1
|
Zhang D, Li G, Liu X, Wang Y, Wu J, Ren Y, She G, Zheng D, Zhao Y, Deng XL, Li M, Zhao L. K Ca3.1 upregulation mediated by Ang II-induced JNK/AP-1 activation contributes to atrial fibrosis. Cell Signal 2025; 131:111731. [PMID: 40064281 DOI: 10.1016/j.cellsig.2025.111731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Atrial fibrillation is strongly associated with an increased risk of embolism, stroke, and heart failure. Current therapeutic approaches often have limited efficacy, and controlling atrial fibrosis remains a critical objective for upstream therapies. The specific mechanisms driving atrial fibrosis remain incompletely understood. The intermediate-conductance calcium-activated potassium channel KCa3.1 has been implicated in promoting fibroblast activation in various fibrotic diseases. This study investigates the role of angiotensin II (Ang II) in regulating KCa3.1, as well as its involvement in the pathogenesis of atrial fibrosis and the underlying signaling mechanisms. In a rat model, chronic Ang II infusion for 4 weeks induced atrial fibrosis, which was significantly attenuated by TRAM-34, a specific KCa3.1 channel blocker. In cultured rat atrial fibroblasts, Ang II treatment promoted fibroblast differentiation, proliferation, migration and collagen production, effects that were suppressed by TRAM-34 and KCa3.1 knockdown. Overexpression of KCa3.1 in fibroblasts further confirmed its pro-fibrotic role. Mechanistically, Ang II upregulated KCa3.1 expression and current density by activating the JNK/AP-1 signaling pathway. This involved phosphorylation of JNK, c-Jun, and c-Fos, leading to the formation of c-Jun/c-Fos heterodimers that directly bound to the KCa3.1 promoter to enhance its transcription. Together, these findings demonstrate that KCa3.1 mediates fibroblast activation and atrial fibrosis through the JNK/AP-1 pathway.
Collapse
Affiliation(s)
- Dongmei Zhang
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Guangyao Li
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Xiang Liu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Jie Wu
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yujie Ren
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Gang She
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Dong Zheng
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China
| | - Yinxia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, 366 North Longchuan Road, Shanghai 200031, China
| | - Xiu-Ling Deng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an 710061, Shaanxi, China
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China.
| | - Limei Zhao
- Department of Pathology and Pathophysiology, Suzhou Medical College of Soochow University, 199 Ren-ai Road, Suzhou 215123, Jiangsu, China; MOE Key Laboratory of Geriatric Diseases and Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
2
|
Li H, Kim JA, Jo SE, Lee H, Kim KC, Choi S, Suh SH. Modafinil exerts anti-inflammatory and anti-fibrotic effects by upregulating adenosine A 2A and A 2B receptors. Purinergic Signal 2024; 20:371-384. [PMID: 37938538 PMCID: PMC11303359 DOI: 10.1007/s11302-023-09973-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Adenosine receptor (AR) suppresses inflammation and fibrosis by activating cyclic adenosine monophosphate (cAMP) signaling. We investigated whether altered AR expression contributes to the development of fibrotic diseases and whether A2AAR and A2BAR upregulation inhibits fibrotic responses. Primary human lung fibroblasts (HLFs) from normal (NHLFs) or patients with idiopathic pulmonary fibrosis (DHLF) were used for in vitro testing. Murine models of fibrotic liver or pulmonary disease were developed by injecting thioacetamide intraperitoneally, by feeding a high-fat diet, or by intratracheal instillation of bleomycin. Modafinil, which activates cAMP signaling via A2AAR and A2BAR, was administered orally. The protein amounts of A2AAR, A2BAR, and exchange protein directly activated by cAMP (Epac) were reduced, while collagen and α-smooth muscle actin (α-SMA) were elevated in DHLFs compared to NHLFs. In liver or lung tissue from murine models of fibrotic diseases, A2AAR and A2BAR were downregulated, but A1AR and A3AR were not. Epac amounts decreased, and amounts of collagen, α-SMA, KCa2.3, and KCa3.1 increased compared to the control. Modafinil restored the amounts of A2AAR, A2BAR, and Epac, and reduced collagen, α-SMA, KCa2.3, and KCa3.1 in murine models of fibrotic diseases. Transforming growth factor-β reduced the amounts of A2AAR, A2BAR, and Epac, and elevated collagen, α-SMA, KCa2.3, and KCa3.1 in NHLFs; however, these alterations were inhibited by modafinil. Our investigation revealed that A2AAR and A2BAR downregulation induced liver and lung fibrotic diseases while upregulation attenuated fibrotic responses, suggesting that A2AAR and A2BAR-upregulating agents, such as modafinil, may serve as novel therapies for fibrotic diseases.
Collapse
Affiliation(s)
- Haiyan Li
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea
| | - Ji Aee Kim
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea
| | - Seong-Eun Jo
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea
| | - Huisu Lee
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea
| | - Kwan-Chang Kim
- Department of Thoracic & Cardiovascular Surgery, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea.
| | - Shinkyu Choi
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea.
| | - Suk Hyo Suh
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07084, Republic of Korea.
| |
Collapse
|
3
|
Ju X, Wang K, Wang C, Zeng C, Wang Y, Yu J. Regulation of myofibroblast dedifferentiation in pulmonary fibrosis. Respir Res 2024; 25:284. [PMID: 39026235 PMCID: PMC11264880 DOI: 10.1186/s12931-024-02898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/29/2024] [Indexed: 07/20/2024] Open
Abstract
Idiopathic pulmonary fibrosis is a lethal, progressive, and irreversible condition that has become a significant focus of medical research due to its increasing incidence. This rising trend presents substantial challenges for patients, healthcare providers, and researchers. Despite the escalating burden of pulmonary fibrosis, the available therapeutic options remain limited. Currently, the United States Food and Drug Administration has approved two drugs for the treatment of pulmonary fibrosis-nintedanib and pirfenidone. However, their therapeutic effectiveness is limited, and they cannot reverse the fibrosis process. Additionally, these drugs are associated with significant side effects. Myofibroblasts play a central role in the pathophysiology of pulmonary fibrosis, significantly contributing to its progression. Consequently, strategies aimed at inhibiting myofibroblast differentiation or promoting their dedifferentiation hold promise as effective treatments. This review examines the regulation of myofibroblast dedifferentiation, exploring various signaling pathways, regulatory targets, and potential pharmaceutical interventions that could provide new directions for therapeutic development.
Collapse
Affiliation(s)
- Xuetao Ju
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Kai Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Congjian Wang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Chenxi Zeng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China
| | - Yi Wang
- Department of Pulmonary and Critical Care Medicine, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China.
| | - Jun Yu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, People's Republic of China.
| |
Collapse
|
4
|
Vittal R, Walker NM, McLinden AP, Braeuer RR, Ke F, Fattahi F, Combs MP, Misumi K, Aoki Y, Wheeler DS, Wilke CA, Huang SK, Moore BB, Cao P, Lama VN. Genetic deficiency of the transcription factor NFAT1 confers protection against fibrogenic responses independent of immune influx. Am J Physiol Lung Cell Mol Physiol 2024; 326:L39-L51. [PMID: 37933452 PMCID: PMC11279780 DOI: 10.1152/ajplung.00045.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is marked by unremitting matrix deposition and architectural distortion. Multiple profibrotic pathways contribute to the persistent activation of mesenchymal cells (MCs) in fibrosis, highlighting the need to identify and target common signaling pathways. The transcription factor nuclear factor of activated T cells 1 (NFAT1) lies downstream of second messenger calcium signaling and has been recently shown to regulate key profibrotic mediator autotaxin (ATX) in lung MCs. Herein, we investigate the role of NFAT1 in regulating fibroproliferative responses during the development of lung fibrosis. Nfat1-/--deficient mice subjected to bleomycin injury demonstrated improved survival and protection from lung fibrosis and collagen deposition as compared with bleomycin-injured wild-type (WT) mice. Chimera mice, generated by reconstituting bone marrow cells from WT or Nfat1-/- mice into irradiated WT mice (WT→WT and Nfat1-/-→WT), demonstrated no difference in bleomycin-induced fibrosis, suggesting immune influx-independent fibroprotection in Nfat1-/- mice. Examination of lung tissue and flow sorted lineageneg/platelet-derived growth factor receptor alpha (PDGFRα)pos MCs demonstrated decreased MC numbers, proliferation [↓ cyclin D1 and 5-ethynyl-2'-deoxyuridine (EdU) incorporation], myofibroblast differentiation [↓ α-smooth muscle actin (α-SMA)], and survival (↓ Birc5) in Nfat1-/- mice. Nfat1 deficiency abrogated ATX expression in response to bleomycin in vivo and MCs derived from Nfat1-/- mice demonstrated decreased ATX expression and migration in vitro. Human IPF MCs demonstrated constitutive NFAT1 activation, and regulation of ATX in these cells by NFAT1 was confirmed using pharmacological and genetic inhibition. Our findings identify NFAT1 as a critical mediator of profibrotic processes, contributing to dysregulated lung remodeling and suggest its targeting in MCs as a potential therapeutic strategy in IPF.NEW & NOTEWORTHY Idiopathic pulmonary fibrosis (IPF) is a fatal disease with hallmarks of fibroblastic foci and exuberant matrix deposition, unknown etiology, and ineffective therapies. Several profibrotic/proinflammatory pathways are implicated in accelerating tissue remodeling toward a honeycombed end-stage disease. NFAT1 is a transcriptional factor activated in IPF tissues. Nfat1-deficient mice subjected to chronic injury are protected against fibrosis independent of immune influxes, with suppression of profibrotic mesenchymal phenotypes including proliferation, differentiation, resistance to apoptosis, and autotaxin-related migration.
Collapse
Affiliation(s)
- Ragini Vittal
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Natalie M Walker
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - A Patrick McLinden
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States
| | - Russell R Braeuer
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Fang Ke
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Fatemeh Fattahi
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Michael P Combs
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Keizo Misumi
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Yoshiro Aoki
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - David S Wheeler
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Carol A Wilke
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States
| | - Steven K Huang
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Bethany B Moore
- Department of Microbiology & Immunology, University of Michigan, Ann Arbor, Michigan, United States
| | - Pengxiu Cao
- Division of Pulmonary & Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Vibha N Lama
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
5
|
Ayilya BL, Balde A, Ramya M, Benjakul S, Kim SK, Nazeer RA. Insights on the mechanism of bleomycin to induce lung injury and associated in vivo models: A review. Int Immunopharmacol 2023; 121:110493. [PMID: 37331299 DOI: 10.1016/j.intimp.2023.110493] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/20/2023]
Abstract
Acute lung injury leads to the development of chronic conditions such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma as well as alveolar sarcoma. Various investigations are being performed worldwide to understand the pathophysiology of these diseases, develop novel bioactive compounds and inhibitors to target the ailment. Generally, in vivo models are used to understand the disease outcome and therapeutic suppressing effects for which the animals are chemically or physically induced to mimic the onset of definite disease conditions. Amongst the chemical inducing agents, Bleomycin (BLM) is the most successful inducer. It is reported to target various receptors and activate inflammatory pathways, cellular apoptosis, epithelial mesenchymal transition leading to the release of inflammatory cytokines, and proteases. Mice is one of the most widely used animal model for BLM induced pulmonary associated studies apart from rat, rabbit, sheep, pig, and monkey. Although, there is considerable variation amongst in vivo studies for BLM induction which suggests a detailed study on the same to understand the mechanism of action of BLM at molecular level. Hence, herein we have reviewed various chemical inducers, mechanism of action of BLM in inducing lung injury in vivo, its advantages and disadvantages. Further, we have also discussed the rationale behind various in vivo models and recent development in BLM induction for various animals.
Collapse
Affiliation(s)
- Bakthavatchalam Loganathan Ayilya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Murugadoss Ramya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Soottawat Benjakul
- Department of Food Technology, Faculty of Agro-Industry, Prince of Songkhla University, 90112 Hat Yai, Songkhla, Thailand
| | - Se-Kwon Kim
- Department of Marine Science and Convergence Engineering, Hanyang University, Ansan 11558, Gyeonggi-do, South Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
6
|
Petersen AG, Lind PC, Mogensen S, Jensen ASB, Granfeldt A, Simonsen U. Treatment with senicapoc, a K Ca 3.1 channel blocker, alleviates hypoxaemia in a mouse model of acute respiratory distress syndrome. Br J Pharmacol 2022; 179:2175-2192. [PMID: 34623632 DOI: 10.1111/bph.15704] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND AND PURPOSE Acute respiratory distress syndrome (ARDS) is characterized by pulmonary oedema and severe hypoxaemia. We investigated whether genetic deficit or blockade of calcium-activated potassium (KCa 3.1) channels would counteract pulmonary oedema and hypoxaemia in ventilator-induced lung injury, an experimental model for ARDS. EXPERIMENTAL APPROACH KCa 3.1 channel knockout (Kccn4-/- ) mice were exposed to ventilator-induced lung injury. Control mice exposed to ventilator-induced lung injury were treated with the KCa 3.1 channel inhibitor, senicapoc. The outcomes were oxygenation (PaO2 /FiO2 ratio), lung compliance, lung wet-to-dry weight and protein and cytokines in bronchoalveolar lavage fluid (BALF). KEY RESULTS Ventilator-induced lung injury resulted in lung oedema, decreased lung compliance, a severe drop in PaO2 /FiO2 ratio, increased protein, neutrophils and tumour necrosis factor-alpha (TNF-α) in BALF from wild-type mice compared with Kccn4-/- mice. Pretreatment with senicapoc (10-70 mg·kg-1 ) prevented the reduction in PaO2 /FiO2 ratio, decrease in lung compliance, increased protein and TNF-α. Senicapoc (30 mg·kg-1 ) reduced histopathological lung injury score and neutrophils in BALF. After injurious ventilation, administration of 30 mg·kg-1 senicapoc also improved the PaO2 /FiO2 ratio and reduced lung injury score and neutrophils in the BALF compared with vehicle-treated mice. In human lung epithelial cells, senicapoc decreased TNF-α-induced permeability. CONCLUSIONS AND IMPLICATIONS Genetic deficiency of KCa 3.1 channels and senicapoc improved the PaO2 /FiO2 ratio and decreased the cytokines after a ventilator-induced lung injury. Moreover, senicapoc directly affects lung epithelial cells and blocks neutrophil infiltration in the injured lung. These findings indicate that blocking KCa 3.1 channels is a potential treatment in ARDS-like disease.
Collapse
Affiliation(s)
- Asbjørn Graver Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Peter Carøe Lind
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Susie Mogensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Anne-Sophie Bonde Jensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Asger Granfeldt
- Department of Clinical Medicine, Anaesthesiology, Aarhus University Hospital, Aarhus, Denmark
- Intensive Care, Aarhus University Hospital, Aarhus, Denmark
- Department of Intensive Care Medicine, Randers Regional Hospital, Randers, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Derseh HB, Goodger JQD, Scheerlinck JPY, Samuel CS, Woodrow IE, Palombo EA, Cumming A, Snibson K. The efficacy and safety of pinocembrin in a sheep model of bleomycin-induced pulmonary fibrosis. PLoS One 2021; 16:e0260719. [PMID: 34855848 PMCID: PMC8638960 DOI: 10.1371/journal.pone.0260719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/15/2021] [Indexed: 11/30/2022] Open
Abstract
The primary flavonoid, pinocembrin, is thought to have a variety of medical uses which relate to its reported anti-oxidant, anti-inflammatory, anti-microbial and anti-cancer properties. Some studies have reported that this flavonoid has anti-fibrotic activities. In this study, we investigated whether pinocembrin would impede fibrosis, dampen inflammation and improve lung function in a large animal model of pulmonary fibrosis. Fibrosis was induced in two localized lung segments in each of the 10 sheep participating in the study. This was achieved via two infusions of bleomycin delivered bronchoscopically at a two-week interval. Another lung segment in the same sheep was left untreated, and was used as a healthy control. The animals were kept for a little over 5 weeks after the final infusion of bleomycin. Pinocembrin, isolated from Eucalyptus leaves, was administered to one of the two bleomycin damaged lung segments at a dose of 7 mg. This dose was given once-weekly over 4-weeks, starting one week after the final bleomycin infusion. Lung compliance (as a measure of stiffness) was significantly improved after four weekly administrations of pinocembrin to bleomycin-damaged lung segments. There were significantly lower numbers of neutrophils and inflammatory cells in the bronchoalveolar lavage of bleomycin-infused lung segments that were treated with pinocembrin. Compared to bleomycin damaged lung segments without drug treatment, pinocembrin administration was associated with significantly lower numbers of immuno-positive CD8+ and CD4+ T cells in the lung parenchyma. Histopathology scoring data showed that pinocembrin treatment was associated with significant improvement in inflammation and overall pathology scores. Hydroxy proline analysis showed that the administration of pinocembrin did not reduce the increased collagen content that was induced by bleomycin in this model. Analyses of Masson’s Trichrome stained sections showed that pinocembrin treatment significantly reduced the connective tissue content in lung segments exposed to bleomycin when compared to bleomycin-infused lungs that did not receive pinocembrin. The striking anti-inflammatory and modest anti-fibrotic remodelling effects of pinocembrin administration were likely linked to the compound’s ability to improve lung pathology and functional compliance in this animal model of pulmonary fibrosis.
Collapse
Affiliation(s)
- Habtamu B. Derseh
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (HBD); (KS)
| | - Jason Q. D. Goodger
- School of Biosciences, University of Melbourne, Parkville, Victoria, Australia
| | - Jean-Pierre Y. Scheerlinck
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S. Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Ian E. Woodrow
- School of Ecosystem and Forest Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Enzo A. Palombo
- Department of Chemistry and Biotechnology, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | | | - Ken Snibson
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Victoria, Australia
- * E-mail: (HBD); (KS)
| |
Collapse
|
8
|
Choi S, Kim JA, Li H, Jo SE, Lee H, Kim TH, Kim M, Kim SJ, Suh SH. Anti-inflammatory and anti-fibrotic effects of modafinil in nonalcoholic liver disease. Biomed Pharmacother 2021; 144:112372. [PMID: 34794237 DOI: 10.1016/j.biopha.2021.112372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Small- and intermediate-conductance Ca2+-activated K+ channels, KCa2.3 and KCa3.1, are involved in cellular signaling processes associated with inflammation and fibrosis. KCa2.3 and KCa3.1 are upregulated by proinflammatory cytokines and profibrotic growth factors. Cyclic AMP, which downregulates KCa2.3 and KCa3.1, is elevated by modafinil in cells; accordingly, we investigated whether modafinil exerts anti-inflammatory and anti-fibrotic responses via KCa2.3- and KCa3.1-mediated pathways in high-fat diet (HFD)- or thioacetamide-induced liver disease models in mice. Modafinil was administered orally in the form of a racemate, (R)-isomer, or (S)-isomer. We also determined whether the treatment targeted the profibrotic activity of hepatic stellate cells using immortalized human hepatic stellate cells (LX-2 cells). Modafinil improved HFD- or thioacetamide-induced changes compared to the control, leading to a reduced inflammatory response, collagen deposition, and α-smooth muscle actin expression both in vivo and in vitro. However, modafinil did not relieve HFD-induced steatosis. There were no significant differences in the effects of the (R)- and (S)-isomers of modafinil. KCa2.3 and KCa3.1 were upregulated and catalase was downregulated in liver tissues from thioacetamide- or HFD-induced liver disease models or in TGF-β-treated LX-2 cells. TGF-β-induced upregulation of KCa2.3, KCa3.1, collagen, and α-smooth muscle actin and downregulation of catalase were reversed by modafinil, polyethylene glycol catalase, N-acetylcysteine, siRNA against KCa2.3 or KCa3.1, and Epac inhibitors. Our investigation revealed that modafinil attenuated inflammatory and fibrotic progression via KCa2.3- and KCa3.1-mediated pathways in nonalcoholic hepatitis, suggesting that inhibiting KCa2.3- and KCa3.1-mediated signaling may serve as a novel therapeutic approach for inflammatory and fibrotic liver diseases.
Collapse
Affiliation(s)
- Shinkyu Choi
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Ji Aee Kim
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Haiyan Li
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Seong-Eun Jo
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Huisu Lee
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Tae Hun Kim
- Department of Internal Medicine, School of Medicine, Ewha Womans University, Seoul, Republic of Korea
| | - Minje Kim
- CellionBioMed Inc., Daejeon, Republic of Korea
| | | | - Suk Hyo Suh
- Department of Physiology, School of Medicine, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Evaluation of microRNA expression in a sheep model for lung fibrosis. BMC Genomics 2021; 22:827. [PMID: 34789159 PMCID: PMC8596952 DOI: 10.1186/s12864-021-08073-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibroproliferative disorder that has one of the poorest prognoses amongst interstitial lung diseases. Recently, the finding of aberrant expression levels of miRNAs in IPF patients has drawn significant attention to the involvement of these molecules in the pathogenesis of this disease. Clarification of the differential expression of miRNAs in health and disease may identify novel therapeutic strategies that can be employed in the future to combat IPF. This study evaluates the miRNA expression profiles in a sheep model for lung fibrosis and compares them to the miRNA profiles of both IPF patients and the mouse bleomycin model for pulmonary fibrosis. Pathway enrichment analyses were performed on differentially expressed miRNAs to illustrate which biological mechanisms were associated with lung fibrosis. RESULTS We discovered 49 differentially expressed miRNAs in the sheep fibrosis model, in which 32 miRNAs were significantly down regulated, while 17 miRNAs were significantly upregulated due to bleomycin-induced lung injury. Moreover, the miRNA families miR-29, miR-26, miR-30, let-7, miR-21, miR-19, miR-17 and miR-199 were aberrantly expressed in both sheep and mouse models, with similar differential miRNAs expression observed in IPF cases. Importantly, 18 miRNAs were aberrantly expressed in both the sheep model and IPF patients, but not in mice. CONCLUSION Together with pathway enrichment analyses, these results show that the sheep model can potentially be used to characterize previously unrecognized biological pathways associated with lung fibrosis.
Collapse
|
10
|
Derseh HB, Perera KUE, Dewage SNV, Stent A, Koumoundouros E, Organ L, Pagel CN, Snibson KJ. Tetrathiomolybdate Treatment Attenuates Bleomycin-Induced Angiogenesis and Lung Pathology in a Sheep Model of Pulmonary Fibrosis. Front Pharmacol 2021; 12:700902. [PMID: 34744706 PMCID: PMC8570673 DOI: 10.3389/fphar.2021.700902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive chronic lung disease characterized by excessive extracellular matrix (ECM) deposition in the parenchyma of the lung. Accompanying the fibrotic remodeling, dysregulated angiogenesis has been observed and implicated in the development and progression of pulmonary fibrosis. Copper is known to be required for key processes involved in fibrosis and angiogenesis. We therefore hypothesized that lowering bioavailable serum copper with tetrathiomolybdate could be of therapeutic value for treating pulmonary fibrosis. This study aimed to investigate the effect of tetrathiomolybdate on angiogenesis and fibrosis induced in sheep lung segments infused with bleomycin. Twenty sheep received two fortnightly infusions of either bleomycin (3U), or saline (control) into two spatially separate lung segments. A week after the final bleomycin/saline infusions, sheep were randomly assigned into two groups (n = 10 per group) and received twice-weekly intravenous administrations of either 50 mg tetrathiomolybdate, or sterile saline (vehicle control), for 6 weeks. Vascular density, expressed as the percentage of capillary area to the total area of parenchyma, was determined in lung tissue sections immuno-stained with antibodies against CD34 and collagen type IV. The degree of fibrosis was assessed by histopathology scoring of H&E stained sections and collagen content using Masson's trichrome staining. Lung compliance was measured via a wedged bronchoscope procedure prior to and 7 weeks following final bleomycin infusion. In this large animal model, we show that copper lowering by tetrathiomolybdate chelation attenuates both bleomycin-induced angiogenesis and pulmonary fibrosis. Moreover, tetrathiomolybdate treatment downregulates vascular endothelial growth factor (VEGF) expression, and improved lung function in bleomycin-induced pulmonary fibrosis. Tetrathiomolybdate also suppressed the accumulation of inflammatory cells in bronchoalveolar lavage fluid 2 weeks after bleomycin injury. The molecular mechanism(s) underpinning copper modulation of fibrotic pathways is an important area for future investigation, and it represents a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Habtamu B Derseh
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, Australia
| | | | - Sasika N Vithana Dewage
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Andrew Stent
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Werribee, VIC, Australia
| | - Emmanuel Koumoundouros
- Department of Biomedical Engineering, Melbourne School of Engineering, University of Melbourne, Parkville, VIC, Australia
| | - Louise Organ
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Charles N Pagel
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Ken J Snibson
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Sørensen LK, Petersen A, Granfeldt A, Simonsen U, Hasselstrøm JB. A validated UHPLC-MS/MS method for rapid determination of senicapoc in plasma samples. J Pharm Biomed Anal 2021; 197:113956. [PMID: 33626443 PMCID: PMC7869607 DOI: 10.1016/j.jpba.2021.113956] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
The clinically tested KCa3.1 channel blocker, senicapoc, has been proven to have excellent pharmacological properties and prior clinical trials found it to be safe for use in patients with sickle cell anaemia. Currently, several preclinical projects are aiming to repurpose senicapoc for other indications, but well-described analytical methods in the literature are lacking. Our aim was to develop a sensitive, rapid and accurate ultra-high-performance liquid chromatography-tandem mass spectrometry method using pneumatically assisted electrospray ionisation (UHPLC-ESI-MS/MS) suitable for the determination of senicapoc in plasma samples. Unfortunately, direct analysis of senicapoc in crude acetonitrile extracts of human plasma samples by UHPLC-ESI-MS/MS was subjected to significant and variable ion suppression from coeluting phospholipids (PLs). The interferences were mainly caused by the presence of phosphatidylcholine and phosphatidylethanolamine classes of PLs, including their lyso-products. However, the PLs were easily removed from crude extracts by filtration through a sorbent with Lewis acid properties which decreased the total ion suppression effect to approximately 5%. Based on this technique, a simple high-throughput UHPLC-MS/MS method was developed and validated for the determination of senicapoc in 100-μL plasma samples. The lower limit of quantification was 0.1 ng/mL. The mean true extraction recovery was close to 100 %. The relative intra-laboratory reproducibility standard deviations of the measured concentrations were 8% and 4% at concentrations of 0.1 ng/mL and 250 ng/mL, respectively. The trueness expressed as the relative bias of the test results was within ± 2% at concentrations of 1 ng/mL or higher.
Collapse
Affiliation(s)
- Lambert K Sørensen
- Section for Forensic Chemistry, Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Asbjørn Petersen
- Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Asger Granfeldt
- Department of Clinical Medicine, Anaesthesiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200, Aarhus N, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000, Aarhus C, Denmark
| | - Jørgen B Hasselstrøm
- Section for Forensic Chemistry, Department of Forensic Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
12
|
Increased Levels of ER Stress and Apoptosis in a Sheep Model for Pulmonary Fibrosis Are Alleviated by In Vivo Blockade of the KCa3.1 Ion Channel. Can Respir J 2021; 2021:6683195. [PMID: 33828632 PMCID: PMC8004363 DOI: 10.1155/2021/6683195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/26/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease, characterized by progressive damage to the lung tissues. Apoptosis and endoplasmic reticulum stress (ER stress) in type II alveolar epithelial cells (AECs) and lung macrophages have been linked with the development of IPF. Therefore, apoptosis- and ER stress-targeted therapies have drawn attention as potential avenues for treatment of IPF. The calcium-activated potassium ion channel KCa3.1 has been proposed as a potential therapeutic target for fibrotic diseases including IPF. While KCa3.1 is expressed in AECs and macrophages, its influence on ER stress and apoptosis during the disease process is unclear. We utilized a novel sheep model of pulmonary fibrosis to demonstrate that apoptosis and ER stress occur in type II AECs and macrophages in sheep with bleomycin-induced lung fibrosis. Apoptosis in type II AEC and macrophages was identified using the TUNEL method of tagging fragmented nuclear DNA, while ER stress was characterized by increased expression of GRP-78 ER chaperone proteins. We demonstrated that apoptosis and ER stress in type II AECs and macrophages increased significantly 2 weeks after the final bleomycin infusion and remained high for up to 7 weeks post-bleomycin injury. Senicapoc treatment significantly reduced the rates of ER stress in type II AECs and macrophages that were resident in bleomycin-infused lung segments. There were also significant reductions in the rates of apoptosis of type II AECs and macrophages in the lung segments of senicapoc-treated sheep. In vivo blockade of the KCa3.1 ion channel alleviates the ER stress and apoptosis in type II AECs and macrophages, and this effect potentially contributes to the anti-fibrotic effects of senicapoc.
Collapse
|
13
|
Roach KM, Castells E, Dixon K, Mason S, Elliott G, Marshall H, Poblocka MA, Macip S, Richardson M, Khalfaoui L, Bradding P. Evaluation of Pirfenidone and Nintedanib in a Human Lung Model of Fibrogenesis. Front Pharmacol 2021; 12:679388. [PMID: 34712131 PMCID: PMC8546112 DOI: 10.3389/fphar.2021.679388] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Introduction: Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal lung disease with a poor prognosis and increasing incidence. Pirfenidone and nintedanib are the only approved treatments for IPF but have limited efficacy and their mechanisms of action are poorly understood. Here we have examined the effects of pirfenidone and nintedanib in a human model of lung fibrogenesis, and compared these with the putative anti-fibrotic compounds Lipoxin A4 (LXA4), and senicapoc, a KCa3.1 ion channel blocker. Methods: Early fibrosis was induced in cultured human lung parenchyma using TGFβ1 for 7 days, ± pirfenidone, nintedanib, or LXA4. Pro-fibrotic responses were examined by RT-PCR, immunohistochemistry and soluble collagen secretion. Results: Thirty six out of eighty four IPF and fibrosis-associated genes tested were significantly upregulated by TGFβ1 in human lung parenchyma with a ≥0.5 log2FC (n = 32). Nintedanib (n = 13) reduced the mRNA expression of 14 fibrosis-associated genes including MMPs (MMP1,-4,-13,-14), integrin α2, CXCR4 and PDGFB, but upregulated α-smooth muscle actin (αSMA). Pirfenidone only reduced mRNA expression for MMP3 and -13. Senicapoc (n = 11) previously attenuated the expression of 28 fibrosis-associated genes, including αSMA, several growth factors, collagen type III, and αV/β6 integrins. Pirfenidone and nintedanib significantly inhibited TGFβ1-induced fibroblast proliferation within the tissue, but unlike senicapoc, neither pirfenidone nor nintedanib prevented increases in tissue αSMA expression. LXA4 was ineffective. Conclusions: Pirfenidone and nintedanib demonstrate modest anti-fibrotic effects and provide a benchmark for anti-fibrotic activity of new drugs in human lung tissue. Based on these data, we predict that the KCa3.1 blocker senicapoc will show greater benefit than either of these licensed drugs in IPF.
Collapse
Affiliation(s)
- K M Roach
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - E Castells
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - K Dixon
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - S Mason
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - G Elliott
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - H Marshall
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - M A Poblocka
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom
| | - S Macip
- Mechanisms of Cancer and Ageing Lab, Department of Molecular and Cell Biology, University of Leicester, Leicester, United Kingdom.,FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - M Richardson
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - L Khalfaoui
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - P Bradding
- Department of Respiratory Sciences, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
14
|
Derseh HB, Dewage SNV, Perera UE, Koumoundouros E, Pagel CN, Organ L, Snibson KJ. Small airway remodeling in a sheep model of bleomycin-induced pulmonary fibrosis. Exp Lung Res 2020; 46:409-419. [PMID: 34346276 DOI: 10.1080/01902148.2020.1834015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Although IPF is described traditionally as a disease affecting lung parenchyma, there is renewed interest in the alterations in the structure and function of the small airways in both IPF patients, and animal models of pulmonary fibrosis. Small airway remodeling may contribute to the pathophysiology of pulmonary fibrosis. Given the dearth of knowledge of small airway changes in pulmonary fibrosis, this study aims to assess the structural remodeling, as well as functional changes associated with bleomycin-injured small airways in a sheep model of pulmonary fibrosis. MATERIALS AND METHODS Two separate lung segments in ten sheep received two challenges of either 3 IU bleomycin, or saline (control), two weeks apart. The animals were euthanized seven weeks after the final bleomycin injury. Airflow resistance in the infused segments was measured with a wedged-bronchoscope procedure. This parameter was measured at baseline before bleomycin/saline-infusion, and at 2-, 4-, and 7-weeks after the final bleomycin-infusion. Inflammation and fibrosis in the airways were assessed by semi-quantitative morphological parameters. The density of blood vessels in the small airway walls was assessed in lung tissue sections immuno-stained with antibodies against collagen type IV. RESULTS There were a number of changes in the distal airways of bleomycin-infused lung segments. Bleomycin exposure significantly elevated airway resistance in these lung segments when compared to saline-infused control lung segments. In the peribronchial and peribronchiolar regions of the small airways, there were significantly increased levels of inflammation, fibrosis, airway wall area, and collagen deposition in bleomycin-infused airways when compared to saline-infused airways. Bronchial blood vessel density was not significantly different between bleomycin-and saline-infused lung segments. CONCLUSIONS In summary, our results indicate that the distal airways are involved in the pathology induced by bleomycin in this sheep model. This suggests that the sheep model may be useful for studying small airway remodeling in pulmonary fibrosis.
Collapse
Affiliation(s)
- Habtamu B Derseh
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| | - Sasika N Vithana Dewage
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| | - Udari E Perera
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| | | | - Charles N Pagel
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| | - Louise Organ
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Ken J Snibson
- Melbourne Veterinary School, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Australia
| |
Collapse
|
15
|
Shrestha S, Cho W, Stump B, Imani J, Lamattina AM, Louis PH, Pazzanese J, Rosas IO, Visner G, Perrella MA, El-Chemaly S. FK506 induces lung lymphatic endothelial cell senescence and downregulates LYVE-1 expression, with associated decreased hyaluronan uptake. Mol Med 2020; 26:75. [PMID: 32736525 PMCID: PMC7395348 DOI: 10.1186/s10020-020-00204-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/24/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Therapeutic lymphangiogenesis in an orthotopic lung transplant model has been shown to improve acute allograft rejection that is mediated at least in part through hyaluronan drainage. Lymphatic vessel endothelial hyaluronan receptor (LYVE-1) expressed on the surface of lymphatic endothelial cells plays important roles in hyaluronan uptake. The impact of current immunosuppressive therapies on lung lymphatic endothelial cells is largely unknown. We tested the hypothesis that FK506, the most commonly used immunosuppressant after lung transplantation, induces lung lymphatic endothelial cell dysfunction. METHODS Lung lymphatic endothelial cells were cultured in vitro and treated with FK506. Telomerase activity was measured using the TRAP assay. Protein expression of LYVE-1 and senescence markers p21 and β-galactosidase was assessed with western blotting. Matrigel tubulation assay were used to investigate the effects of FK506 on TNF-α-induced lymphangiogenesis. Dual luciferase reporter assay was used to confirm NFAT-dependent transcriptional regulation of LYVE-1. Flow cytometry was used to examine the effects of FK506 on LYVE-1 in precision-cut-lung-slices ex vivo and on hyaluronan uptake in vitro. RESULTS In vitro, FK506 downregulated telomerase reverse transcriptase expression, resulting in decreased telomerase activity and subsequent induction of p21 expression and cell senescence. Treatment with FK506 decreased LYVE-1 mRNA and protein levels and resulted in decreased LEC HA uptake. Similar result showing reduction of LYVE-1 expression when treated with FK506 was observed ex vivo. We identified a putative NFAT binding site on the LYVE-1 promoter and cloned this region of the promoter in a luciferase-based reporter construct. We showed that this NFAT binding site regulates LYVE-1 transcription, and mutation of this binding site blunted FK506-dependent downregulation of LYVE-1 promoter-dependent transcription. Finally, FK506-treated lymphatic endothelial cells show a blunted response to TNF-α-mediated lymphangiogenesis. CONCLUSION FK506 alters lymphatic endothelial cell molecular characteristics and causes lymphatic endothelial cell dysfunction in vitro and ex vivo. These effects of FK506 on lymphatic endothelial cell may impair the ability of the transplanted lung to drain hyaluronan macromolecules in vivo. The implications of our findings on the long-term health of lung allografts merit more investigation.
Collapse
Affiliation(s)
- Shikshya Shrestha
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Woohyun Cho
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Present Address: Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Benjamin Stump
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jewel Imani
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Anthony M Lamattina
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Pierce H Louis
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - James Pazzanese
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gary Visner
- Deparmtent of Pediatrics, Boston Children Hospital, Harvard Medical School, Boston, MA, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
Roach KM, Bradding P. Ca 2+ signalling in fibroblasts and the therapeutic potential of K Ca3.1 channel blockers in fibrotic diseases. Br J Pharmacol 2020; 177:1003-1024. [PMID: 31758702 DOI: 10.1111/bph.14939] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
The role of Ca2+ signalling in fibroblasts is of great interest in fibrosis-related diseases. Intracellular free Ca2+ ([Ca2+ ]i ) is a ubiquitous secondary messenger, regulating a number of cellular functions such as secretion, metabolism, differentiation, proliferation and contraction. The intermediate conductance Ca2+ -activated K+ channel KCa 3.1 is pivotal in Ca2+ signalling and plays a central role in fibroblast processes including cell activation, migration and proliferation through the regulation of cell membrane potential. Evidence from a number of approaches demonstrates that KCa 3.1 plays an important role in the development of many fibrotic diseases, including idiopathic pulmonary, renal tubulointerstitial fibrosis and cardiovascular disease. The KCa 3.1 selective blocker senicapoc was well tolerated in clinical trials for sickle cell disease, raising the possibility of rapid translation to the clinic for people suffering from pathological fibrosis. This review after analysing all the data, concludes that targeting KCa 3.1 should be a high priority for human fibrotic disease.
Collapse
Affiliation(s)
- Katy M Roach
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Peter Bradding
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
17
|
Derseh HB, Dewage SNV, Perera KUE, Pagel CN, Koumoundouros E, Organ L, Snibson KJ. K Ca3.1 channel blockade attenuates microvascular remodelling in a large animal model of bleomycin-induced pulmonary fibrosis. Sci Rep 2019; 9:19893. [PMID: 31882807 PMCID: PMC6934539 DOI: 10.1038/s41598-019-56412-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 12/11/2019] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with limited therapeutic options and poor prognosis. IPF has been associated with aberrant vascular remodelling, however the role of vascular remodelling in pulmonary fibrosis is poorly understood. Here, we used a novel segmental challenge model of bleomycin-induced pulmonary fibrosis in sheep to evaluate the remodelling of the pulmonary vasculature, and to investigate the changes to this remodelling after the administration of the KCa3.1 channel inhibitor, senicapoc, compared to the FDA-approved drug pirfenidone. We demonstrate that in vehicle-treated sheep, bleomycin-infused lung segments had significantly higher blood vessel density when compared to saline-infused control segments in the same sheep. These microvascular density changes were significantly attenuated by senicapoc treatment. The increases in vascular endothelial growth factor (VEGF) expression and endothelial cell proliferation in bleomycin-infused lung segments were significantly reduced in sheep treated with the senicapoc, when compared to vehicle-treated controls. These parameters were not significantly suppressed with pirfenidone treatment. Senicapoc treatment attenuated vascular remodelling through inhibition of capillary endothelial cell proliferation and VEGF expression. These findings suggest a potential new mode of action for the novel drug senicapoc which may contribute to its efficacy in combatting pulmonary fibrosis.
Collapse
Affiliation(s)
- Habtamu B Derseh
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia.
| | - Sasika N Vithana Dewage
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Kopiyawaththage U E Perera
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Charles N Pagel
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Emmanuel Koumoundouros
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Victoria, Australia
| | - Louise Organ
- Division of Respiratory Medicine, University of Nottingham, Nottingham, UK
| | - Ken J Snibson
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
18
|
Dewage SNV, Organ L, Koumoundouros E, Derseh HB, Perera KUE, Samuel CS, Stent AW, Snibson KJ. The efficacy of pirfenidone in a sheep model of pulmonary fibrosis. Exp Lung Res 2019; 45:310-322. [PMID: 31762329 DOI: 10.1080/01902148.2019.1695019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrotic lung disease with unknown cause. While the drugs nintedanib and pirfenidone have been approved for the treatment of IPF, they only slow disease progression and can induce several side-effects, suggesting that there is still an unmet need to develop new efficacious drugs, and interventions strategies, to combat this disease. We have recently developed a sheep model of pulmonary fibrosis for the preclinical testing of novel anti-fibrotic drugs. The aim of this study was to assess the effects of pirfenidone to ascertain its suitability as a benchmark for comparing other novel therapeutics in this sheep model. To initiate localized fibrosis, sheep were given two infusions of bleomycin (0.6 U/ml per infusion), a fortnight apart, to a specific lung segment. The contralateral lung segment in each sheep was infused with saline to act as an internal control. Two weeks after the final bleomycin infusion, either pirfenidone or methylcellulose (vehicle control) were administered orally to sheep twice daily for 5 weeks. Results showed that sheep treated with pirfenidone had improved lung function, ameliorated fibrotic pathology, lower numbers of active myofibroblasts, and reduced extra cellular matrix deposition when compared with the relevant measurements obtained from control sheep treated with vehicle. This study showed that pirfenidone can attenuate bleomycin-induced pulmonary fibrosis in sheep, and can therefore be used as a positive control to assess other novel therapeutics for IPF in this model.
Collapse
Affiliation(s)
- Sasika N V Dewage
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| | - Louise Organ
- Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Emmanuel Koumoundouros
- Department of Electrical and Electronic Engineering, The University of Melbourne, Parkville, Australia
| | - Habtamu B Derseh
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| | | | | | - Andrew W Stent
- Faculty of Veterinary Science and Agricultural Science, The University of Melbourne, Werribee, Australia
| | - Ken J Snibson
- Faculty of Veterinary and Agricultural Science, The University of Melbourne, Parkville, Australia
| |
Collapse
|
19
|
Brown BM, Shim H, Christophersen P, Wulff H. Pharmacology of Small- and Intermediate-Conductance Calcium-Activated Potassium Channels. Annu Rev Pharmacol Toxicol 2019; 60:219-240. [PMID: 31337271 DOI: 10.1146/annurev-pharmtox-010919-023420] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The three small-conductance calcium-activated potassium (KCa2) channels and the related intermediate-conductance KCa3.1 channel are voltage-independent K+ channels that mediate calcium-induced membrane hyperpolarization. When intracellular calcium increases in the channel vicinity, it calcifies the flexible N lobe of the channel-bound calmodulin, which then swings over to the S4-S5 linker and opens the channel. KCa2 and KCa3.1 channels are highly druggable and offer multiple binding sites for venom peptides and small-molecule blockers as well as for positive- and negative-gating modulators. In this review, we briefly summarize the physiological role of KCa channels and then discuss the pharmacophores and the mechanism of action of the most commonly used peptidic and small-molecule KCa2 and KCa3.1 modulators. Finally, we describe the progress that has been made in advancing KCa3.1 blockers and KCa2.2 negative- and positive-gating modulators toward the clinic for neurological and cardiovascular diseases and discuss the remaining challenges.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | - Heesung Shim
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| | | | - Heike Wulff
- Department of Pharmacology, University of California, Davis, California 95616, USA;
| |
Collapse
|
20
|
Pethő Z, Najder K, Bulk E, Schwab A. Mechanosensitive ion channels push cancer progression. Cell Calcium 2019; 80:79-90. [PMID: 30991298 DOI: 10.1016/j.ceca.2019.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
In many cases, the mechanical properties of a tumor are different from those of the host tissue. Mechanical cues regulate cancer development by affecting both tumor cells and their microenvironment, by altering cell migration, proliferation, extracellular matrix remodeling and metastatic spread. Cancer cells sense mechanical stimuli such as tissue stiffness, shear stress, tissue pressure of the extracellular space (outside-in mechanosensation). These mechanical cues are transduced into a cellular response (e. g. cell migration and proliferation; inside-in mechanotransduction) or to a response affecting the microenvironment (e. g. inducing a fibrosis or building up growth-induced pressure; inside-out mechanotransduction). These processes heavily rely on mechanosensitive membrane proteins, prominently ion channels. Mechanosensitive ion channels are involved in the Ca2+-signaling of the tumor and stroma cells, both directly, by mediating Ca2+ influx (e. g. Piezo and TRP channels), or indirectly, by maintaining the electrochemical gradient necessary for Ca2+ influx (e. g. K2P, KCa channels). This review aims to discuss the diverse roles of mechanosenstive ion channels in cancer progression, especially those involved in Ca2+-signaling, by pinpointing their functional relevance in tumor pathophysiology.
Collapse
Affiliation(s)
- Zoltán Pethő
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany.
| | - Karolina Najder
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Etmar Bulk
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Albrecht Schwab
- Institut für Physiologie II, Robert-Koch-Str. 27b, 48149 Münster, Germany
| |
Collapse
|
21
|
Abstract
Fibrosis is a medical condition characterized by an excessive deposition of extracellular matrix compounds such as collagen in tissues. Fibrotic lesions are present in many diseases and can affect all organs. The excessive extracellular matrix accumulation in these conditions can often have serious consequences and in many cases be life-threatening. A typical event seen in many fibrotic conditions is a profound accumulation of mast cells (MCs), suggesting that these cells can contribute to the pathology. Indeed, there is now substantialv evidence pointing to an important role of MCs in fibrotic disease. However, investigations from various clinical settings and different animal models have arrived at partly contradictory conclusions as to how MCs affect fibrosis, with many studies suggesting a detrimental role of MCs whereas others suggest that MCs can be protective. Here, we review the current knowledge of how MCs can affect fibrosis.
Collapse
Affiliation(s)
- Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|
22
|
Matsui M, Terasawa K, Kajikuri J, Kito H, Endo K, Jaikhan P, Suzuki T, Ohya S. Histone Deacetylases Enhance Ca 2+-Activated K⁺ Channel K Ca3.1 Expression in Murine Inflammatory CD4⁺ T Cells. Int J Mol Sci 2018; 19:ijms19102942. [PMID: 30262728 PMCID: PMC6213394 DOI: 10.3390/ijms19102942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022] Open
Abstract
The up-regulated expression of the Ca2+-activated K+ channel KCa3.1 in inflammatory CD4+ T cells has been implicated in the pathogenesis of inflammatory bowel disease (IBD) through the enhanced production of inflammatory cytokines, such as interferon-γ (IFN-γ). However, the underlying mechanisms have not yet been elucidated. The objective of the present study is to clarify the involvement of histone deacetylases (HDACs) in the up-regulation of KCa3.1 in the CD4+ T cells of IBD model mice. The expression levels of KCa3.1 and its regulators, such as function-modifying molecules and transcription factors, were quantitated using a real-time polymerase chain reaction (PCR) assay, Western blotting, and depolarization responses, which were induced by the selective KCa3.1 blocker TRAM-34 (1 μM) and were measured using a voltage-sensitive fluorescent dye imaging system. The treatment with 1 μM vorinostat, a pan-HDAC inhibitor, for 24 h repressed the transcriptional expression of KCa3.1 in the splenic CD4+ T cells of IBD model mice. Accordingly, TRAM-34-induced depolarization responses were significantly reduced. HDAC2 and HDAC3 were significantly up-regulated in the CD4+ T cells of IBD model mice. The down-regulated expression of KCa3.1 was observed following treatments with the selective inhibitors of HDAC2 and HDAC3. The KCa3.1 K+ channel regulates inflammatory cytokine production in CD4+ T cells, mediating epigenetic modifications by HDAC2 and HDAC3.
Collapse
Affiliation(s)
- Miki Matsui
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Kyoko Terasawa
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Kyoko Endo
- Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan.
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | - Pattaporn Jaikhan
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 403-8334, Japan.
| | - Takayoshi Suzuki
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 403-8334, Japan.
| | - Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| |
Collapse
|
23
|
Xie H, Lu J, Zhu Y, Meng X, Wang R. The KCa3.1 blocker TRAM-34 inhibits proliferation of fibroblasts in paraquat-induced pulmonary fibrosis. Toxicol Lett 2018; 295:408-415. [PMID: 30036685 DOI: 10.1016/j.toxlet.2018.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 07/15/2018] [Accepted: 07/19/2018] [Indexed: 01/30/2023]
Abstract
KCa3.1, a Ca2+-activated K+ channel, plays an important role in modulating calcium signaling and maintaining membrane potential during cell activation. It has been reported to promote fibroblast function in many fibrotic diseases. However, the role of KCa3.1 in the pathophysiology of pulmonary fibrosis after paraquat (PQ) poisoning has not been studied. A rat model of PQ poisoning was used. After treatment with TRAM-34, which is a highly selective KCa3.1 blocker, the expression of KCa3.1, TGF-β1 and α-SMA were evaluated via Western blot, histology and other assays. Bromodeoxyuridine (BrdU) marking and MTT assay were used to measure primary rat pulmonary fibroblast proliferation. The results showed that KCa3.1 expression was elevated after PQ poisoning. Blockade of KCa3.1 alleviated PQ-induced pulmonary inflammation and fibrosis. Blockade of KCa3.1 also attenuated the level of collagen I and α-SMA and the proliferation of fibroblasts. However, TGF-β1 expression remained unaffected by blockade of KCa3.1 in rat lung tissues after PQ poisoning. The present study suggests that KCa3.1 expression increased and might promote pulmonary fibroblast proliferation in PQ-induced pulmonary fibrosis. In addition, we confirmed that TRAM-34 attenuates proliferation and collagen secretion of fibroblasts. Our findings indicated that TRAM-34 might inhibit PQ-induced proliferation of pulmonary fibroblasts and prevent progression of lung fibrosis.
Collapse
Affiliation(s)
- Hui Xie
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, China
| | - Jian Lu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, China
| | - Yong Zhu
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 201620, China
| | - Ruilan Wang
- Department of Critical Care Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 201620, China.
| |
Collapse
|
24
|
Anumanthan G, Gupta S, Fink MK, Hesemann NP, Bowles DK, McDaniel LM, Muhammad M, Mohan RR. KCa3.1 ion channel: A novel therapeutic target for corneal fibrosis. PLoS One 2018; 13:e0192145. [PMID: 29554088 PMCID: PMC5858751 DOI: 10.1371/journal.pone.0192145] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/17/2018] [Indexed: 12/16/2022] Open
Abstract
Vision impairment from corneal fibrosis is a common consequence of irregular corneal wound healing after injury. Intermediate-conductance calmodulin/calcium-activated K+ channels 3.1 (KCa3.1) play an important role in cell cycle progression and cellular proliferation. Proliferation and differentiation of corneal fibroblasts to myofibroblasts can lead to corneal fibrosis after injury. KCa3.1 has been shown in many non-ocular tissues to promote fibrosis, but its role in corneal fibrosis is still unknown. In this study, we characterized the expression KCa3.1 in the human cornea and its role in corneal wound healing in vivo using a KCa3.1 knockout (KCa3.1-/-) mouse model. Additionally, we tested the hypothesis that blockade of KCa3.1 by a selective KCa3.1 inhibitor, TRAM-34, could augment a novel interventional approach for controlling corneal fibrosis in our established in vitro model of corneal fibrosis. The expression of KCa3.1 gene and protein was analyzed in human and murine corneas. Primary human corneal fibroblast (HCF) cultures were used to examine the potential of TRAM-34 in treating corneal fibrosis by measuring levels of pro-fibrotic genes, proteins, and cellular migration using real-time quantitative qPCR, Western blotting, and scratch assay, respectively. Cytotoxicity of TRAM-34 was tested with trypan blue assay, and pro-fibrotic marker expression was tested in KCa3.1-/-. Expression of KCa3.1 mRNA and protein was detected in all three layers of the human cornea. The KCa3.1-/- mice demonstrated significantly reduced corneal fibrosis and expression of pro-fibrotic marker genes such as collagen I and α-smooth muscle actin (α-SMA), suggesting that KCa3.1 plays an important role corneal wound healing in vivo. Pharmacological treatment with TRAM-34 significantly attenuated corneal fibrosis in vitro, as demonstrated in HCFs by the inhibition TGFβ-mediated transcription of pro-fibrotic collagen I mRNA and α-SMA mRNA and protein expression (p<0.001). No evidence of cytotoxicity was observed. Our study suggests that KCa3.1 regulates corneal wound healing and that blockade of KCa3.1 by TRAM-34 offers a potential therapeutic strategy for developing therapies to cure corneal fibrosis in vivo.
Collapse
Affiliation(s)
- Govindaraj Anumanthan
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| | - Suneel Gupta
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| | - Michael K. Fink
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| | - Nathan P. Hesemann
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Douglas K. Bowles
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Lindsey M. McDaniel
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Maaz Muhammad
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
| | - Rajiv R. Mohan
- Harry S. Truman Memorial Veteran Hospital, Columbia, Missouri, United States of America
- Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, United States of America
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, Missouri, United States of America
- Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
25
|
Roach KM, Sutcliffe A, Matthews L, Elliott G, Newby C, Amrani Y, Bradding P. A model of human lung fibrogenesis for the assessment of anti-fibrotic strategies in idiopathic pulmonary fibrosis. Sci Rep 2018; 8:342. [PMID: 29321510 PMCID: PMC5762721 DOI: 10.1038/s41598-017-18555-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 12/14/2017] [Indexed: 11/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with limited therapeutic options. KCa3.1 ion channels play a critical role in TGFβ1-dependent pro-fibrotic responses in human lung myofibroblasts. We aimed to develop a human lung parenchymal model of fibrogenesis and test the efficacy of the selective KCa3.1 blocker senicapoc. 2 mm3 pieces of human lung parenchyma were cultured for 7 days in DMEM ± TGFβ1 (10 ng/ml) and pro-fibrotic pathways examined by RT-PCR, immunohistochemistry and collagen secretion. Following 7 days of culture with TGFβ1, 41 IPF- and fibrosis-associated genes were significantly upregulated. Immunohistochemical staining demonstrated increased expression of ECM proteins and fibroblast-specific protein after TGFβ1-stimulation. Collagen secretion was significantly increased following TGFβ1-stimulation. These pro-fibrotic responses were attenuated by senicapoc, but not by dexamethasone. This 7 day ex vivo model of human lung fibrogenesis recapitulates pro-fibrotic events evident in IPF and is sensitive to KCa3.1 channel inhibition. By maintaining the complex cell-cell and cell-matrix interactions of human tissue, and removing cross-species heterogeneity, this model may better predict drug efficacy in clinical trials and accelerate drug development in IPF. KCa3.1 channels are a promising target for the treatment of IPF.
Collapse
Affiliation(s)
- Katy M Roach
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK.
| | - Amanda Sutcliffe
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Laura Matthews
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Gill Elliott
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Chris Newby
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Yassine Amrani
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Peter Bradding
- Institute for Lung Health, Respiratory Medicine, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| |
Collapse
|
26
|
Brown BM, Pressley B, Wulff H. KCa3.1 Channel Modulators as Potential Therapeutic Compounds for Glioblastoma. Curr Neuropharmacol 2018; 16:618-626. [PMID: 28676010 PMCID: PMC5997873 DOI: 10.2174/1570159x15666170630164226] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The intermediate-conductance Ca2+-activated K+ channel KCa3.1 is widely expressed in cells of the immune system such as T- and B-lymphocytes, mast cells, macrophages and microglia, but also found in dedifferentiated vascular smooth muscle cells, fibroblasts and many cancer cells including pancreatic, prostate, leukemia and glioblastoma. In all these cell types KCa3.1 plays an important role in cellular activation, migration and proliferation by regulating membrane potential and Ca2+ signaling. METHODS AND RESULTS KCa3.1 therefore constitutes an attractive therapeutic target for diseases involving excessive proliferation or activation of one more of these cell types and researchers both in academia and in the pharmaceutical industry have developed several potent and selective small molecule inhibitors of KCa3.1. This article will briefly review the available compounds (TRAM-34, senicapoc, NS6180), their binding sites and mechanisms of action, and then discuss the potential usefulness of these compounds for the treatment of brain tumors based on their brain penetration and their efficacy in reducing microglia activation in animal models of ischemic stroke and Alzheimer's disease. CONCLUSION Senicapoc, which has previously been in Phase III clinical trials, would be available for repurposing, and could be used to quickly translate findings made with other KCa3.1 blocking tool compounds into clinical trials.
Collapse
Affiliation(s)
- Brandon M Brown
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Brandon Pressley
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA 95616, United States
| |
Collapse
|