1
|
Makled MN, El-Sheakh AR. Fingolimod attenuates ovalbumin-induced airway inflammation via inhibiting MAPK/ERK signaling in mice. J Biochem Mol Toxicol 2023; 37:e23266. [PMID: 36468814 DOI: 10.1002/jbt.23266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/06/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
The current study was designed to investigate the potential anti-inflammatory and antioxidant effects of fingolimod against Ovalbumin (Ova)-induced allergic airway inflammation compared to dexamethasone. Fingolimod was given (0.5 mg/kg/day, p.o.) for sensitized mice 1 h before Ova challenge from Days 19 to 24. Fingolimod significantly inhibited Ova-induced elevation of inflammatory cells and eosinophils numbers in bronchoalveolar lavage fluid (BALF) and reduced concentrations of immunoglobulin E in serum and of sphingosine-1-phosphate, interleukin (IL)-4, and IL-13 in BALF. Fingolimod inhibited microvascular leakage and edema as reflected by the decreased lung/body weight index. These findings were supported by histopathological examination results showing that fingolimod substantially decreased perivascular edema and inflammatory cell infiltration. Fingolimod also attenuated Ova-induced oxidative stress as evidenced by decreased malondialdehyde concentration along with increasing concentrations of reduced glutathione and superoxide dismutase in lung tissues. Fingolimod also significantly decreased monocyte chemoattractant protein-1 (MCP-1), p-ERK, and p-P38 in lung tissues of Ova-challenged mice. In conclusion, the current study demonstrated the anti-inflammatory and antioxidant effects of fingolimod in allergic airway inflammation that might be associated with the downregulation of mitogen activated kinases signaling to decrease T helper 2 cytokine secretion (IL-4 and IL-13) and MCP-1 expression, along with the inhibition of oxidative stress.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.,Future Studies and Risks Management & National Committee of Drugs, Academy of Scientific Research, Ministry of Higher Education, ElSayeda Zeinab, Egypt
| |
Collapse
|
2
|
Prospective use of amniotic mesenchymal stem cell metabolite products for tissue regeneration. J Biol Eng 2023; 17:11. [PMID: 36759827 PMCID: PMC9912508 DOI: 10.1186/s13036-023-00331-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Chronic disease can cause tissue and organ damage constituting the largest obstacle to therapy which, in turn, reduces patients' quality-adjusted life-year. Degenerative diseases such as osteoporosis, Alzheimer's disease, Parkinson's disease, and infectious conditions such as hepatitis, cause physical injury to organs. Moreover, damage resulting from chronic conditions such as diabetes can also culminate in the loss of organ function. In these cases, organ transplantation constitutes the therapy of choice, despite the associated problems of immunological rejection, potential disease transmission, and high morbidity rates. Tissue regeneration has the potential to heal or replace tissues and organs damaged by age, disease, or trauma, as well as to treat disabilities. Stem cell use represents an unprecedented strategy for these therapies. However, product availability and mass production remain challenges. A novel therapeutic alternative involving amniotic mesenchymal stem cell metabolite products (AMSC-MP) has been developed using metabolites from stem cells which contain cytokines and growth factors. Its potential role in regenerative therapy has recently been explored, enabling broad pharmacological applications including various gastrointestinal, lung, bladder and renal conditions, as well as the treatment of bone wounds, regeneration and skin aging due to its low immunogenicity and anti-inflammatory effects. The various kinds of growth factors present in AMSC-MP, namely bFGF, VEGF, TGF-β, EGF and KGF, have their respective functions and activities. Each growth factor is formed by different proteins resulting in molecules with various physicochemical properties and levels of stability. This knowledge will assist in the manufacture and application of AMSC-MP as a therapeutic agent.
Collapse
|
3
|
Immunophenotypic characterization and therapeutics effects of human bone marrow- and umbilical cord-derived mesenchymal stromal cells in an experimental model of sepsis. Exp Cell Res 2021; 399:112473. [PMID: 33428902 DOI: 10.1016/j.yexcr.2021.112473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/11/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022]
Abstract
Sepsis is a complicated multi-system disorder characterized by a dysregulated host response to infection. Despite substantial progress in the understanding of mechanisms of sepsis, translation of these advances into clinically effective therapies remains challenging. Mesenchymal Stromal Cells (MSCs) possess immunomodulatory properties that have shown therapeutic promise in preclinical models of sepsis. The therapeutic effects of MSCs may vary depending on the source and type of these cells. In this comparative study, the gene expression pattern and surface markers of bone marrow-derived MSCs (BM-MSCs) and umbilical cord-derived MSCs (UC-MSCs) as well as their therapeutic effects in a clinically relevant mouse model of polymicrobial sepsis, cecal ligation and puncture (CLP), were investigated. The results showed remarkable differences in gene expression profile, surface markers and therapeutic potency in terms of enhancing survival and pro/anti-inflammatory responses between the two MSC types. BM-MSCs improved survival concomitant with an enhanced systemic bacterial clearance and improved inflammatory profile post CLP surgery. Despite some improvement in the inflammatory profile of the septic animals, treatment with UC-MSCs did not enhance survival or bacterial clearance. Overall, the beneficial therapeutic effects of BM-MSCs over UC-MSCs may likely be attributed to their pro-inflammatory function, and to some extent anti-inflammatory features, reflected in their gene expression pattern enhancing macrophage polarization to M1/M2 phenotypes resulting in a balanced pro- and anti-inflammatory response against polymicrobial sepsis.
Collapse
|
4
|
Airway Epithelial Dysfunction in Asthma: Relevant to Epidermal Growth Factor Receptors and Airway Epithelial Cells. J Clin Med 2020; 9:jcm9113698. [PMID: 33217964 PMCID: PMC7698733 DOI: 10.3390/jcm9113698] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Airway epithelium plays an important role as the first barrier from external pathogens, including bacteria, viruses, chemical substances, and allergic components. Airway epithelial cells also have pivotal roles as immunological coordinators of defense mechanisms to transfer signals to immunologic cells to eliminate external pathogens from airways. Impaired airway epithelium allows the pathogens to remain in the airway epithelium, which induces aberrant immunological reactions. Dysregulated functions of asthmatic airway epithelium have been reported in terms of impaired wound repair, fragile tight junctions, and excessive proliferation, leading to airway remodeling, which contributes to aberrant airway responses caused by external pathogens. To maintain airway epithelium integrity, a family of epidermal growth factor receptors (EGFR) have pivotal roles in mechanisms of cell growth, proliferation, and differentiation. There are extensive studies focusing on the relation between EGFR and asthma pathophysiology, which describe airway remodeling, airway hypermucus secretion, as well as immunological responses of airway inflammation. Furthermore, the second EGFR family member, erythroblastosis oncogene B2 (ErbB2), has been recognized to be involved with impaired wound recovery and epithelial differentiation in asthmatic airway epithelium. In this review, the roles of the EGFR family in asthmatic airway epithelium are focused on to elucidate the pathogenesis of airway epithelial dysfunction in asthma.
Collapse
|
5
|
Wang J, Sun H, Liu Y. The proliferative and anti-apoptosis functions of KGF/KGFR contributes to bronchial epithelial repair in asthma. Pulm Pharmacol Ther 2020; 63:101931. [PMID: 32818548 DOI: 10.1016/j.pupt.2020.101931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/05/2020] [Accepted: 07/12/2020] [Indexed: 10/23/2022]
Abstract
This study aimed to investigate the effect of keratinocyte growth factor (KGF) on the apoptosis, proliferation, damage repair, intercellular adhesion, and inflammatory cytokine release of cultured 16HBE 14o-bronchial ECs in vitro. Bronchial epithelial cells (ECs) from all subjects were obtained by bronchoscopic brushing. The expression levels of KGF and its receptor KGFR in collected cells were determined using RT-qPCR and Western blotting. The apoptosis and adhesion molecules expression by KGF administration were determined using flow cytometry and Western blotting. This occurred when 16HBE 14o-cell lines cultured and were exposed to interferon-γ (IFN-γ) and tumor necrosis factor-alpha (TNF-α) in vitro. The role of KGF on proliferation and damage repair were analyzed using CCK-8, EdU and wound closure assays after 16HBE 14o-cells were scraped. The effect of KGF on the release of inflammation related cytokines by damaged ECs was measured using ELISA kits. Compared with healthy controls, the KGF and KGFR expression and apoptosis significantly increased in collected cells from asthma patients. In vitro, treatment of KGF may limit IFN-γ and TNF-α induced apoptosis by inhibiting apoptosis-associated markers in the TNF signaling pathway. Besides, KGF could limit the release of TSLP, IL-25 and IL-33 by damaged 16HBE 14o-cells. On the contrary, KGF could promote the intercellular adhesion and wound closure of cultured 16HBE 14o-cells via the increased expression level of intercellular junction proteins ICAM-1, β-catenin, E-cad, and Dsc3. In conclusion, KGF and KGFR may help bronchial ECs repair in asthma via the inhibition apoptosis of ECs while the promotion of proliferation and migration of ECs.
Collapse
Affiliation(s)
- Jiongbo Wang
- The Clinic of Retired Cadres, Qingdao Municipal Hospital (East Campus), Qingdao, 266071, China
| | - Hongju Sun
- Department of General Medicine, Qingdao Central Hospital, Qingdao, 266042, China
| | - Yunshun Liu
- The Clinic of Retired Cadres, Qingdao Municipal Hospital (East Campus), Qingdao, 266071, China.
| |
Collapse
|
6
|
Effects of Pelargonium sidoides and Coptis Rhizoma 2 : 1 Mixed Formula (PS + CR) on Ovalbumin-Induced Asthma in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:9135637. [PMID: 32190091 PMCID: PMC7066403 DOI: 10.1155/2020/9135637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 11/18/2022]
Abstract
Pelargonium sidoides (PS) is traditionally used to treat respiratory and gastrointestinal infections, dysmenorrhea, and hepatic disorders in South Africa. Coptis Rhizoma (CR) is used to treat gastroenteric disorders, cardiovascular diseases, and cancer in East Asia. In the present study, we intended to observe the possible beneficial antiasthma effects of PS and CR on the ovalbumin- (OVA-) induced asthma C57BL/6J mice. Asthma in mice was induced by OVA sensitization and subsequent boosting. PS + CR (300 and 1,000 mg/kg; PO) or dexamethasone (IP) was administered once a day for 16 days. The changes in the body weight and gains, lung weights and gross inspections, total and differential cell counts of leukocytes in bronchoalveolar lavage fluid (BALF), serum OVA-specific immunoglobulin E (OVA-sIgE) levels, interleukin-4 (IL-4) and IL-5 levels in BALF and lung tissue homogenate, and IL-4 and IL-5 mRNA levels in lung tissue homogenates were analyzed with lung histopathology: mean alveolar surface area (ASA), alveolar septal thickness, numbers of inflammatory cells, mast cells, and eosinophils infiltrated in the alveolar regions, respectively. Significant increases in lung weights, total and differential cell counts of leukocytes in BALF, serum OVA-sIgE levels, and IL-4 and IL-5 levels in BALF and lung tissue homogenate were observed in OVA control as compared to those of intact control. In addition, OVA control showed a significant decrease in mean ASA and increases in alveolar septal thickness, numbers of inflammatory cells, mast cells, and eosinophils infiltrated in alveolar regions. However, these allergic and inflammatory asthmatic changes were significantly inhibited by PS + CR in a dose-dependent manner. In this study, PS + CR showed dose-dependent beneficial effects on OVA-induced asthma in mice through anti-inflammatory and antiallergic activities. Therefore, it is expected that PS + CR have enough potential as a new therapeutic agent or as an ingredient of a medicinal agent for various allergic and inflammatory respiratory diseases including asthma.
Collapse
|
7
|
Ingawale DK, Mandlik SK, Patel SS. Combination of Sarsasapogenin and Fluticasone attenuates ovalbumin-induced airway inflammation in a mouse asthma model. Immunopharmacol Immunotoxicol 2020; 42:128-137. [PMID: 32070162 DOI: 10.1080/08923973.2020.1728541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Objective: Asthma is a very common airway inflammatory disease for which the existing drug therapy options are insufficient. In this study, we explored the mechanisms underlying the anti-inflammatory potential of Sarsapogenin (SG) and its combination with Fluticasone (FC) in ovalbumin (OVA)-induced allergic asthma in mice.Methods: In a standard experimental model, asthma in mice was sensitized and challenged by OVA. The mice were treated with SG and SG + FC during OVA challenge. At the completion, lung weight, inflammatory cell count in bronchoalveolar lavage fluid (BALF), serum cytokines levels, immunoglobulin E (IgE) levels, lung nitrate/nitrite (NO) levels, and lung tissue oxidative stress biomarkers were determined. Histopathological evaluation of the lung tissue was also performed.Key findings: Treatment of mice with SG and SG + FC combination intensely diminished the trafficking of total and differential inflammatory cells count into BALF. SG and SG + FC administration significantly reduced the production of inflammatory cytokines, serum IgE levels and restoration of antioxidant stress markers. Histopathological analysis of lung samples effectually weakened bronchial inflammation and mucus production in the lung with a significant reduction in inflammation and mucus score.Conclusion: Our study results suggested that SG and SG + FC effectively reduced allergic airway inflammation via inhibiting pro-inflammatory cytokines, NO expressions and oxidative stress parameters. So, it could be used as a therapeutic potential agent for the treatment of asthma by decreasing its dose in combination with FC to avoid the chronic adverse effects of FC.
Collapse
Affiliation(s)
- Deepa K Ingawale
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | | | - Snehal S Patel
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
8
|
Ha SG, Dileepan M, Ge XN, Kang BN, Greenberg YG, Rao A, Muralidhar G, Medina-Kauwe L, Thompson MA, Pabelick CM, O'Grady SM, Rao SP, Sriramarao P. Knob protein enhances epithelial barrier integrity and attenuates airway inflammation. J Allergy Clin Immunol 2018; 142:1808-1817.e3. [PMID: 29522849 PMCID: PMC6126992 DOI: 10.1016/j.jaci.2018.01.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 12/12/2017] [Accepted: 01/29/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Altered epithelial physical and functional barrier properties along with TH1/TH2 immune dysregulation are features of allergic asthma. Regulation of junction proteins to improve barrier function of airway epithelial cells has the potential for alleviation of allergic airway inflammation. OBJECTIVE We sought to determine the immunomodulatory effect of knob protein of the adenoviral capsid on allergic asthma and to investigate its mechanism of action on airway epithelial junction proteins and barrier function. METHODS Airway inflammation, including junction protein expression, was evaluated in allergen-challenged mice with and without treatment with knob. Human bronchial epithelial cells were exposed to knob, and its effects on expression of junction proteins and barrier integrity were determined. RESULTS Administration of knob to allergen-challenged mice suppressed airway inflammation (eosinophilia, airway hyperresponsiveness, and IL-5 levels) and prevented allergen-induced loss of airway epithelial occludin and E-cadherin expression. Additionally, knob decreased expression of TH2-promoting inflammatory mediators, specifically IL-33, by murine lung epithelial cells. At a cellular level, treatment of human bronchial epithelial cells with knob activated c-Jun N-terminal kinase, increased expression of occludin and E-cadherin, and enhanced epithelial barrier integrity. CONCLUSION Increased expression of junction proteins mediated by knob leading to enhanced epithelial barrier function might mitigate the allergen-induced airway inflammatory response, including asthma.
Collapse
Affiliation(s)
- Sung Gil Ha
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - Mythili Dileepan
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - Xiao Na Ge
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - Bit Na Kang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - Yana G Greenberg
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - Amrita Rao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | | | - Lali Medina-Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center and Geffen School of Medicine, University of California-Los Angeles, Los Angeles, Calif
| | | | - Christina M Pabelick
- Departments of Anesthesiology and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minn
| | - Scott M O'Grady
- Departments of Animal Science and Integrative Biology and Physiology, University of Minnesota, St Paul, Minn
| | - Savita P Rao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn
| | - P Sriramarao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St Paul, Minn; Department of Medicine, University of Minnesota, Minneapolis, Minn.
| |
Collapse
|
9
|
Abdеlaziz RR, Еlmahdy MK, Suddek GM. Flavocoxid attenuates airway inflammation in ovalbumin-induced mouse asthma model. Chem Biol Interact 2018; 292:15-23. [PMID: 29986831 DOI: 10.1016/j.cbi.2018.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/25/2018] [Accepted: 07/01/2018] [Indexed: 01/05/2023]
Abstract
Asthma is a common airways inflammatory disease. This study provides evidence on the efficacy of flavocoxid against ovalbumin (OVA)-induced allergic airways inflammation in a mouse model of asthma. Airway inflammation was induced by intrapеritonеal injection of 10 mg ovalbumin (OVA) on day zero and day 7 followed by OVA challenge starting from 14th day to 16th day. Beclomethasone; a standard anti-inflammatory agent was selected as a drug in asthma. Flavocoxid (20 mg/kg, i. p.) was administered on day zero till 16th day followed by OVA challenge. At the end of the study, lung weight index, bronchoalveolar lavage fluid (BALF) content of total and differential WBCs, interleukin-13(IL-13), in addition to lung tissue nitrate/nitrite (NO) and oxidative stress biomarkers were measured. Also, histological and immunohistochemical analysis were conducted. Daily i. p. injection of flavocoxid (20 mg/kg) significantly improved airway inflammation. Inflammatory cells in BALF, malondialdehyde (MDA), NO and IL-13 significantly declined with concomitant increase in superoxide dismutase (SOD) activity. Histopathological examination and immunohistochеmical staining of mast cells were correlated with observed biochemical improvements. Collectively, these results demonstrate that flavocoxid mitigates the allergic airway inflammation induced by ovalbumin through attenuation of IL-13, NO expressions and oxidative stress.
Collapse
Affiliation(s)
- Rania R Abdеlaziz
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Mohammеd Kh Еlmahdy
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Ghada M Suddek
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| |
Collapse
|
10
|
Looi K, Buckley AG, Rigby PJ, Garratt LW, Iosifidis T, Zosky GR, Larcombe AN, Lannigan FJ, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Knight DA, Kicic A, Stick SM. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin Exp Allergy 2018; 48:513-524. [PMID: 29350877 DOI: 10.1111/cea.13097] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.
Collapse
Affiliation(s)
- K Looi
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - A G Buckley
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - P J Rigby
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - L W Garratt
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - T Iosifidis
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia
| | - G R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hohart, TAS, Australia
| | - A N Larcombe
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - F J Lannigan
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, Notre Dame University, Fremantle, WA, Australia
| | - K-M Ling
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - K M Martinovich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E Kicic-Starcevich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - N C Shaw
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E N Sutanto
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - D A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - A Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - S M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
11
|
Buckley AG, Looi K, Iosifidis T, Ling KM, Sutanto EN, Martinovich KM, Kicic-Starcevich E, Garratt LW, Shaw NC, Lannigan FJ, Larcombe AN, Zosky G, Knight DA, Rigby PJ, Kicic A, Stick SM. Visualisation of Multiple Tight Junctional Complexes in Human Airway Epithelial Cells. Biol Proced Online 2018; 20:3. [PMID: 29434527 PMCID: PMC5793437 DOI: 10.1186/s12575-018-0070-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/22/2018] [Indexed: 01/30/2023] Open
Abstract
Background Apically located tight junctions in airway epithelium perform a fundamental role in controlling macromolecule migration through paracellular spaces. Alterations in their expression may lead to disruptions in barrier integrity, which subsequently facilitates entry of potential bacterial and other pathogens into the host. Furthermore, there is emerging evidence that the barrier integrity of the airway in certain airway inflammatory diseases may be altered. However, there is little consensus on the way this is assessed and measured and the type of cells used to achieve this. Methods Here, we assessed four fixation methods including; (i) 4% (v/v) paraformaldehyde; (ii) 100% methanol; (iii) acetone or; (iv) 1:1 methanol: acetone. Pre-extraction with Triton X-100 was also performed and assessed on cells prior to fixation with either methanol or paraformaldehyde. Cells were also permeabilized with 0.1% (v/v) Saponin in 1× TBS following fixation and subsequently stained for tight junction proteins. Confocal microscopy was then used to visualise, compare and evaluate staining intensity of the tight junctional complexes in order to determine a standardised workflow of reproducible staining. Results Positive staining was observed following methanol fixation for claudin-1 and ZO-1 tight junction proteins but no staining was detected for occludin in 16HBE14o- cells. Combinatorial fixation with methanol and acetone also produced consistent positive staining for both occludin and ZO-1 tight junction proteins in these cells. When assessed using primary cells cultured at air-liquid interface, similar positive staining for claudin-1 and ZO-1 was observed following methanol fixation, while similar positive staining for occludin and ZO-1 was observed following the same combinatorial fixation with methanol and acetone. Conclusions The present study demonstrates the importance of a personalised approach to optimise staining for the visualisation of different tight junction proteins. Of significance, the workflow, once optimised, can readily be translated into primary airway epithelial cell air-liquid interface cultures where it can be used to assess barrier integrity in chronic lung diseases.
Collapse
Affiliation(s)
- Alysia G Buckley
- 1Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Kevin Looi
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Thomas Iosifidis
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Kak-Ming Ling
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Erika N Sutanto
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia
| | - Kelly M Martinovich
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Elizabeth Kicic-Starcevich
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Luke W Garratt
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia
| | - Nicole C Shaw
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Francis J Lannigan
- School of Medicine, Notre Dame University, Fremantle, Western Australia 6160 Australia
| | - Alexander N Larcombe
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Graeme Zosky
- 4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,7School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania 7000 Australia
| | - Darryl A Knight
- 8School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales Australia.,9Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales Australia.,10Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Paul J Rigby
- 1Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia 6009 Australia
| | - Anthony Kicic
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia.,11School of Public Health, Curtin University, Bentley, Western Australia 6102 Australia.,12Telethon Kids Institute, Subiaco, Perth, Western Australia 6008 Australia
| | - Stephen M Stick
- 2School of Paediatrics and Child Health, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,3Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia 6009 Australia.,4Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia 6009 Australia.,5Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia 6001 Australia
| |
Collapse
|
12
|
Biphasic lung injury during Streptococcus pneumoniae infection in a murine model. Med Mal Infect 2017; 48:103-113. [PMID: 29191391 DOI: 10.1016/j.medmal.2017.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 05/03/2017] [Accepted: 11/07/2017] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Streptococcus pneumoniae is the leading cause of community-acquired pneumonia. We aimed to analyze the epithelial response to S. pneumoniae-induced lung injury. METHODS Using an in vitro model with 16HBE cells and experimental in vivo murine model of acute lung injury, we analyzed the epithelial response to S. pneumoniae. Lung epithelial cell monolayers were exposed to S. pneumoniae and permeability was assessed by transepithelial resistance (TER) measurement and organization and expression of junction proteins. Functional consequences were studied with an in vivo murine model measuring alveolar permeability, distal alveolar fluid clearance (DAFC), and the alveolar inflammatory response. RESULTS In vitro, S. pneumoniae induced a dose-dependent decrease in transepithelial resistance, which was associated with significant modifications in the organization of junction proteins assessed by immunofluorescence staining and expression after 6hours of exposure. In vivo, S. pneumoniae induced a transient increase in alveolar permeability with an adequate increase in DAFC 6hours post infection. In a second phase, a permanent increased permeability was associated with a major decrease in DAFC. CONCLUSION Overall, the epithelial response to S. pneumoniae followed a biphasic pattern with an initial reversible increase in permeability related to the alteration of tight and adherens junctions and a second phase associated with an epithelial injury with a major increase in permeability with a decreased DAFC reflecting an injured alveolar capillary barrier.
Collapse
|
13
|
London NR, Lane AP. Innate immunity and chronic rhinosinusitis: What we have learned from animal models. Laryngoscope Investig Otolaryngol 2016; 1:49-56. [PMID: 28459101 PMCID: PMC5409101 DOI: 10.1002/lio2.21] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Chronic rhinosinusitis (CRS) is a heterogeneous and multifactorial disease characterized by dysregulated inflammation. Abnormalities in innate immune function including sinonasal epithelial cell barrier function, mucociliary clearance, response to pathogen-associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs), and the contribution of innate immune cells will be highlighted in this review. DATA SOURCES PubMed literature review. REVIEW METHODS A review of the literature was conducted to determine what we have learned from animal models in relation to innate immunity and chronic rhinosinusitis. RESULTS Dysregulation of innate immune mechanisms including sinonasal barrier function, mucociliary clearance, PAMPs, and innate immune cells such as eosinophils, mast cells, and innate lymphoid cells may contribute to CRS pathogenesis. Sinonasal inflammation has been studied using mouse, rat, rabbit, pig, and sheep explant or in vivo models. Study using these models has allowed for analysis of experimental therapeutics and furthered our understanding of the aforementioned aspects of the innate immune mechanism as it relates to sinonasal inflammation. These include augmenting mucociliary clearance through activation of the cystic fibrosis transmembrane conductance regulator (CFTR) and study of drug toxicity on ciliary beat frequency. Knockout models of Toll-like receptors (TLR) have demonstrated the critical role these PRRs play in allergic inflammation as loss of TLR2 and TLR4 leads to decreased lower airway inflammation. Mast cell deficient mice are less susceptible to ovalbumin-induced sinonasal inflammation. CONCLUSION Animal models have shed light as to the potential contribution of dysregulated innate immunity in chronic sinonasal inflammation.
Collapse
Affiliation(s)
- Nyall R. London
- Department of OtolaryngologyHead and Neck Surgery, Johns Hopkins School of MedicineBaltimoreMarylandU.S.A.
| | - Andrew P. Lane
- Department of OtolaryngologyHead and Neck Surgery, Johns Hopkins School of MedicineBaltimoreMarylandU.S.A.
| |
Collapse
|
14
|
Abstract
The bronchial epithelium is constantly exposed to a wide range of environmental materials present in inhaled air, including noxious gases and anthropogenic and natural particulates, such as gas and particles from car emissions, tobacco smoke, pollens, animal dander, and pathogens. As a fully differentiated, pseudostratified mucociliary epithelium, the bronchial epithelium protects the internal milieu of the lung from these agents by forming a physical barrier involving adhesive complexes and a chemical barrier involving secretion of mucus, which traps inhaled particles that can be cleared by the mucociliary escalator. It is a testament to the effectiveness of these two barriers that most environmental challenges are largely overcome without the need to develop an inflammatory response. However, as the initial cell of contact with the environment, the bronchial epithelium also plays a pivotal role in immune surveillance and appropriate activation of immune effector cells and antigen presenting cells in the presence of pathogens or other danger signals. Thus, the bronchial epithelium plays a central role in controlling tissue homeostasis and innate immunity. This review will discuss these barrier properties and how dysregulation of these homeostatic mechanisms can contribute to disease pathologies such as asthma.
Collapse
|
15
|
Finch PW, Mark Cross LJ, McAuley DF, Farrell CL. Palifermin for the protection and regeneration of epithelial tissues following injury: new findings in basic research and pre-clinical models. J Cell Mol Med 2014; 17:1065-87. [PMID: 24151975 PMCID: PMC4118166 DOI: 10.1111/jcmm.12091] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 05/06/2013] [Accepted: 05/15/2013] [Indexed: 02/06/2023] Open
Abstract
Keratinocyte growth factor (KGF) is a paracrine-acting epithelial mitogen produced by cells of mesenchymal origin, that plays an important role in protecting and repairing epithelial tissues. Pre-clinical data initially demonstrated that a recombinant truncated KGF (palifermin) could reduce gastrointestinal injury and mortality resulting from a variety of toxic exposures. Furthermore, the use of palifermin in patients with hematological malignancies reduced the incidence and duration of severe oral mucositis experienced after intensive chemoradiotherapy. Based upon these findings, as well as the observation that KGF receptors are expressed in many, if not all, epithelial tissues, pre-clinical studies have been conducted to determine the efficacy of palifermin in protecting different epithelial tissues from toxic injury in an attempt to model various clinical situations in which it might prove to be of benefit in limiting tissue damage. In this article, we review these studies to provide the pre-clinical background for clinical trials that are described in the accompanying article and the rationale for additional clinical applications of palifermin.
Collapse
|
16
|
Park SW, Lee EH, Lee EJ, Kim HJ, Bae DJ, Han S, Kim D, Jang AS, Uh ST, Kim YH, Erle DJ, Park CS. Apolipoprotein A1 potentiates lipoxin A4 synthesis and recovery of allergen-induced disrupted tight junctions in the airway epithelium. Clin Exp Allergy 2014; 43:914-27. [PMID: 23889245 DOI: 10.1111/cea.12143] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 04/21/2013] [Accepted: 04/29/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND Asthma is characterized by chronic airway inflammation triggered by various allergens in the environment. Defects in the bronchial epithelial interface with the external environment are the hallmark of asthma. Apolipoprotein A-1 (ApoA1) or ApoA1 mimetics have demonstrated anti-inflammatory activity and preventive effects in mouse models. OBJECTIVE We investigated airway levels of ApoA1 in asthmatics and the possible role of ApoA1 in protection of the bronchial epithelium and in resolution of inflammation in cellular and animal models of asthma. METHODS ApoA1 levels were measured in bronchoalveolar lavage fluid (BALF) from asthmatics and healthy controls. With treatment of ApoA1, mouse model of house dust mite (HDM)-driven asthma and cultured primary bronchial epithelial cells obtained from asthmatics were examined. Tight junction (TJ) expression in the bronchial epithelial cells was assessed by using confocal microscopy and immunoblot. RESULTS Asthmatics showed significantly lower ApoA1 levels in bronchoalveolar lavage fluid than did healthy controls. Local ApoA1 treatment significantly decreased lung IL-25, IL-33, and thymic stromal lymphopoietin levels in HDM-challenged mice and inhibited allergen-induced production of these cytokines in cultured primary bronchial epithelial cells. ApoA1 promoted recovery of disrupted TJ proteins zonula occludens-1 and occludin in cultured primary bronchial epithelium obtained from asthmatics. ApoA1-induced increases in the TJ proteins were dependent on increased production of lipoxin A4 (LX A4). CONCLUSIONS AND CLINICAL RELEVANCE ApoA1 enhances resolution of allergen-induced airway inflammation through promoting recovery of damaged TJs in the bronchial epithelium. ApoA1 could be a therapeutic strategy in chronic airway inflammatory diseases that are associated with a defective epithelial barrier, including asthma.
Collapse
Affiliation(s)
- S-W Park
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-Do, South Korea.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Choi YS, Kim YC, Ji S, Choi Y. Increased bacterial invasion and differential expression of tight-junction proteins, growth factors, and growth factor receptors in periodontal lesions. J Periodontol 2014; 85:e313-22. [PMID: 24527855 DOI: 10.1902/jop.2014.130740] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Many pathogens are known to modulate epithelial physical barriers, particularly tight-junction (TJ) proteins, to enter host cells and/or tissues. Growth factors have been implicated in the regulation of TJ proteins. The aim of this study is to determine differences in the levels of TJ proteins, growth factors, and their receptors in relation to bacterial invasion in diseased gingival tissues obtained from patients with periodontitis. METHODS The presence of bacteria and expression of junctional adhesion molecule (JAM)-A, occludin, epidermal growth factor (EGF), keratinocyte growth factor (KGF), insulin-like growth factor-I (IGF-I), EGF receptor, KGF receptor, and IGF-1 receptor (IGF-1R) were evaluated in gingival tissues from healthy (n = 10) and diseased (n = 10) sites in patients with periodontitis by in situ hybridization and immunohistochemistry. RESULTS The bacterial invasion of gingival tissue was increased in periodontal lesions compared with healthy sites. Although the levels of JAM-A and occludin were not significantly different between the healthy and diseased sites, aberrant cytoplasmic expression of JAM-A and occluding was often observed in the lesions. In addition, more leukocytes expressing JAM-A or occludin were observed within the disease-associated epithelia. Compared with the healthy sites, the differential expression of KGF, IGF-I, and IGF-1R was observed in the periodontal lesions. The levels of TJ proteins showed positive correlations with those of growth factors. CONCLUSION The aberrant expression of growth factors and TJ proteins may contribute to increased bacterial invasion and disease progression in periodontal lesions.
Collapse
Affiliation(s)
- Yun S Choi
- Department of Immunology and Molecular Microbiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | | | | | | |
Collapse
|
18
|
β-Catenin promotes host resistance against Pseudomonas aeruginosa keratitis. J Infect 2013; 67:584-94. [PMID: 23911965 DOI: 10.1016/j.jinf.2013.07.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 06/19/2013] [Accepted: 07/23/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To explore the role of β-catenin in Pseudomonas aeruginosa (PA) keratitis. METHODS Western-blot and immunostaining assay were used to determine the β-catenin protein expression in C57BL/6 (B6) corneas and in in vitro cultured murine cells including macrophage-like RAW264.7 cells, bone marrow-derived neutrophils and A6(1) corneal epithelial cells. B6 mice were subconjunctivally injected with lentivirus expressing active mutant of β-catenin (β-cat-lentivirus) vs appropriate control (Ctl-lentivirus), and then infected with PA. Pro-inflammatory cytokine levels were examined using real-time PCR and ELISA, and bacterial burden was assessed using plate count assays both in vivo and in vitro. RESULTS β-Catenin protein expression was decreased in B6 corneas, murine macrophage-like RAW264.7 cells, mouse bone marrow-derived neutrophils and mouse A6(1) corneal epithelial cells after PA infection. Over-expression of β-catenin in B6 corneas significantly reduced the severity of corneal disease after PA infection, by decreasing pro-inflammatory cytokine expression and bacterial burden. In vitro data further demonstrated that over-expression of β-catenin suppressed pro-inflammatory cytokine production but enhanced bacterial clearance in macrophages and neutrophils. CONCLUSIONS β-Catenin reduces the severity of PA keratitis by decreasing corneal inflammation and bacterial burden.
Collapse
|
19
|
Hallstrand TS, Kippelen P, Larsson J, Bougault V, van Leeuwen JC, Driessen JMM, Brannan JD. Where to from here for exercise-induced bronchoconstriction: the unanswered questions. Immunol Allergy Clin North Am 2013; 33:423-42, ix. [PMID: 23830134 DOI: 10.1016/j.iac.2013.02.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The role of epithelial injury is an unanswered question in those with established asthma and in elite athletes who develop features of asthma and exercise-induced bronchorestriction (EIB) after years of training. The movement of water in response to changes in osmolarity is likely to be an important signal to the epithelium that may be central to the onset of EIB. It is generally accepted that the mast cell and its mediators play a major role in EIB and the presence of eosinophils is likely to enhance EIB severity.
Collapse
Affiliation(s)
- Teal S Hallstrand
- Division of Pulmonary and Critical Care, University of Washington, Department of Medicine, 1959 NE Pacific Street, Box 356166, Seattle, WA 98195-6522, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Conway J, Fleming J, Bennett M, Havelock T. The co-imaging of gamma camera measurements of aerosol deposition and respiratory anatomy. J Aerosol Med Pulm Drug Deliv 2013; 26:123-30. [PMID: 23517170 DOI: 10.1089/jamp.2011.0960] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The use of gamma camera imaging following the inhalation of a radiolabel has been widely used by researchers to investigate the fate of inhaled aerosols. The application of two-dimensional (2D) planar gamma scintigraphy and single-photon emission computed tomography (SPECT) to the study of inhaled aerosols is discussed in this review. Information on co-localized anatomy can be derived from other imaging techniques such as krypton ventilation scans and low- and high-resolution X-ray computed tomography (CT). Radionuclide imaging, combined with information on anatomy, is a potentially useful approach when the understanding of regional deposition within the lung is central to research objectives for following disease progression and for the evaluation of therapeutic intervention.
Collapse
Affiliation(s)
- Joy Conway
- Faculty of Health Sciences, University of Southampton, Southampton, UK SO16 6YD.
| | | | | | | |
Collapse
|
21
|
Ramakrishna L, de Vries VC, Curotto de Lafaille MA. Cross-roads in the lung: immune cells and tissue interactions as determinants of allergic asthma. Immunol Res 2012; 53:213-28. [PMID: 22447350 DOI: 10.1007/s12026-012-8296-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Allergic asthma is a chronic disease of the lung characterized by underlying Th2- and IgE-mediated inflammation, structural alterations of the bronchial wall, and airway hyperresponsiveness. Initial allergic sensitization and later development of chronic disease are determined by close interactions between lung structural cells and the resident and migratory immune cells in the lung. Epithelial cells play a crucial role in allergic sensitization by directly influencing dendritic cells induction of tolerant or effector T cells and production of type 2 cytokines by innate immune cells. During chronic disease, the bronchial epithelium, stroma, and smooth muscle become structurally and functionally altered, contributing to the perpetuation of tissue remodeling. Thus, targeting tissue-driven pathology in addition to inflammation may increase the effectiveness of asthma treatment.
Collapse
Affiliation(s)
- Lakshmi Ramakrishna
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, #4-06 Immunos, Singapore
| | | | | |
Collapse
|
22
|
Abdel Aziz RR, Helaly NY, Zalata KR, Gameil NM. Influence of inhaled beclomethasone and montelukast on airway remodeling in mice. Inflammopharmacology 2012; 21:55-66. [PMID: 22354687 DOI: 10.1007/s10787-012-0127-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 02/07/2012] [Indexed: 01/06/2023]
Abstract
This study examined the effect of montelukast and beclomethasone on airway remodeling in murine model of asthma. Mice were sensitized by i.p. injection of ovalbumin (OVA) on days 0 and 14, and then challenged by nebulization of 1% OVA 3 days/week for 6 or 10 weeks. Results of 6-week OVA-challenged group showed moderate inflammation, but the 10-week OVA-challenged group exhibited mild inflammation. The OVA challenge (6 and 10 weeks) exhibited marked airway fibrosis, illustrated by significant increase in goblet cell hyperplasia and epithelial thickness, increased lung content of collagen and transforming growth factor-β(1), together with a decrease in nitric oxide production; also, there was an increase in bronchoalveolar lavage fluid level of interleukin-13. Administration of montelukast or beclomethasone before each OVA challenge was capable of restoring most of the measured parameters to near normal levels. Inhalation of beclomethasone has a similar role in airway remodeling as montelukast, but its effects in regulating inflammatory changes is less pronounced than montelukast.
Collapse
Affiliation(s)
- Rania R Abdel Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | | | | | | |
Collapse
|
23
|
Sekiyama A, Gon Y, Terakado M, Takeshita I, Kozu Y, Maruoka S, Matsumoto K, Hashimoto S. Glucocorticoids enhance airway epithelial barrier integrity. Int Immunopharmacol 2011; 12:350-7. [PMID: 22210372 DOI: 10.1016/j.intimp.2011.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 11/25/2011] [Accepted: 12/06/2011] [Indexed: 12/22/2022]
Abstract
Asthma is a chronic inflammatory disorder of the airways, but its pathogenesis is incompletely understood. While asthma is a complex disease caused by multiple factors, epithelial barrier damage is a cardinal feature. Glucocorticoids (GCs) are the most effective anti-inflammatory drugs in the treatment of asthma. However, the effects of GCs on the airway epithelial barrier have not been evaluated. Epithelial barrier functions were evaluated in cultured human airway epithelial cell monolayers, Calu-3 and 16HBE. Then, the cells were treated with dexamethasone (Dex), fulticasone propionate (FP), or budesonide (BD) for 5 days. Permeability measured by transepithelial electrical resistance was increased by treatment with Dex, FP, and BD in a dose-dependent manner. Permeability to fluorescein isothiocyanate-labeled dextran was markedly reduced by these treatments. Immunocytostaining revealed that Dex treatment potentiated tight junction formation in these polarized epithelial cells. Knockdown of epidermal growth factor receptor (EGFR) by small interference RNA blunted the effects of Dex on barrier integrity. Although EGFR expression was not affected by Dex treatment, EGFR phosphorylation was enhanced in Dex-treated cells. This is suggesting that EGFR are important for this phenomenon. These findings suggest that GC inhalation therapy can improve epithelial barrier integrity and might contribute to the therapeutic effects of GCs for treating asthma.
Collapse
Affiliation(s)
- Akiko Sekiyama
- Division of Respiratory Medicine, Department of Internal Medicine, Nihon University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
BACKGROUND Asthma is a disorder of the conducting airways that contract too easily and too much to cause variable airflow obstruction with symptoms of wheeze, cough, chest tightness and shortness of breath. Based on this knowledge, initial treatments were directed to dilating the contracted airways with anticholinergic and adrenergic drugs. The recognition that allergic-type inflammation underlay the hyperresponsive airways in asthma led to the introduction of anti-inflammatory drugs such as sodium cromoglicate and corticosteroids. Over the 2 decades that followed, these drugs have been progressively improved by increasing their therapeutic index and duration of action. METHODS A review of the recent literature indicates that since the 1980s, the explosive increase in knowledge of the cell and mediator mechanisms of asthma has only led to modest improvements in therapy including the introduction of leukotriene modifiers and a blocking monoclonal antibody against IgE. Indeed, biologics targeting allergic cytokines and effector cells have on the whole proven disappointing despite initial promise being shown in animal models. RESULTS Part of the difficulty lies in the oversimplified concept that asthma is only driven by allergic processes when in reality there are many environmental causes and triggers and the view that it is a homogeneous disorder only varying in severity. CONCLUSIONS The more recent views that asthma is a complex disorder made up of different subtypes with differing causes, treatment responses and natural histories creates a new opportunity for stratified medicine in which therapies acting upstream selectively target specific disease subtypes identified by specific diagnostic biomarkers.
Collapse
|
25
|
Xiao C, Puddicombe SM, Field S, Haywood J, Broughton-Head V, Puxeddu I, Haitchi HM, Vernon-Wilson E, Sammut D, Bedke N, Cremin C, Sones J, Djukanović R, Howarth PH, Collins JE, Holgate ST, Monk P, Davies DE. Defective epithelial barrier function in asthma. J Allergy Clin Immunol 2011; 128:549-56.e1-12. [PMID: 21752437 DOI: 10.1016/j.jaci.2011.05.038] [Citation(s) in RCA: 449] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/23/2011] [Accepted: 05/27/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND Asthma is a complex disease involving gene and environment interactions. Although atopy is a strong predisposing risk factor for asthma, local tissue susceptibilities are required for disease expression. The bronchial epithelium forms the interface with the external environment and is pivotally involved in controlling tissue homeostasis through provision of a physical barrier controlled by tight junction (TJ) complexes. OBJECTIVES To explain the link between environment exposures and airway vulnerability, we hypothesized that epithelial TJs are abnormal in asthma, leading to increased susceptibility to environmental agents. METHODS Localization of TJs in bronchial biopsies and differentiated epithelial cultures was assessed by electron microscopy or immunostaining. Baseline permeability and the effect of cigarette smoke and growth factor were assessed by measurement of transepithelial electrical resistance and passage of fluorescently labeled dextrans. RESULTS By using immunostaining, we found that bronchial biopsies from asthmatic subjects displayed patchy disruption of TJs. In differentiated bronchial epithelial cultures, TJ formation and transepithelial electrical resistance were significantly lower (P < .05) in cultures from asthmatic donors (n = 43) than from normal controls (n = 40) and inversely correlated with macromolecular permeability. Cultures from asthmatic donors were also more sensitive to disruption by cigarette smoke extract. Epidermal growth factor enhanced basal TJ formation in cultures from asthmatic subjects (P < .01) and protected against cigarette smoke-induced barrier disruption (P < .01). CONCLUSIONS Our results show that the bronchial epithelial barrier in asthma is compromised. This defect may facilitate the passage of allergens and other agents into the airway tissue, leading to immune activation and may thus contribute to the end organ expression of asthma.
Collapse
Affiliation(s)
- Chang Xiao
- Synairgen Research Ltd, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hirota JA, Hackett TL, Inman MD, Knight DA. Modeling asthma in mice: what have we learned about the airway epithelium? Am J Respir Cell Mol Biol 2010; 44:431-8. [PMID: 20724554 DOI: 10.1165/rcmb.2010-0146tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Clinical reports of areas of damaged airway epithelium associated with shed epithelial cells in bronchoalveolar lavage fluid, aberrant epithelial repair processes, and altered cytokine and growth factor release have highlighted some fundamental differences between the airway epithelium in individuals with and without asthma. However, the consequences of these epithelial changes are not clearly defined, and may be difficult to assess in the clinic. In this Review, we answer the two questions. (1) What in vivo models and methods have been used to inform us about airway epithelium damage, repair, and immune responses? Our response focuses on genetic influences as well as allergen exposure, environmental/chemical, and mechanical models. (2) How can we improve on existing mouse models to understand changes in airway epithelium biology in asthma? In answering the second question, we include exciting recent studies that have combined multiple exposure methods and/or epithelium-centric outcome measurements. By addressing these two questions, we propose that future interrogation of epithelial responses of both existing and nascent mouse models may provide greater understanding of the mechanisms underlying airway inflammation and remodeling in asthma with hope of generating novel therapeutic targets.
Collapse
Affiliation(s)
- Jeremy A Hirota
- University of British Columbia James Hogg Research Centre--Heart and Lung Institute, St. Paul's Hospital, Vancouver, British Columbia, Canada.
| | | | | | | |
Collapse
|
27
|
Swindle EJ, Collins JE, Davies DE. Breakdown in epithelial barrier function in patients with asthma: identification of novel therapeutic approaches. J Allergy Clin Immunol 2009; 124:23-34; quiz 35-6. [PMID: 19560576 DOI: 10.1016/j.jaci.2009.05.037] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 05/26/2009] [Accepted: 05/27/2009] [Indexed: 01/19/2023]
Abstract
The bronchial epithelium is pivotally involved in the provision of chemical, physical, and immunologic barriers to the inhaled environment. These barriers serve to maintain normal homeostasis, but when compromised, the immunologic barrier becomes activated to protect the internal milieu of the lung. We discuss what is currently understood about abnormalities in these barrier functions in patients with asthma and consider novel therapeutic opportunities that target this key structure.
Collapse
Affiliation(s)
- Emily J Swindle
- Division of Infection, Inflammation and Immunity, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | | | | |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW The use of inhaled corticosteroids, short and long acting beta2-adrenoceptor agonists and inhibitors of leukotrienes provide most asthmatic patients with good disease control. However, none of these therapies are specifically directed to the underlying causal pathways of asthma. In this review the role of selective inhibitors of the inflammatory cascade are presented with a particular emphasis on biologics. RECENT FINDINGS Apart from antihuman immunoglobulin E, biologics have had little impact on this disease. However, with the definition of critical pathways in driving ongoing inflammation and airway remodelling, the situation is about to change with several exciting new approaches being on the horizon. Specific cytokines that are considered central to the Th2 inflammatory response as therapeutic targets are discussed along with some entirely new approaches such as restoration of mucosal innate immunity and epithelial barrier function and the application of radiofrequency ablation of airway smooth muscle or thermal bronchoplasty. SUMMARY What is becoming clear in filling the pipeline with new asthma therapies that treat the underlying disease causes is the need for closer working between clinical academics and industry to ensure that there is a rapid and sustained transfer of knowledge on novel targets through to their validation, proof of concept studies and clinical trials.
Collapse
|
29
|
Van Hove CL, Maes T, Joos GF, Tournoy KG. Chronic inflammation in asthma: a contest of persistence vs resolution. Allergy 2008; 63:1095-109. [PMID: 18616676 DOI: 10.1111/j.1398-9995.2008.01772.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent investigations have highlighted that endogenous anti-inflammatory mediators and immune regulating mechanisms are important for the resolution of inflammatory processes. A disruption of these mechanisms can be causally related not only to the initiation of unnecessary inflammation, but also to the persistence of several chronic inflammatory diseases. In asthma, chronic Th-2 driven eosinophilic inflammation of the airways is one of the central abnormalities. To date, elucidating the role of the different pro-inflammatory mediators involved in orchestrating the inflammatory processes in asthma has been the subject of intense research in both humans and animal models. However, the counter-regulatory mechanisms that co-determine the outcome in the contest of resolution vs persistence of the eosinophilic airway inflammation remain poorly understood. These are currently being investigated in animal models of chronic asthma. Elucidating these mechanisms is of relevance, since it can give rise to a new therapeutic approach in the treatment of chronic airway inflammation in asthmatics. This novel concept of treatment involves the stimulation of endogenous anti-inflammatory pathways, rather than solely antagonising the various pro-inflammatory mediators. Here, we review and discuss the current knowledge about these endogenous anti-inflammatory mediators in clinical and experimental asthma.
Collapse
Affiliation(s)
- C L Van Hove
- Department of Respiratory Medicine, Laboratory of Translational Research in Obstructive Pulmonary Diseases, Ghent University, Ghent, Belgium
| | | | | | | |
Collapse
|
30
|
Yildirim AO, Veith M, Rausch T, Müller B, Kilb P, Van Winkle LS, Fehrenbach H. Keratinocyte growth factor protects against Clara cell injury induced by naphthalene. Eur Respir J 2008; 32:694-704. [PMID: 18385170 DOI: 10.1183/09031936.00155107] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Airway epithelial cells are exposed to environmental toxicants that result in airway injury. Naphthalene (NA) causes site-selective damage to Clara cells in mouse distal airways. N-terminally truncated recombinant human keratinocyte growth factor (DeltaN23-KGF) protects against acute lung injury. The present study investigated whether or not DeltaN23-KGF protects against NA-induced acute Clara cell damage by measuring airway responses specifically and in order to identify underlying molecular mechanisms. Mice were treated with DeltaN23-KGF or PBS 33 h prior to injection of 200 mg.kg body weight(-1) NA. Lung function was analysed by head-out body plethysmography. Distal airways isolated by microdissection were assessed for cell permeability using ethidium homodimer-1. Immunohistochemistry of Clara cell-specific protein in conjunction with a physical dissector was used to quantify Clara cell numbers. RNA was isolated from frozen airways in order to analyse gene expression using quantitative RT-PCR. DeltaN23-KGF prevented NA-induced airflow limitation and Clara cell permeability, and resulted in twice as many Clara cells compared with PBS pre-treatment. DeltaN23-KGF-pre-treated mice exhibited increased expression of proliferating cell nuclear antigen mRNA. Cytochrome P(450) isoform 2F2, which converts NA into its toxic metabolite, was reduced by approximately 50%. The present results demonstrate that pre-treatment with N-terminally truncated recombinant human keratinocyte growth factor protects against naphthalene-induced injury. This suggests that N-terminally truncated recombinant human keratinocyte growth factor exerts its beneficial effect through a decrease in the expression of cytochrome P(450) isoform 2F2.
Collapse
Affiliation(s)
- A O Yildirim
- Clinical Research Group Chronic Airway Diseases, Medical Faculty, Philipps University Marburg, Hans-Meerwein Str.1, 35043 Marburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Allergic diseases have reached epidemic proportions worldwide. An understanding of the cellular and soluble mediators that are involved in allergic inflammatory responses not only helps in understanding the mechanisms of current treatments, but is also important for the identification of new targets that are amenable to both small-molecule and biological interventions. There is now considerable optimism with regards to tackling the allergy epidemic in light of improvements in systemic and mucosal allergen-specific immunotherapy, the identification of key cytokines and their receptors that drive T-helper-2-cell polarization, a clearer understanding of the pathways of leukocyte recruitment and the signalling pathways that are involved in cell activation and mediator secretion, and new approaches to vaccine development.
Collapse
|
32
|
Lorenowicz MJ, Fernandez-Borja M, van Stalborch AMD, van Sterkenburg MAJA, Hiemstra PS, Hordijk PL. Microtubule dynamics and Rac-1 signaling independently regulate barrier function in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2007; 293:L1321-31. [PMID: 17827248 DOI: 10.1152/ajplung.00443.2006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cadherin-mediated cell-cell adhesion controls the morphology and function of epithelial cells and is a critical component of the pathology of chronic inflammatory disorders. Dynamic interactions between cadherins and the actin cytoskeleton are required for stable cell-cell contact. Besides actin, microtubules also target intercellular, cadherin-based junctions and contribute to their formation and stability. Here, we studied the role of microtubules in conjunction with Rho-like GTPases in the regulation of lung epithelial barrier function using real-time monitoring of transepithelial electrical resistance. Unexpectedly, we found that disruption of microtubules promotes epithelial cell-cell adhesion. This increase in epithelial barrier function is accompanied by the accumulation of beta-catenin at cell-cell junctions, as detected by immunofluorescence. Moreover, we found that the increase in cell-cell contact, induced by microtubule depolymerization, requires signaling through a RhoA/Rho kinase pathway. The Rac-1 GTPase counteracts this pathway, because inhibition of Rac-1 signaling rapidly promotes epithelial barrier function, in a microtubule- and RhoA-independent fashion. Together, our data suggest that microtubule-RhoA-mediated signaling and Rac-1 control lung epithelial integrity through counteracting independent pathways.
Collapse
Affiliation(s)
- Magdalena J Lorenowicz
- Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, Univ. of Amsterdam, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|