can be initiated in early life and be re-activated by environmental triggers later in life. Many studies further support the idea that TGF-β plays the central role in the pathogenesis of remodeling and fibrosis. However, the activation pathways and the end-effect of TGF-β activation seems to be distinctive of disease and effecter cell specific patterns. The existing animal models do not properly reflect the human disease and thus have to be further improved. SUMMARY The central role of TGF-β on pathological mechanisms leading to remodeling and fibrosis has been further confirmed. However, the questions of why TGF-β is activated as well as its disease and cell type specific mode of action remain to be answered. Based on clinical data redefining the term 'tissue remodeling' in a disease and cell type specific way should be considered.
Collapse
|
79
|
Zaidi AK, Mussarat A, Mishra A. Diagnostic and therapeutic strategies for eosinophilic esophagitis. ACTA ACUST UNITED AC 2014; 11:351-367. [PMID: 25400904 DOI: 10.2217/cpr.14.31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Eosinophilic esophagitis (EoE) is a recently recognized allergic disorder, characterized by eosophageal dysfunction, accumulation of ≥15 eosinophils/high-powered field, eosinophil microabssess, basal cell hyperplasia, extracellular eosinophilic granules in the esophageal epithelial mucosal biopsy and a lack of response to a 8-week proton pump inhibitor treatment. Despite the increased incidences and considerable progress made in understanding EoE pathogenesis, there are limited diagnostic and therapeutic options available for EoE. Currently, the only criterion for diagnosing EoE is repetitive esophageal endoscopic biopsies and histopathological evaluation. Antigen elimination or corticosteroid therapies are effective therapies for EoE but are expensive and have limitations, if continued in the long term. Hence, there is a great necessity for novel noninvasive diagnostic biomarkers that can easily diagnose EoE and assess effectiveness of therapy. Herein, we have provided an update on key molecules involved in the disease initiation, and progression and proposed novel noninvasive diagnostic molecules and strategies for EoE therapy.
Collapse
Affiliation(s)
- Asifa K Zaidi
- Department of Medicine, Section of Pulmonary Diseases Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112-2699, USA
| | - Ahad Mussarat
- Department of Medicine, Section of Pulmonary Diseases Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112-2699, USA
| | - Anil Mishra
- Department of Medicine, Section of Pulmonary Diseases Tulane Eosinophilic Disorder Center, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112-2699, USA
| |
Collapse
|
80
|
Ding WY, Liu L, Wang ZH, Tang MX, Ti Y, Han L, Zhang L, Zhang Y, Zhong M, Zhang W. FP-receptor gene silencing ameliorates myocardial fibrosis and protects from diabetic cardiomyopathy. J Mol Med (Berl) 2014; 92:629-40. [PMID: 24500109 DOI: 10.1007/s00109-013-1119-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 12/02/2013] [Accepted: 12/06/2013] [Indexed: 11/25/2022]
Abstract
UNLABELLED Prostaglandin F2(α)-F-prostanoid (PGF2(α)-FP) receptor is closely related to insulin resistance, which plays a causal role in the pathogenesis of diabetic cardiomyopathy (DCM). We sought to reveal whether PGF2(α)-FP receptor plays an important part in modulating DCM and the mechanisms involved. We established the type 2 diabetes rat model by high-fat diet and low-dose streptozotocin (STZ) and then evaluated its characteristics by metabolite tests, Western blot analysis for FP-receptor expression, histopathologic analyses of cardiomyocyte density and fibrosis area. Next, we used gene silencing to investigate the role of FP receptor in the pathophysiologic features of DCM. Our study showed elevated cholesterol, triglyceride, glucose, and insulin levels, severe insulin resistance, and FP-receptor overexpression in diabetic rats. The collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) were higher in the diabetic group than the control group (CVF% 10.99 ± 0.99 vs 1.59 ± 0.18, P < 0.05; PVCA/LA% 17.07 ± 2.61 vs 2.86 ± 0.69, P < 0.05). We found that the silencing of FP receptor decreased cholesterol, triglyceride, glucose, and insulin levels and ameliorated insulin resistance. The CVF and PVCF/LA were significantly downregulated in FP-receptor short hairpin RNA (shRNA) treatment group (FP-receptor shRNA group vs vehicle group: CVF% 5.59 ± 0.92 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 4.74 ± 1.57 vs 14.79 ± 2.22, P < 0.05; FP-receptor shRNA + PGF2(α) group vs vehicle group : CVF% 5.19 ± 0.79 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 5.96 ± 1.15 vs 14.79 ± 2.22, P < 0.05, respectively). Furthermore, with FP-receptor gene silencing, the activated protein kinase C (PKC) and Rho kinase were significantly decreased, and the blunted phosphorylation of Akt was restored. FP-receptor gene silencing may exert a protective effect on DCM by improving myocardial fibrosis, suggesting a new therapeutic approach for human DCM. KEY MESSAGES FP-receptor gene silencing improves glucose tolerance and insulin resistance in type 2 diabetes (T2D). FP-receptor gene silencing modulates the activities of PKC/Rho and Akt signaling pathways in T2D. FP-receptor gene silencing decreases collagen expression and ameliorates myocardial fibrosis in T2D. FP-receptor gene silencing protects from diabetic cardiomyopathy in T2D.
Collapse
Affiliation(s)
- Wen-yuan Ding
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Moiseeva EP, Straatman KR, Leyland ML, Bradding P. CADM1 controls actin cytoskeleton assembly and regulates extracellular matrix adhesion in human mast cells. PLoS One 2014; 9:e85980. [PMID: 24465823 PMCID: PMC3899107 DOI: 10.1371/journal.pone.0085980] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/03/2013] [Indexed: 12/21/2022] Open
Abstract
CADM1 is a major receptor for the adhesion of mast cells (MCs) to fibroblasts, human airway smooth muscle cells (HASMCs) and neurons. It also regulates E-cadherin and alpha6beta4 integrin in other cell types. Here we investigated a role for CADM1 in MC adhesion to both cells and extracellular matrix (ECM). Downregulation of CADM1 in the human MC line HMC-1 resulted not only in reduced adhesion to HASMCs, but also reduced adhesion to their ECM. Time-course studies in the presence of EDTA to inhibit integrins demonstrated that CADM1 provided fast initial adhesion to HASMCs and assisted with slower adhesion to ECM. CADM1 downregulation, but not antibody-dependent CADM1 inhibition, reduced MC adhesion to ECM, suggesting indirect regulation of ECM adhesion. To investigate potential mechanisms, phosphotyrosine signalling and polymerisation of actin filaments, essential for integrin-mediated adhesion, were examined. Modulation of CADM1 expression positively correlated with surface KIT levels and polymerisation of cortical F-actin in HMC-1 cells. It also influenced phosphotyrosine signalling and KIT tyrosine autophosphorylation. CADM1 accounted for 46% of surface KIT levels and 31% of F-actin in HMC-1 cells. CADM1 downregulation resulted in elongation of cortical actin filaments in both HMC-1 cells and human lung MCs and increased cell rigidity of HMC-1 cells. Collectively these data suggest that CADM1 is a key adhesion receptor, which regulates MC net adhesion, both directly through CADM1-dependent adhesion, and indirectly through the regulation of other adhesion receptors. The latter is likely to occur via docking of KIT and polymerisation of cortical F-actin. Here we propose a stepwise model of adhesion with CADM1 as a driving force for net MC adhesion.
Collapse
Affiliation(s)
- Elena P. Moiseeva
- Institute for Lung Health, Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
- * E-mail:
| | - Kees R. Straatman
- Centre for Core Biotechnology Services, University of Leicester, Leicester, United Kingdom
| | - Mark L. Leyland
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
| | - Peter Bradding
- Institute for Lung Health, Dept. of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
82
|
Clarke DL, Carruthers AM, Mustelin T, Murray LA. Matrix regulation of idiopathic pulmonary fibrosis: the role of enzymes. FIBROGENESIS & TISSUE REPAIR 2013; 6:20. [PMID: 24279676 PMCID: PMC4176485 DOI: 10.1186/1755-1536-6-20] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/13/2013] [Indexed: 12/22/2022]
Abstract
Repairing damaged tissues is an essential homeostatic mechanism that enables clearance of dead or damaged cells after injury, and the maintenance of tissue integrity. However, exaggeration of this process in the lung can lead to the development of fibrotic scar tissue. This is characterized by excessive accumulation of extracellular matrix (ECM) components such as fibronectin, proteoglycans, hyaluronic acid, and interstitial collagens. After tissue injury, or a breakdown of tissue integrity, a cascade of events unfolds to maintain normal tissue homeostasis. Inflammatory mediators are released from injured epithelium, leading to both platelet activation and inflammatory cell migration. Inflammatory cells are capable of releasing multiple pro-inflammatory and fibrogenic mediators such as transforming growth factor (TGF)β and interleukin (IL)-13, which can trigger myofibroblast proliferation and recruitment. The myofibroblast population is also expanded as a result of epithelial cells undergoing epithelial-to-mesenchymal transition and of the activation of resident fibroblasts, leading to ECM deposition and tissue remodeling. In the healthy lung, wound healing then proceeds to restore the normal architecture of the lung; however, fibrosis can develop when the wound is severe, the tissue injury persists, or the repair process becomes dysregulated. Understanding the processes regulating aberrant wound healing and the matrix in the chronic fibrotic lung disease idiopathic pulmonary fibrosis (IPF), is key to identifying new treatments for this chronic debilitating disease. This review focuses primarily on the emerging role of enzymes in the lungs of patients with IPF. Elevated expression of a number of enzymes that can directly modulate the ECM has been reported, and recent data indicates that modulating the activity of these enzymes can have a downstream effect on fibrotic tissue remodeling.
Collapse
|