1
|
Dorababu A, Maraswami M. Recent Advances (2015-2020) in Drug Discovery for Attenuation of Pulmonary Fibrosis and COPD. Molecules 2023; 28:molecules28093674. [PMID: 37175084 PMCID: PMC10179756 DOI: 10.3390/molecules28093674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
A condition of scarring of lung tissue due to a wide range of causes (such as environmental pollution, cigarette smoking (CS), lung diseases, some medications, etc.) has been reported as pulmonary fibrosis (PF). This has become a serious problem all over the world due to the lack of efficient drugs for treatment or cure. To date, no drug has been designed that could inhibit fibrosis. However, few medications have been reported to reduce the rate of fibrosis. Meanwhile, ongoing research indicates pulmonary fibrosis can be treated in its initial stages when symptoms are mild. Here, an attempt is made to summarize the recent studies on the effects of various chemical drugs that attenuate PF and increase patients' quality of life. The review is classified based on the nature of the drug molecules, e.g., natural/biomolecule-based, synthetic-molecule-based PF inhibitors, etc. Here, the mechanisms through which the drug molecules attenuate PF are discussed. It is shown that inhibitory molecules can significantly decrease the TGF-β1, profibrotic factors, proteins responsible for inflammation, pro-fibrogenic cytokines, etc., thereby ameliorating the progress of PF. This review may be useful in designing better drugs that could reduce the fibrosis process drastically or even cure the disease to some extent.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Chemistry, SRMPP Government First Grade College, Huvinahadagali 583219, India
| | - Manikantha Maraswami
- Department of Chemistry, Abzena LLC., 360 George Patterson Blvd, Bristol, PA 19007, USA
| |
Collapse
|
2
|
Kayalar O, Oztay F. CGRP induces myofibroblast differentiation and the production of extracellular matrix in MRC5s via autocrine and paracrine signalings. J Biochem Mol Toxicol 2022; 36:e23204. [PMID: 36056781 DOI: 10.1002/jbt.23204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 04/05/2022] [Accepted: 08/12/2022] [Indexed: 11/08/2022]
Abstract
There are contradictory views on which calcitonin gene-related peptide (CGRP) causes pulmonary fibrosis. Fibrotic potency of CGRP was tested and compared to that of transforming growth factor-β (TGF-β). Myofibroblast differentiation, cell proliferation, and activations of TGF-β and Wnt pathways were examined for 24, 48, and 72 h in A549 and MRC5 cell lines stimulated with CGRP and TGF-β. CGRP-induced cell proliferation in MRC5s early on while cell proliferation in A549 occurred progressively. CGRP promoted fibroblast-myofibroblast differentiation by inducing the transcription of ACTA2, COL1A1, SMAD2/3, and SMAD4 genes, the production of collagen, fibronectin, α-smooth muscle actin, and activation of TGF-β signaling starting from 24 h. Additionally, TGF-β signaling induced by CGRP decreased the DKK1 level and activated the Wnt signaling in MRC5s. After CGRP stimulation, Wnt7a levels were increased from 24 to 72 h, while Wnt5a levels were elevated at 72 h in MRC5s. CGRP did not induce epithelial-mesenchymal transition in A549s, unlike TGF-β. A comparison of fibrotic potency of CGRP and TGF-β showed that TGF-β is a powerful profibrotic molecule and induces earlier myofibroblast differentiation. Even so, CGRP promotes myofibroblast differentiation and extracellular matrix production by inducing Smad-dependent-TGF-β and Wnt signalings via autocrine and paracrine signalings in MRC5s.
Collapse
Affiliation(s)
- Ozgecan Kayalar
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey.,Koç University Research Centre for Translational Medicine (KUTTAM), School of Medicine, Koç University, Istanbul, Turkey
| | - Fusun Oztay
- Department of Biology, Science Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
3
|
Promises and Challenges of Cell-Based Therapies to Promote Lung Regeneration in Idiopathic Pulmonary Fibrosis. Cells 2022; 11:cells11162595. [PMID: 36010671 PMCID: PMC9406501 DOI: 10.3390/cells11162595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 12/17/2022] Open
Abstract
The lung epithelium is constantly exposed to harmful agents present in the air that we breathe making it highly susceptible to damage. However, in instances of injury to the lung, it exhibits a remarkable capacity to regenerate injured tissue thanks to the presence of distinct stem and progenitor cell populations along the airway and alveolar epithelium. Mechanisms of repair are affected in chronic lung diseases such as idiopathic pulmonary fibrosis (IPF), a progressive life-threatening disorder characterized by the loss of alveolar structures, wherein excessive deposition of extracellular matrix components cause the distortion of tissue architecture that limits lung function and impairs tissue repair. Here, we review the most recent findings of a study of epithelial cells with progenitor behavior that contribute to tissue repair as well as the mechanisms involved in mouse and human lung regeneration. In addition, we describe therapeutic strategies to promote or induce lung regeneration and the cell-based strategies tested in clinical trials for the treatment of IPF. Finally, we discuss the challenges, concerns and limitations of applying these therapies of cell transplantation in IPF patients. Further research is still required to develop successful strategies focused on cell-based therapies to promote lung regeneration to restore lung architecture and function.
Collapse
|
4
|
Saygili E, Devamoglu U, Goker-Bagca B, Goksel O, Biray-Avci C, Goksel T, Yesil-Celiktas O. A drug-responsive multicellular human spheroid model to recapitulate drug-induced pulmonary fibrosis. Biomed Mater 2022; 17. [PMID: 35617946 DOI: 10.1088/1748-605x/ac73cd] [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: 12/31/2021] [Accepted: 05/26/2022] [Indexed: 11/12/2022]
Abstract
Associated with a high mortality rate, pulmonary fibrosis (PF) is the end stage of several interstitial lung diseases. Although many factors are linked to PF progression, initiation of the fibrotic process remains to be studied. Current research focused on generating new strategies to gain a better understanding of the underlying disease mechanism as the animal models remain insufficient to reflect human physiology. Herein, to account complex cellular interactions within the fibrotic tissue, a multicellular spheroid (MCS) model where human bronchial epithelial cells incorporated with human lung fibroblasts was generated and treated with bleomycin (BLM) to emulate drug-induced PF. Recapitulating the epithelial-interstitial microenvironment, the findings successfully reflected the PF disease, where excessive alpha smooth muscle actin (α-SMA) and collagen type I secretion were noted along with the morphological changes in response to BLM. Moreover, increased levels of fibrotic linked COL13A1, MMP2, WNT3 and decreased expression level of CDH1 provide evidence for the model reliability on fibrosis modelling. Subsequent administration of the FDA approved nintedanib and pirfenidone anti-fibrotic drugs proved the drug-responsiveness of the model.
Collapse
Affiliation(s)
- Ecem Saygili
- Department of Bioengineering, Ege University, Department of Bioengineering, Bornova, Izmir, 35040, TURKEY
| | - Utku Devamoglu
- Department of Bioengineering, Ege University, Department of Bioengineering, Bornova, Izmir, 35040, TURKEY
| | - Bakiye Goker-Bagca
- Department of Medical Biology, Adnan Menderes University, Department of Medical Biology, Aydin, Aydin, 09010, TURKEY
| | - Ozlem Goksel
- Department of Pulmonary Medicine / EgeSAM-Ege University Translational Pulmonary Research Center, Ege University, Bornova, Izmir, 35040, TURKEY
| | - Cigir Biray-Avci
- Department of Medical Biology, Ege University, Bornova, Izmir, 35040, TURKEY
| | - Tuncay Goksel
- Department of Pulmonary Medicine / EgeSAM-Ege University Translational Pulmonary Research Center, Ege University, Bornova, Izmir, 35040, TURKEY
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering / EgeSAM-Ege University Translational Pulmonary Research Center, Ege University, Bornova, Izmir, 35040, TURKEY
| |
Collapse
|
5
|
Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30:210093. [PMID: 34615700 PMCID: PMC9489085 DOI: 10.1183/16000617.0093-2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
First described in the mid-1960s, acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure with an overall mortality rate of approximately 40%. Despite significant advances in the understanding and treatment of ARDS, no substantive pharmacologic therapy has proven to be beneficial, and current management continues to be primarily supportive. Beyond their antibacterial activity, several antibiotics such as macrolides and tetracyclines exert pleiotropic immunomodulatory effects that might be able to rectify the dysregulated inflammatory response present in patients with ARDS. This review aims to provide an overview of preclinical and clinical studies that describe the immunomodulatory effects of antibiotics in ARDS. Moreover, the underlying mechanisms of their immunomodulatory properties will be discussed. Further studies are necessary to investigate their full therapeutic potential and to identify ARDS phenotypes which are most likely to benefit from their immunomodulatory effects.
Collapse
Affiliation(s)
- Andrea Sauer
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Konrad Peukert
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Inomata M, Kamio K, Azuma A, Matsuda K, Usuki J, Morinaga A, Tanaka T, Kashiwada T, Atsumi K, Hayashi H, Fujita K, Saito Y, Kubota K, Seike M, Gemma A. Rictor-targeting exosomal microRNA-16 ameliorates lung fibrosis by inhibiting the mTORC2-SPARC axis. Exp Cell Res 2020; 398:112416. [PMID: 33307020 DOI: 10.1016/j.yexcr.2020.112416] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 10/30/2020] [Accepted: 11/26/2020] [Indexed: 12/31/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF), a progressive disorder of unknown etiology, is characterized by pathological lung fibroblast activation and proliferation resulting in abnormal deposition of extracellular matrix proteins within the lung parenchyma. The pathophysiological roles of exosomal microRNAs in pulmonary fibrosis remain unclear; therefore, we aimed to identify and characterize fibrosis-responsive exosomal microRNAs. We used microRNA array analysis and profiled the expression of exosome-derived miRNA in sera of C57BL/6 mice exhibiting bleomycin-induced pulmonary fibrosis. The effect of microRNAs potentially involved in fibrosis was then evaluated in vivo and in vitro. The expression of exosomal microRNA-16 was increased by up to 8.0-fold on day 14 in bleomycin-treated mice, compared to vehicle-treated mice. MicroRNA-16 mimic administration on day 14 after bleomycin challenge ameliorated pulmonary fibrosis and suppressed lung and serum expression of secreted protein acidic and rich in cysteine (SPARC). Pretreatment of human lung fibroblasts with the microRNA-16 mimic decreased the expression of rapamycin-insensitive companion of mTOR (Rictor) and TGF-β1-induced expression of SPARC. This is the first study reporting the anti-fibrotic properties of microRNA-16 and demonstrating that these effects occur via the mTORC2 pathway. These findings support that microRNA-16 may be a promising therapeutic target for IPF.
Collapse
Affiliation(s)
- Minoru Inomata
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Koichiro Kamio
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan.
| | - Arata Azuma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Kuniko Matsuda
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Jiro Usuki
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Akemi Morinaga
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Toru Tanaka
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Takeru Kashiwada
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Kenichiro Atsumi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Hiroki Hayashi
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Kazue Fujita
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Yoshinobu Saito
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Kaoru Kubota
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, 113-8603, Japan
| |
Collapse
|
7
|
Antifibrotic therapy by sustained release of low molecular weight heparin from poly(lactic-co-glycolic acid) microparticles on bleomycin-induced pulmonary fibrosis in mice. Sci Rep 2020; 10:19019. [PMID: 33149192 PMCID: PMC7642430 DOI: 10.1038/s41598-020-76034-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/20/2020] [Indexed: 12/01/2022] Open
Abstract
Heparin and low molecular weight heparin (LMWH) have recently been considered useful treatment tools for inflammation. Heparin has antifibrotic activity, mediated by cellular secretion of hepatocyte growth factor (HGF). HGF has antifibrotic properties demonstrated in experimental models of lung, kidney, heart, skin, and liver fibrosis. The ability of LMWH for HGF secretion is similar to that of normal heparin. Poly (lactic-co-glycolic acid) (PLGA) is widely used for sustained drug release, because of its biocompatibility and low toxicity. LMWH-loaded PLGA microparticles are prepared by a conventional water-in-oil-in-water emulsion method. Interstitial pneumonia is a life-threatening pathological condition that causes respiratory failure when it progresses. In the present study, we investigated the therapeutic effect of LMWH-loaded PLGA microparticles in a mouse model of bleomycin-induced lung fibrosis. The ratios of fibrotic area to total area were significantly lower in mice administered LMWH-loaded microparticles than in mice administered bleomycin alone. The microparticle administration did not further enhance the gene expression for inflammatory cytokines. In a cell culture study, HGF secretion by mouse and human lung fibroblasts was significantly increased by LMWH addition. We conclude that LMWH showed anti-inflammatory activity, through the effects of LMWH-loaded PLGA microparticles on cells at sites of inflammation.
Collapse
|
8
|
Gastrin-releasing peptide induces fibrotic response in MRC5s and proliferation in A549s. Cell Commun Signal 2020; 18:96. [PMID: 32552754 PMCID: PMC7301567 DOI: 10.1186/s12964-020-00585-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 04/27/2020] [Indexed: 01/08/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a complex lung disease, whose build-up scar tissue is induced by several molecules. Gastrin-releasing peptide (GRP) is released from pulmonary neuroendocrine cells, alveolar macrophages, and some nerve endings in the lung. A possible role of GRP in IPF is unclear. We aimed to investigate the fibrotic response to GRP, at the cellular level in MRC5 and A549 cell lines. The proliferative and fibrotic effects of GRP on these cells were evaluated by using BrdU, immunoblotting, immunofluorescence and qRT-PCR for molecules associated with myofibroblast differentiation, TGF-β and Wnt signalling. All doses of GRP increased the amount of BrdU incorporation in A549 cells. In contrast, the amount of BrdU increased in MRC5 cells in the first 24 h, though progressively decreased by 72 h. GRP did not stimulate epithelial-mesenchymal transition in A549 cells, rather, it stimulated the differentiation of MRC5 cells into myofibroblasts. Furthermore, GRP induced gene and protein expressions of p-Smad2/3 and Smad4, and reduced the levels of Smad7 in MRC5 cells. In addition, GRP decreased Wnt5a protein levels and stimulated β-catenin activation by increasing Wnt4, Wnt7a and β-catenin protein levels. GRP caused myofibroblast differentiation by inducing TGF-βand Wnt pathways via paracrine and autocrine signalling in MRC5 cells. In conclusion, GRP may lead to pulmonary fibrosis due to its proliferative and fibrotic effects on lung fibroblasts. The abrogation of GRP-mediated signal activation might be considered as a treatment modality for fibrotic lung diseases. Video Abstract.
Collapse
|
9
|
Kim YS, Li Q, Youn HY, Kim DY. Oral Administration of Chitosan Attenuates Bleomycin-induced Pulmonary Fibrosis in Rats. In Vivo 2020; 33:1455-1461. [PMID: 31471392 DOI: 10.21873/invivo.11624] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIM Idiopathic pulmonary fibrosis (PF) is a fatal disorder of unknown aetiology with limited treatment options. Chitosan has antibacterial, antifungal, antioxidant, antitumour, and anti-inflammatory effects. This study aimed to investigate the effects of chitosan administration on bleomycin (BLM)-induced PF in rats. MATERIALS AND METHODS A PF rat model was established by endotracheal instillation of 5 mg/kg BLM; then, chitosan was administered in drinking water for 3 weeks. Histology, cell counts, and cytokine responses in the bronchoalveolar lavage fluid (BALF) and weight measurements (body and lung) were analyzed to assess its therapeutic effects. RESULTS Chitosan administration tended to reduce transforming growth factor (TGF)-β1 and interferon (IFN)-γ levels in BALF, and histopathological examination confirmed that chitosan attenuated the degree of inflammation and fibrosis in the lung. CONCLUSION This study revealed that oral chitosan exhibits potential antifibrotic effects, as measured by decreased proinflammatory cytokine levels and histological evaluation, in a BLM-induced PF rat model.
Collapse
Affiliation(s)
- You-Seok Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,KPC Corporation, Oporo, Gwangju, Republic of Korea
| | - Qiang Li
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dae Young Kim
- Department of Life Science, College of Bio-nano Technology, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
10
|
Krempaska K, Barnowski S, Gavini J, Hobi N, Ebener S, Simillion C, Stokes A, Schliep R, Knudsen L, Geiser TK, Funke-Chambour M. Azithromycin has enhanced effects on lung fibroblasts from idiopathic pulmonary fibrosis (IPF) patients compared to controls [corrected]. Respir Res 2020; 21:25. [PMID: 31941499 PMCID: PMC6964061 DOI: 10.1186/s12931-020-1275-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/01/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fatal lung disease without a cure and new drug strategies are urgently needed. Differences in behavior between diseased and healthy cells are well known and drug response can be different between cells isolated from IPF patients and controls. The macrolide Azithromycin (AZT) has anti-inflammatory and immunomodulatory properties. Recently anti-fibrotic effects have been described. However, the anti-fibrotic effects on primary IPF-fibroblasts (FB) directly compared to control-FB are unknown. We hypothesized that IPF-FB react differently to AZT in terms of anti-fibrotic effects. METHODS Primary normal human lung and IPF-FB were exposed to TGF-β (5 ng/ml), Azithromycin (50 μM) alone or in combination prior to gene expression analysis. Pro-collagen Iα1 secretion was assessed by ELISA and protein expression by western blot (αSMA, Fibronectin, ATP6V1B2, LC3 AB (II/I), p62, Bcl-xL). Microarray analysis was performed to screen involved genes and pathways after Azithromycin treatment in control-FB. Apoptosis and intraluminal lysosomal pH were analyzed by flow cytometry. RESULTS AZT significantly reduced collagen secretion in TGF-β treated IPF-FB compared to TGF-β treatment alone, but not in control-FB. Pro-fibrotic gene expression was similarly reduced after AZT treatment in IPF and control-FB. P62 and LC3II/I western blot revealed impaired autophagic flux after AZT in both control and IPF-FB with significant increase of LC3II/I after AZT in control and IPF-FB, indicating enhanced autophagy inhibition. Early apoptosis was significantly higher in TGF-β treated IPF-FB compared to controls after AZT. Microarray analysis of control-FB treated with AZT revealed impaired lysosomal pathways. The ATPase and lysosomal pH regulator ATP6V0D2 was significantly less increased after additional AZT in IPF-FB compared to controls. Lysosomal function was impaired in both IPF and control FB, but pH was significantly more increased in TGF-β treated IPF-FB. CONCLUSION We report different treatment responses after AZT with enhanced anti-fibrotic and pro-apoptotic effects in IPF compared to control-FB. Possibly impaired lysosomal function contributes towards these effects. In summary, different baseline cell phenotype and behavior of IPF and control cells contribute to enhanced anti-fibrotic and pro-apoptotic effects in IPF-FB after AZT treatment and strengthen its role as a new potential anti-fibrotic compound, that should further be evaluated in clinical studies.
Collapse
Affiliation(s)
- Kristina Krempaska
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Sandra Barnowski
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Jacopo Gavini
- Department of Visceral Surgery and Medicine, Department for BioMedical Research, Inselspital, Bern University Hospital and University of Bern, 3010, Bern, Switzerland
| | - Nina Hobi
- AlveoliX AG, Murtenstrasse 50, 3008, Bern, Switzerland
- ARTORG Center for Biomedical Engineering Research, Organs-on-Chip Technologies, University of Bern, Bern, Switzerland
| | - Simone Ebener
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Cedric Simillion
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bioinformatics Unit and SIB Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Andrea Stokes
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ronja Schliep
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Thomas K Geiser
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, CH-3010, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
11
|
Experimental Models of Pulmonary Fibrosis and their Translational Potential. ACTA MEDICA MARTINIANA 2019. [DOI: 10.2478/acm-2019-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Pulmonary fibrosis, represented mainly by idiopathic pulmonary fibrosis, develops chronic and progressive changes in lung parenchyma with high mortality and limited therapeutic options. The aim of this review was to summarize the most common experimental models used in the research of pulmonary fibrosis. Lung damage associated with development of pulmonary fibrosis can be caused by irradiation or by instillation of bleomycin, fluorescein isothiocyanate (FITC), silicon dioxide (silica), asbestos, etc. This article reviews the characteristics of the most frequently used animal models of fibrosis, including the limitations of their use. Although none of the used animal models resembles completely the changes in human pulmonary fibrosis, similarities between them allow preclinical testing of novel treatment approaches or their combinations in the laboratory conditions before their use in the clinical practice.
Collapse
|
12
|
Chen H, Yang R, Tang Y, Fu X. Effects of curcumin on artery blood gas index of rats with pulmonary fibrosis caused by paraquat poisoning and the expression of Smad 4, Smurf 2, interleukin-4 and interferon-γ. Exp Ther Med 2019; 17:3664-3670. [PMID: 30988750 PMCID: PMC6447781 DOI: 10.3892/etm.2019.7341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Effects of curcumin on artery blood gas index of rats with pulmonary fibrosis caused by paraquat poisoning and the expression of Mothers against decapentaplegic homolog 4 (Smad4), Smad ubiquitination regulatory factor 2 (Smurf2), interleukin-4 (IL-4) and interferon-γ (IFN-γ) were explored. A total of 54 Wistar rats were randomly selected, of which 36 rats were selected for paraquat poisoning pulmonary fibrosis modeling, and 18 were used in the control group for normal feeding. Then, 18 rats were randomly selected from the modeled groups and injected with curcumin and classified as the curcumin group. The remaining 18 rats were not processed and 17 were successfully modeled as the paraquat group. The expression of SMAD4, Smurf2, IL-4 and INF-γ was detected by enzyme-linked immunosorbent assay. Abdominal aortic blood was taken for determination of pH, arterial partial pressure of oxygen (PaO2) and arterial partial pressure of carbon dioxide (PaCO2). The artery blood PaO2 and serum INF-γ of the curcumin and paraquat groups were significantly higher on day 1 than those on day 5 (P<0.05). The artery blood PaO2 and serum INF-γ in the curcumin group were higher than those in the paraquat group (P<0.05). The artery blood PaCO2, serum Smad4, Smurf2 and IL-4 in the curcumin group were significantly lower on day 1 than those on day 5 (P<0.05). The artery blood PaCO2, serum Smad4, Smurf2 and IL-4 in the paraquat group were significantly lower on day 1 than those on day 5 (P<0.05). The PaCO2, serum Smad4, Smurf2 and IL-4 in the curcumin group were lower than those in the paraquat group (P<0.05). In conclusion, curcumin can effectively improve pulmonary fibrosis in rats after treatment with paraquat poisoning. The results show that it is expected to be an effective drug for the treatment of paraquat, and provide effective reference and guidance for clinical treatment.
Collapse
Affiliation(s)
- Honggang Chen
- Department of Emergency, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Rongjia Yang
- Department of Emergency, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yan Tang
- Department of Emergency, The Second People's Hospital of Lanzhou City, Lanzhou, Gansu 730000, P.R. China
| | - Xiaoyan Fu
- Department of Nursing, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
13
|
Jin YK, Li XH, Wang W, Liu J, Zhang W, Fang YS, Zhang ZF, Dai HP, Ning W, Wang C. Follistatin-Like 1 Promotes Bleomycin-Induced Pulmonary Fibrosis through the Transforming Growth Factor Beta 1/Mitogen-Activated Protein Kinase Signaling Pathway. Chin Med J (Engl) 2018; 131:1917-1925. [PMID: 30082522 PMCID: PMC6085847 DOI: 10.4103/0366-6999.238151] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Follistatin-like 1 (FSTL1) is a novel profibrogenic factor that induces pulmonary fibrosis (PF) through the transforming growth factor-beta 1 (TGF-β1)/Smad signaling. Little is known about its effects on PF through the non-Smad signaling, like the mitogen-activated protein kinase (MAPK) pathway. Therefore, this study aimed to investigate the role of FSTL1 in PF through the MAPK signaling pathway and its mechanisms in lung fibrogenesis. Methods PF was induced in Fstl1+/-and wild-type (WT) C57BL/6 mice with bleomycin. After 14 days, the mice were sacrificed, and lung tissues were stained with hematoxylin and eosin; the hydroxyproline content was measured to confirm PF. The mRNA and protein level of FSTL1 and the change of MAPK phosphorylation were measured by quantitative polymerase chain reaction and Western blotting. The effect of Fstl1 deficiency on fibroblasts differentiation was measured by Western blotting and cell immunofluorescence. MAPK signaling activation was measured by Western blotting in Fstl1+/- and WT fibroblasts treated with recombinant human FSTL1 protein. We pretreated mouse lung fibroblast cells with inhibitors of the extracellular signal-regulated kinase (ERK), p38, and Jun N-terminal kinase (JNK) signaling and analyzed their differentiation, proliferation, migration, and invasion by Western blotting, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide analysis, and transwell assays. The Student's t-test was used to compare the differences between two groups. Results Fstl1 deficiency attenuated phosphorylation of the ERK, p38, and JNK signaling in bleomycin-induced fibrotic lung tissue 14 days after injury (0.67 ± 0.05 vs. 1.22 ± 0.03, t = 14.92, P = 0.0001; 0.41 ± 0.01 vs. 1.15 ± 0.07; t = 11.19; P = 0.0004; and 0.41 ± 0.01 vs. 1.07 ± 0.07, t = 8.92, P = 0.0009; respectively), compared with WT lungs at the same time and in primary lung fibroblasts (0.82 ± 0.01 vs. 1.01 ± 0.04, t = 4.06, P = 0.0150; 1.04 ± 0.03 vs. 1.24 ± 0.03, t = 4.44, P = 0.0100; and 0.76 ± 0.05 vs. 0.99 ± 0.05, t = 4.48, P = 0.0100; respectively), compared with TGF-β1-stimulated WT group. Recombinant human FSTL1 protein in lung fibroblasts enhanced TGF-β1-mediated phosphorylation of the ERK (1.19 ± 0.08 vs. 0.55 ± 0.04, t = 6.99, P = 0.0020), p38 (1.18 ± 0.04 vs. 0.66 ± 0.03, t = 11.20, P = 0.0020), and JNK (1.11 ± 0.01 vs. 0.84 ± 0.04, t = 6.53, P = 0.0030), compared with the TGF-β1-stimulated WT group. Fstl1-deficient fibroblasts showed reduced alpha-smooth muscle actin (α-SMA) expression (0.70 ± 0.06 vs. 1.28 ± 0.11, t = 4.65, P = 0.0035, compared with the untreated WT group; 1.40 ± 0.05 vs. 1.76 ± 0.02, t = 6.31, P = 0.0007; compared with the TGF-β1-treated WT group). Compared with the corresponding condition in the control group, the TGF-β1/FSTL1-mediated α-SMA expression was significantly suppressed by pretreatment with an inhibitor of p38 (0.73 ± 0.01 vs. 1.13 ± 0.10, t = 3.92, P = 0.0078) and JNK (0.78 ± 0.03 vs. 1.08 ± 0.06, t = 4.40, P = 0.0046) signaling. The proliferation of mouse lung fibroblast cells (MLgs) significantly decreased after treatment of an inhibitor of p38 (0.30 ± 0.01 vs. 0.46 ± 0.03, t = 4.64, P = 0.0009), JNK (0.30 ± 0.01 vs. 0.49 ± 0.01, t = 12.84, P = 0.0001), and Smad2/3 (0.18 ± 0.02 vs. 0.46 ± 0.02, t = 12.69, P = 0.0001) signaling compared with the dimethylsulfoxide group. The migration and invasion cells of MLgs significantly decreased in medium pretreated with an inhibitor of p38 (70.17 ± 3.28 vs. 116.30 ± 7.11, t = 5.89, P = 0.0042 for the migratory cells; 19.87 ± 0.84 vs. 32.70 ± 0.95, t = 10.14, P = 0.0005 for the invasive cells), JNK (72.30 ± 3.85 vs. 116.30 ± 7.11, t = 5.44, P = 0.0056 for the migratory cells; 18.03 ± 0.94 vs. 32.70 ± 0.95, t = 11.00, P = 0.0004 for the invasive cells), and Smad2/3 (64.76 ± 1.41 vs. 116.30 ± 7.11, t = 7.11, P = 0.0021 for the migratory cells; 18.03 ± 0.94 vs. 32.70 ± 0.95, t = 13.29, P = 0.0002 for the invasive cells) signaling compared with the corresponding condition in the dimethylsulfoxide group. Conclusion FSTL1 affects lung fibroblast differentiation, proliferation, migration, and invasion through p38 and JNK signaling, and in this way, it might influence the development of PF.
Collapse
Affiliation(s)
- Yan-Kun Jin
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University; Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| | - Xiao-He Li
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wang Wang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Jie Liu
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Wei Zhang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Yin-Shan Fang
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhi-Fei Zhang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Physiology and Pathophysiology, Capital Medical University, Beijing 100069, China
| | - Hua-Ping Dai
- Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| | - Wen Ning
- Department of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chen Wang
- Department of Respiratory Disease, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Capital Medical University; Department of Respiratory Disease, Capital Medical University, Beijing 100020, China
| |
Collapse
|
14
|
Carrington R, Jordan S, Pitchford S, Page C. Use of animal models in IPF research. Pulm Pharmacol Ther 2018; 51:73-78. [DOI: 10.1016/j.pupt.2018.07.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/04/2018] [Accepted: 07/05/2018] [Indexed: 01/10/2023]
|
15
|
Zhang Z, Zhao Y, Chen G, Li R, Yang J, Sun D. Study of lung toxicity in rats exposed to silica powder with different hard metal constituents. Toxicol Ind Health 2018; 34:449-457. [PMID: 29669483 DOI: 10.1177/0748233718758586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The objective of this study was to assess the lung toxicity induced by the inhalation of different hard metal constituents and silica powder and screen for potential toxicity biomarkers. Rats were randomly divided into saline, cobalt, tungsten carbide, silica, and hard metal (HM) groups and were administered a single 10-mg dose of the respective treatments. After 8 weeks, the lung tissue structure in the HM group was deformed, numerous nucleated giant and epithelial-like cells appeared in the stroma, and the computed tomography scanning images appeared abnormal. Krebs von den Lungen-6 (KL-6), transforming growth factor (TGF)-β1, and TGF-β2 expression in bronchoalveolar lavage fluid (BALF) significantly differed between the groups ( p < 0.05). Serum KL-6 and TGF-β1, but not TGF-β2, levels significantly differed between some groups ( p < 0.05). We observed multinucleated giant cells in the rat lung tissue. While the serum and BALF levels of KL-6 and TGF-β2 are not highly specific, TGF-β1 may be a valuable reference diagnostic marker in HM lung disease.
Collapse
Affiliation(s)
- Zhansai Zhang
- 1 Department of Occupational Disease, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanfang Zhao
- 2 Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Gang Chen
- 3 Department of Orthopedics, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruoming Li
- 4 Department of Public Health, Lingyun Community Health Service Center, Shanghai, China
| | - Jun Yang
- 5 Department of Spine Surgery, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Daoyuan Sun
- 1 Department of Occupational Disease, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
The Nonantibiotic Macrolide EM703 Improves Survival in a Model of Quinolone-Treated Pseudomonas aeruginosa Airway Infection. Antimicrob Agents Chemother 2017; 61:AAC.02761-16. [PMID: 28652240 DOI: 10.1128/aac.02761-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 06/16/2017] [Indexed: 11/20/2022] Open
Abstract
Macrolide antibiotics are used as anti-inflammatory agents, e.g., for prevention of exacerbations in chronic obstructive pulmonary disease and cystic fibrosis. Several studies have shown improved outcomes after the addition of macrolides to β-lactam antibiotics for treatment of severe community-acquired pneumonia. However, a beneficial effect of macrolides in treating Gram-negative bacterial airway infections, e.g., those caused by Pseudomonas aeruginosa, remains to be shown. Macrolide antibiotics have significant side effects, in particular, motility-stimulating activity in the gastrointestinal tract and promotion of bacterial resistance. In this study, EM703, a modified macrolide lacking antibiotic and motility-stimulating activities but with retained anti-inflammatory properties, was used as an adjunct treatment for experimental P. aeruginosa lung infection, in combination with a conventional antibiotic. Airway infections in BALB/cJRj mice were induced by nasal instillation of P. aeruginosa; this was followed by treatment with the quinolone levofloxacin in the absence or presence of EM703. Survival, inflammatory responses, and cellular influx to the airways were monitored. Both pretreatment and simultaneous administration of EM703 dramatically improved survival in levofloxacin-treated mice with P. aeruginosa airway infections. In addition, EM703 reduced the levels of proinflammatory cytokines, increased the numbers of leukocytes in bronchoalveolar lavage fluid, and reduced the numbers of neutrophils present in lung tissue. In summary, the findings of this study show that the immunomodulatory properties of the modified macrolide EM703 can be important when treating Gram-negative pneumonia, as exemplified by P. aeruginosa infection in this study.
Collapse
|
17
|
XPLN is modulated by HDAC inhibitors and negatively regulates SPARC expression by targeting mTORC2 in human lung fibroblasts. Pulm Pharmacol Ther 2017; 44:61-69. [DOI: 10.1016/j.pupt.2017.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/12/2017] [Indexed: 11/19/2022]
|
18
|
Nozoe K, Aida Y, Fukuda T, Sanui T, Nishimura F. Mechanisms of the Macrolide-Induced Inhibition of Superoxide Generation by Neutrophils. Inflammation 2017; 39:1039-48. [PMID: 26983705 DOI: 10.1007/s10753-016-0333-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The effect of macrolides on the superoxide (O2 (-)) production by neutrophils was studied. Resting neutrophils become primed by lipopolysaccharide (LPS) or N-formyl-methionyl-leucyl-phenylalanine (fMLP), and primed neutrophils generate O2 (-) in response to fMLP or adhesion, respectively. Both LPS-primed fMLP-stimulated O2 (-) generation by macrolide-treated neutrophils and adhesion-stimulated O2 (-) generation by macrolide-treated fMLP-primed neutrophils were inhibited. Macrolide inhibition of O2 (-) generation was dependent on serum or pH. Serum could be substituted by NaHCO3. The intensity of inhibition was azithromycin = roxithromycin > clarithromycin > erythromycin, in that order. Non-antimicrobial derivatives of erythromycin, that is, EM703 and EM900, inhibited O2 (-) generation at pH 7.4. NH4Cl abolished the activity of azithromycin (AZ) only when added to neutrophils with AZ but not after incubation with AZ, suggesting that NH4Cl prevented the influx of AZ. AZ did not affect the expression of alkaline phosphatase, CD11b, and cytochrome b558 in both resting and LPS-primed neutrophils. These results suggested that macrolides did not affect granule mobilization but inhibited O2 (-) generation selectively.
Collapse
Affiliation(s)
- Kohji Nozoe
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshitomi Aida
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takao Fukuda
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Terukazu Sanui
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Fusanori Nishimura
- Section of Periodontology, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
19
|
Chang XH, Zhu A, Liu FF, Zou LY, Su L, Liu SK, Zhou HH, Sun YY, Han AJ, Sun YF, Li S, Li J, Sun YB. Nickel oxide nanoparticles induced pulmonary fibrosis via TGF-β1 activation in rats. Hum Exp Toxicol 2016; 36:802-812. [DOI: 10.1177/0960327116666650] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nano nickel oxide (NiO), widely used in industry, has recently been discovered to have pulmonary toxicity. However, no subchronic exposure studies about nano NiO-induced pulmonary fibrosis have been reported. The objective of this study was to investigate pulmonary fibrosis induced by nano NiO and its potential mechanism in rats. Male Wistar rats ( n = 40, 200–240 g) were randomized into control group, nano NiO groups (0.015, 0.06, and 0.24 mg/kg), and micro NiO group (0.024 mg/kg). All rats were killed to collect lung tissue after intratracheal instillation of NiO particles twice a week for 6 weeks. To identify pulmonary fibrosis, Masson trichrome staining, hydroxyproline content, and collagen protein expression were performed. The results showed widespread lung fibrotic injury in histological examination and increased content of hydroxyproline, collagen types I and III in rat lung tissue exposed to nano NiO. To explore the potential pulmonary fibrosis mechanism, transforming growth factor beta 1 (TGF- β1) content was measured by enzyme-linked immunosorbent assay, and the messenger RNA expression of key indicators was detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The TGF- β1 content was increased in nano NiO exposure groups, as well as the upregulated gene expression of TGF- β1, Smad2, Smad4, matrix metalloproteinase, and tissue inhibitor of metalloproteinase. The findings indicated that nano NiO could induce pulmonary fibrosis, which may be related to TGF- β1 activation.
Collapse
Affiliation(s)
- XH Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - A Zhu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - FF Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - LY Zou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - L Su
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - SK Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - HH Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - YY Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - AJ Han
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - YF Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - S Li
- Lanzhou Municipal Center for Disease Control, Lanzhou, China
| | - J Li
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - YB Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
20
|
Togami K, Miyao A, Miyakoshi K, Kanehira Y, Tada H, Chono S. Efficient delivery to human lung fibroblasts (WI-38) of pirfenidone incorporated into liposomes modified with truncated basic fibroblast growth factor and its inhibitory effect on collagen synthesis in idiopathic pulmonary fibrosis. Biol Pharm Bull 2015; 38:270-6. [PMID: 25747986 DOI: 10.1248/bpb.b14-00659] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In the present in vitro study, we assessed the delivery of pirfenidone incorporated into liposomes modified with truncated basic fibroblast growth factor (tbFGF) to lung fibroblasts and investigated the anti-fibrotic effect of the drug. The tbFGF peptide, KRTGQYKLC, was used to modify the surface of liposomes (tbFGF-liposomes). We used the thin-layer evaporation method, followed by sonication, to prepare tbFGF-liposomes containing pirfenidone. The cellular accumulation of tbFGF-liposomes was 1.7-fold greater than that of non-modified liposomes in WI-38 cells used as a model of lung fibroblasts. Confocal laser scanning microscopy showed that tbFGF-liposomes were widely localized in WI-38 cells. The inhibitory effects of pirfenidone incorporated into tbFGF-liposomes on transforming growth factor-β1 (TGF-β1)-induced collagen synthesis in WI-38 cells were evaluated by measuring the level of intracellular hydroxyproline, a major component of the protein collagen. Pirfenidone incorporated into tbFGF-liposomes at concentrations of 10, 30, and 100 µM significantly decreased the TGF-β1-induced hydroxyproline content in WI-38 cells. The anti-fibrotic effect of pirfenidone incorporated into tbFGF-liposomes was enhanced compared with that of pirfenidone solution. These results indicate that tbFGF-liposomes are a useful drug delivery system of anti-fibrotic drugs to lung fibroblasts for the treatment of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Kohei Togami
- Division of Pharmaceutics, Hokkaido Pharmaceutical University School of Pharmacy
| | | | | | | | | | | |
Collapse
|
21
|
Non-antibiotic 12-membered macrolides: design, synthesis and biological evaluation in a cigarette-smoking model. J Antibiot (Tokyo) 2015; 69:319-26. [PMID: 26419414 DOI: 10.1038/ja.2015.91] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/09/2015] [Accepted: 08/10/2015] [Indexed: 12/18/2022]
Abstract
The 14-membered macrolide erythromycin A expresses three distinct biological properties, including antibacterial activity, gastrointestinal motor-stimulating activity and anti-inflammatory and/or immunomodulatory effects. Although low-dose, long-term therapy using 14- and 15-membered macrolides displaying anti-inflammatory and/or immunomodulatory activity effectively treats diffuse panbronchiolitis and chronic sinusitis, bacterial resistance may emerge. To address this issue, we developed the 12-membered non-antibiotic macrolide (8R,9S)-8,9-dihydro-6,9-epoxy-8,9-anhydropseudoerythromycin A (EM900) that promotes monocyte to macrophage differentiation, a marker for anti-inflammatory and/or immunomodulatory effects, without possessing antibacterial activity. In this article, we report that the new macrolide derivative (8R,9S) -de(3'-N-methyl)-3'-N-(p-chlorobenzyl)-de(3-O-cladinosyl)-3-dehydro-8,9-dihydro-6,9-epoxy-8,9-anhydropseudoerythromycin A 12,13-carbonate (EM939) exhibited stronger promotive activity for monocyte to macrophage differentiation than that of the parent compound EM900 in addition to reduced cytotoxicity toward THP-1 cells and antibacterial inactivity. In a cigarette-smoking model used to simulate chronic obstructive pulmonary disease (COPD), the EM900 derivatives significantly attenuated lung and alveolar inflations, functionally and histologically, via oral administration. Because of these marked therapeutic effects, non-antibiotic EM900 derivatives may become central to the treatment of chronic inflammatory diseases such as COPD.
Collapse
|
22
|
Nintedanib modulates surfactant protein-D expression in A549 human lung epithelial cells via the c-Jun N-terminal kinase-activator protein-1 pathway. Pulm Pharmacol Ther 2015; 32:29-36. [PMID: 25843005 DOI: 10.1016/j.pupt.2015.03.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/17/2015] [Accepted: 03/24/2015] [Indexed: 11/21/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease with a high mortality rate. Signalling pathways activated by several tyrosine kinase receptors are known to be involved in lung fibrosis, and this knowledge has led to the development of the triple tyrosine kinase inhibitor nintedanib, an inhibitor of vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and fibroblast growth factor receptor (FGFR), for the treatment of IPF. Pulmonary surfactant protein D (SP-D), an important biomarker of IPF, reportedly attenuates bleomycin-induced pulmonary fibrosis in mice. In this study, we investigated whether nintedanib modulates SP-D expression in human lung epithelial (A549) cells using quantitative real-time reverse transcriptase polymerase chain reaction and western blotting. To investigate the mechanisms underlying the effects of nintedanib, we evaluated the phosphorylation of c-Jun N-terminal kinase (JNK) and its downstream target c-Jun. The effect of the JNK inhibitor SP600125 on c-Jun phosphorylation was also tested. Activation of activator protein-1 (AP-1) was examined using an enzyme-linked immunosorbent assay-based test, and cell proliferation assays were performed to estimate the effect of nintedanib on cell proliferation. Furthermore, we treated mice with nintedanib to examine its in vivo effect on SP-D levels in lungs. These experiments showed that nintedanib up-regulated SP-D messenger RNA expression in a dose-dependent manner at concentrations up to 5 μM, with significant SP-D induction observed at concentrations of 3 μM and 5 μM, in comparison with that observed in vehicle controls. Nintedanib stimulated a rapid increase in phosphorylated JNK in A549 cells within 30 min of treatment and stimulated c-Jun phosphorylation, which was inhibited by the JNK inhibitor SP600125. Additionally, nintedanib was found to activate AP-1. A549 cell proliferation was not affected by nintedanib at any of the tested concentrations. Moreover, blocking FGFR, PDGFR, and VEGFR function did not affect nintedanib-induced SP-D expression, suggesting that nintedanib mediates its effects through a mechanism that is distinct from its known role as a tyrosine kinase inhibitor. Nintedanib is also reported to inhibit Src kinase although pre-treatment of cells with a Src kinase inhibitor had no effect on nintedanib-induced SP-D expression. Increased expression of SFTPD mRNA and SP-D protein in the lungs of nintedanib-treated mice was also observed. In this work, we demonstrated that nintedanib up-regulated SP-D expression in A549 cells via the JNK-AP-1 pathway and did not affect cell proliferation. This is the first report describing SP-D induction by nintedanib.
Collapse
|
23
|
The design of novel classes of macrolides for neutrophil-dominated inflammatory diseases. Future Med Chem 2015; 6:657-74. [PMID: 24895894 DOI: 10.4155/fmc.14.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neutrophil-dominated inflammatory diseases, like chronic obstructive pulmonary disease, cystic fibrosis, bronchiectasis, bronchiolitis obliteras syndrome and non-eosinophilic asthma, present a significant medical problem lacking adequate therapy. Macrolide antibiotics have been reported to be effective in the treatment of the aforementioned diseases, for reasons unrelated to their antibacterial action. This has resulted in research activities aimed at gaining a better understanding of the immunomodulatory actions of macrolides and the synthesis of various novel anti-inflammatory macrolides without antimicrobial activity. Despite the difficult chemistry and lack of an extensive knowledge for their mechanism of action, several interesting molecules from this class, including potential clinical candidates, are on the horizon.
Collapse
|
24
|
Do genetic susceptibility, Toll-like receptors, and pathogen-associated molecular patterns modulate the effects of wear? Clin Orthop Relat Res 2014; 472:3709-17. [PMID: 25034980 PMCID: PMC4397765 DOI: 10.1007/s11999-014-3786-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Overwhelming evidence supports the concept that wear particles are the primary initiator of aseptic loosening of orthopaedic implants. It is likely, however, that other factors modulate the biologic response to wear particles. This review focuses on three potential other factors: genetic susceptibility, Toll-like receptors (TLRs), and bacterial pathogen-associated molecular patterns (PAMPs). WHERE ARE WE NOW?: Considerable evidence is emerging that both genetic susceptibility and TLR activation are important factors that modulate the biologic response to wear particles, but it remains controversial whether bacterial PAMPs also do so. WHERE DO WE NEED TO GO?: Detailed understanding of the roles of these other factors may lead to identification of novel therapeutic targets for patients with aseptic loosening. HOW DO WE GET THERE?: Highest priority should be given to polymorphism replication studies with large numbers of patients and studies to replicate the reported correlation between bacterial biofilms and the severity of aseptic loosening.
Collapse
|
25
|
Wang L, Sun Y, Ruan C, Liu B, Zhao L, Gu X. Angelica sinensis is effective in treating diffuse interstitial pulmonary fibrosis in rats. BIOTECHNOL BIOTEC EQ 2014; 28:923-928. [PMID: 26019579 PMCID: PMC4433952 DOI: 10.1080/13102818.2014.957487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 06/03/2014] [Indexed: 11/05/2022] Open
Abstract
The aim of this study was to investigate the therapeutic effect of Angelica sinensis on a rat model of diffuse interstitial pulmonary fibrosis induced by bleomycin A5. The mechanism by which A. sinensis exerts its effect is also discussed. A diffuse interstitial pulmonary fibrosis model was established in 36 male Wistar rats by an endotracheal injection of bleomycin A5 (5 mg/kg). Then, these rats were randomly divided into the model group (n = 18) and the treatment group (treated with A. sinensis after modelling, n = 18). Control rats (n = 6) received an equal volume of saline. Hematoxylin–eosin (HE) staining was performed to analyse alveolitis and Masson staining, to observe pulmonary fibrosis. Collagen content was determined by hydroxyproline assay. Nuclear factor kappa B (NF-κB) activity was measured by electrophoretic mobility shift assay. Transforming growth factor-β (TGF-β) expression at mRNA level was detected by northern blotting and at protein level by enzyme-linked immunosorbent assay. The results obtained showed that the alveolitis and pulmonary fibrosis of the rats treated with A. sinensis was significantly alleviated compared with that of the rats in the model group. Treatment with A. sinensis also lowered the content of collagen, decreased NF-κB activity in alveolar macrophages and reduced the TGF-β expression at the mRNA and protein level. These results indicated that A. sinensis is effective in treating and alleviating interstitial pulmonary fibrosis, possibly by lowering collagen, inhibiting the activity of NF-κB and reducing the TGF-β expression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Yanmei Sun
- Department of Internal Medicine, Qinghua Hospital , Xi'an , Shaanxi , P.R. China
| | - Cailian Ruan
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Bofeng Liu
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Lin Zhao
- Department of Human Anatomy, School of Medical Sciences, Yan'an University , Yan'an , Shaanxi , P.R. China
| | - Xiujuan Gu
- Department of Inspection Division, Yan'an University Affiliated Hospital , Yan'an , Shaanxi , P.R. China
| |
Collapse
|
26
|
Saputra D, Yoon JH, Park H, Heo Y, Yang H, Lee EJ, Lee S, Song CW, Lee K. Inhalation of carbon black nanoparticles aggravates pulmonary inflammation in mice. Toxicol Res 2014; 30:83-90. [PMID: 25071917 PMCID: PMC4112069 DOI: 10.5487/tr.2014.30.2.083] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/02/2014] [Accepted: 06/15/2014] [Indexed: 01/17/2023] Open
Abstract
An increasing number of recent studies have focused on the impact of particulate matter on human health. As a model for atmospheric particulate inhalation, we investigated the effects of inhaled carbon black nanoparticles (CBNP) on mice with bleomycin-induced pulmonary fibrosis. The CNBPs were generated by a novel aerosolization process, and the mice were exposed to the aerosol for 4 hours. We found that CBNP inhalation exacerbated lung inflammation, as evidenced by histopathology analysis and by the expression levels of interleukin-6 protein, fibronectin, and interferon-γ mRNAs in lung tissues. Notably, fibronectin mRNA expression showed a statistically significant increase in expression after CBNP exposure. These data suggest that the concentration of CBNPs delivered (calculated to be 12.5 μg/m(3)) can aggravate lung inflammation in mice. Our results also suggest that the inhalation of ultrafine particles like PM 2.5 is an impactful environmental risk factor for humans, particularly in susceptible populations with predisposing lung conditions.
Collapse
Affiliation(s)
- Devina Saputra
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea ; Toxicology and Pharmacology, Korea University of Science and Technology, Daejeon, Korea
| | - Jin-Ha Yoon
- Institute for Occupational Health, Yonsei University College of Medicine, Seoul, Korea
| | - Hyunju Park
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Yongju Heo
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Hyoseon Yang
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Eun Ji Lee
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Sangjin Lee
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Chang-Woo Song
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center, Korea Institute of Toxicology, Jeonbuk, Korea ; Toxicology and Pharmacology, Korea University of Science and Technology, Daejeon, Korea
| |
Collapse
|
27
|
Nyp MF, Navarro A, Rezaiekhaligh MH, Perez RE, Mabry SM, Ekekezie II. TRIP-1 via AKT modulation drives lung fibroblast/myofibroblast trans-differentiation. Respir Res 2014; 15:19. [PMID: 24528651 PMCID: PMC3946032 DOI: 10.1186/1465-9921-15-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 02/11/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Myofibroblasts are the critical effector cells in the pathogenesis of pulmonary fibrosis which carries a high degree of morbidity and mortality. We have previously identified Type II TGFβ receptor interacting protein 1 (TRIP-1), through proteomic analysis, as a key regulator of collagen contraction in primary human lung fibroblasts--a functional characteristic of myofibroblasts, and the last, but critical step in the process of fibrosis. However, whether or not TRIP-1 modulates fibroblast trans-differentiation to myofibroblasts is not known. METHODS TRIP-1 expression was altered in primary human lung fibroblasts by siRNA and plasmid transfection. Transfected fibroblasts were then analyzed for myofibroblast features and function such as α-SMA expression, collagen contraction ability, and resistance to apoptosis. RESULTS The down-regulation of TRIP-1 expression in primary human lung fibroblasts induces α-SMA expression and enhances resistance to apoptosis and collagen contraction ability. In contrast, TRIP-1 over-expression inhibits α-SMA expression. Remarkably, the effects of the loss of TRIP-1 are not abrogated by blockage of TGFβ ligand activation of the Smad3 pathway or by Smad3 knockdown. Rather, a TRIP-1 mediated enhancement of AKT phosphorylation is the implicated pathway. In TRIP-1 knockdown fibroblasts, AKT inhibition prevents α-SMA induction, and transfection with a constitutively active AKT construct drives collagen contraction and decreases apoptosis. CONCLUSIONS TRIP-1 regulates fibroblast acquisition of phenotype and function associated with myofibroblasts. The importance of this finding is it suggests TRIP-1 expression could be a potential target in therapeutic strategy aimed against pathological fibrosis.
Collapse
Affiliation(s)
- Michael F Nyp
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Angels Navarro
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Mohammad H Rezaiekhaligh
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Ricardo E Perez
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Sherry M Mabry
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Ikechukwu I Ekekezie
- Department of Pediatrics, Section of Neonatal-Perinatal Medicine, Children’s Mercy Hospitals & Clinics, 2401 Gillham Road, 64108 Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| |
Collapse
|
28
|
Inomata M, Kamio K, Azuma A, Matsuda K, Kokuho N, Miura Y, Hayashi H, Nei T, Fujita K, Saito Y, Gemma A. Pirfenidone inhibits fibrocyte accumulation in the lungs in bleomycin-induced murine pulmonary fibrosis. Respir Res 2014; 15:16. [PMID: 24507087 PMCID: PMC3930125 DOI: 10.1186/1465-9921-15-16] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 02/04/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bone marrow-derived fibrocytes reportedly play important roles in the pathogenesis of idiopathic pulmonary fibrosis. Pirfenidone is an anti-fibrotic agent; however, its effects on fibrocytes have not been investigated. The aim of this study was to investigate whether pirfenidone inhibits fibrocyte pool size in the lungs of bleomycin-treated mice. METHODS Bleomycin (100 mg/kg) was infused with osmotic pumps into C57BL/6 mice, and pirfenidone (300 mg/kg/day) was orally administered daily for 2 wk. The lungs were removed, and single-cell suspensions were subjected to fluorescence-activated cell sorter (FACS) analysis to detect fibrocytes, which were defined as CD45 and collagen-I double-positive cells. Immunohistochemistry was performed on the lung specimens to quantify fibrocytes. Chemokines in the lung digests were measured with enzyme-linked immunosorbent assay. The effect of pirfenidone on alveolar macrophages was evaluated with bronchoalveolar lavage (BAL). In a therapeutic setting, pirfenidone administration was initiated 10 days after bleomycin treatment. For chemotaxis assay, lung fibrocytes were isolated with immunomagnetic selection (CD45-positive mesenchymal cells) after culture and allowed to migrate toward chemokines in the presence or absence of pirfenidone. Moreover, the effect of pirfenidone on the expression of chemokine receptors on fibrocytes was evaluated. RESULTS Pirfenidone significantly ameliorated bleomycin-induced pulmonary fibrosis as assessed with quantitative histology and collagen measurement. Fibrocyte pool size in bleomycin-treated mice lungs was attenuated from 26.5% to 13.7% by pirfenidone on FACS analysis. This outcome was also observed in a therapeutic setting. Immunohistochemistry revealed that fibrocytes were significantly decreased by pirfenidone administration compared with those in bleomycin-treated mice (P = 0.0097). Increased chemokine (CC motif) ligand-2 (CCL2) and CCL12 production in bleomycin-treated mouse lungs was significantly attenuated by pirfenidone (P = 0.0003 and P < 0.0001, respectively). Pirfenidone also attenuated macrophage counts stimulated by bleomycin in BAL fluid. Fibrocyte migration toward CCL2 and chemokine (CC motif) receptor-2 expression on fibrocytes was significantly inhibited by pirfenidone in vitro. CONCLUSIONS Pirfenidone attenuated the fibrocyte pool size in bleomycin-treated mouse lungs via attenuation of CCL2 and CCL12 production in vivo, and fibrocyte migration was inhibited by pirfenidone in vitro. Fibrocyte inhibition is considered a mechanism of anti-fibrotic action of pirfenidone.
Collapse
Affiliation(s)
| | | | - Arata Azuma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Clinical trials with new drugs for chronic obstructive pulmonary disease (COPD) have been performed. Viruses exacerbate COPD and bacteria may play a part in severe COPD; therefore, antibiotic and antiviral approaches have a sound rationale. Antiinflammatory approaches have been studied. Advances in understanding the molecular basis of other processes have resulted in novel drugs to target reactive oxidant species, mucus, proteases, fibrosis, cachexia, and muscle wasting, and accelerated aging. Studies with monoclonal antibodies have been disappointing, highlighting the tendency for infections and malignancies during treatment. Promising future directions are lung regeneration with retinoids and stem cells.
Collapse
Affiliation(s)
- Clare L Ross
- Imperial Clinical Respiratory Research Unit (ICRRU), Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), National Heart and Lung Institute (NHLI), St Mary's Hospital, Imperial College, Praed Street, Paddington, London W2 INY, UK
| | - Trevor T Hansel
- Imperial Clinical Respiratory Research Unit (ICRRU), Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), National Heart and Lung Institute (NHLI), St Mary's Hospital, Imperial College, Praed Street, Paddington, London W2 INY, UK.
| |
Collapse
|
30
|
Usuki J, Matsuda K, Azuma A, Kudoh S, Gemma A. Sequential analysis of myofibroblast differentiation and transforming growth factor-β1/Smad pathway activation in murine pulmonary fibrosis. J NIPPON MED SCH 2013; 79:46-59. [PMID: 22398790 DOI: 10.1272/jnms.79.46] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Myofibroblasts play a critical role in tissue fibrosis. However, the intracellular signaling pathways in myofibroblast differentiation are poorly understood. Here, we studied the relationship between transforming growth factor-β (TGF-β)/Smad pathway activation and myofibroblast differentiation in both in vivo and in vitro experiments. In murine bleomycin-induced pulmonary fibrosis, nuclear localization of phosphorylated Smad2/3 (p-Smad2/3) was observed in pulmonary fibrotic lesions 7 days after bleomycin injection, whereas α-smooth muscle actin (ASMA)-positive myofibroblasts appeared in the lesions at 14 days, when the cytoplasmic localization of p-Smad2/3 was observed. We also compared the effects of TGF-β1 on myofibroblast differentiation and on type I collagen expression in a murine lung fibroblast cell line (MLg2908). TGF-β1 induced rapid expression of p-Smad2/3 in nuclei, after which ASMA organization in the cytoplasm of fibroblasts was observed. However, TGF-β1 produced no effect on the quantity of ASMA, either in mRNA levels or protein levels, even after the phosphorylation of Smad2/3. In contrast, TGF-β1 upregulated the expression of type I collagen mRNA. These findings suggest that in pulmonary fibrosis the molecular mechanism of myofibroblast differentiation is complex and that the difference between ASMA expression and type I collagen expression is mediated by the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Jiro Usuki
- Division of Pulmonary Medicine, Infection Diseases and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
31
|
McGowan SE, McCoy DM. Platelet-derived growth factor-A and sonic hedgehog signaling direct lung fibroblast precursors during alveolar septal formation. Am J Physiol Lung Cell Mol Physiol 2013; 305:L229-39. [PMID: 23748534 DOI: 10.1152/ajplung.00011.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alveolar septal formation is required to support the respiration of growing mammals; in humans effacement of the alveolar surface and impaired gas exchange are critical features of emphysema and pulmonary fibrosis. Platelet-derived growth factor-A (PDGF-A) and its receptor PDGF-receptor-α (PDGFRα) are required for secondary septal elongation in mice during postnatal days 4 through 12 and they regulate the proliferation and septal location of interstitial fibroblasts. We examined lung fibroblasts (LF) to learn whether PDGFRα expression distinguished a population of precursor cells, with enhanced proliferative and migratory capabilities. We identified a subpopulation of LF that expresses sonic hedgehog (Shh) and stem cell antigen-1 (Sca1). PDGF-A and Shh both increased cytokinesis and chemotaxis in vitro, but through different mechanisms. In primary LF cultures, Shh signaled exclusively through a noncanonical pathway involving generation of Rac1-GTP, whereas both the canonical and noncanonical pathways were used by the Mlg neonatal mouse LF cell line. LF preferentially oriented their primary cilia toward their anterior pole during migration. Furthermore, a larger proportion of PDGFRα-expressing LF, which are more abundant at the septal tips, bore primary cilia compared with other alveolar cells. In pulmonary emphysema, destroyed alveolar septa do not regenerate, in part because cells fail to assume a configuration that allows efficient gas exchange. Better understanding how LF are positioned during alveolar development could identify signaling pathways, which promote alveolar septal regeneration.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service, Iowa City, IA, USA.
| | | |
Collapse
|
32
|
Li YJ, Shimizu T, Hirata Y, Inagaki H, Takizawa H, Azuma A, Kawada T, Sugawara I, Kudoh S, Sunazuka T, Omura S. EM, EM703 inhibit NF-kB activation induced by oxidative stress from diesel exhaust particle in human bronchial epithelial cells: Importance in IL-8 transcription. Pulm Pharmacol Ther 2013; 26:318-24. [DOI: 10.1016/j.pupt.2012.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 12/13/2012] [Accepted: 12/24/2012] [Indexed: 01/17/2023]
|
33
|
Bagnato G, Bitto A, Pizzino G, Irrera N, Sangari D, Cinquegrani M, Roberts WN, Matucci Cerinic M, Squadrito F, Altavilla D, Bagnato G, Saitta A. Simvastatin attenuates the development of pulmonary and cutaneous fibrosis in a murine model of systemic sclerosis. Rheumatology (Oxford) 2013; 52:1377-86. [DOI: 10.1093/rheumatology/ket144] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
34
|
Bagnato G, Bitto A, Irrera N, Pizzino G, Sangari D, Cinquegrani M, Roberts WN, Atteritano M, Altavilla D, Squadrito F, Bagnato G, Saitta A. Propylthiouracil prevents cutaneous and pulmonary fibrosis in the reactive oxygen species murine model of systemic sclerosis. Arthritis Res Ther 2013; 15:R120. [PMID: 24286160 PMCID: PMC3978728 DOI: 10.1186/ar4300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 09/16/2013] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Recent advances suggest that the cellular redox state may play a significant role in the progression of fibrosis in systemic sclerosis (SSc). Another, and as yet poorly accounted for, feature of SSc is its overlap with thyroid abnormalities. Previous reports demonstrate that hypothyroidism reduces oxidant stress. The aim of this study was therefore to evaluate the effect of propylthiouracil (PTU), and of the hypothyroidism induced by it, on the development of cutaneous and pulmonary fibrosis in the oxidant stress murine model of SSc. METHODS Chronic oxidant stress SSc was induced in BALB/c mice by daily subcutaneous injections of hypochlorous acid (HOCl) for 6 weeks. Mice (n = 25) were randomized into three arms: HOCl (n = 10), HOCl plus PTU (n = 10) or vehicle alone (n = 5). PTU administration was initiated 30 minutes after HOCl subcutaneous injection and continued daily for 6 weeks. Skin and lung fibrosis were evaluated by histologic methods. Immunohistochemical staining for alpha-smooth muscle actin (α-SMA) in cutaneous and pulmonary tissues was performed to evaluate myofibroblast differentiation. Lung and skin concentrations of vascular endothelial growth factor (VEGF), extracellular signal-related kinase (ERK), rat sarcoma protein (Ras), Ras homolog gene family (Rho), and transforming growth factor (TGF) β were analyzed by Western blot. RESULTS Injections of HOCl induced cutaneous and lung fibrosis in BALB/c mice. PTU treatment prevented both dermal and pulmonary fibrosis. Myofibroblast differentiation was also inhibited by PTU in the skin and lung. The increase in cutaneous and pulmonary expression of VEGF, ERK, Ras, and Rho in mice treated with HOCl was significantly prevented in mice co-administered with PTU. CONCLUSIONS PTU, probably through its direct effect on reactive oxygen species or indirectly through thyroid function inhibition, prevents the development of cutaneous and pulmonary fibrosis by blocking the activation of the Ras-ERK pathway in the oxidant-stress animal model of SSc.
Collapse
Affiliation(s)
- Gianluca Bagnato
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Gabriele Pizzino
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Donatella Sangari
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Maurizio Cinquegrani
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| | - William Neal Roberts
- Department of Internal Medicine, Division of Rheumatology, University of
Louisville, Louisville, KY 40292, Kentucky, USA
| | - Marco Atteritano
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Domenica Altavilla
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, Division of Pharmacology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Gianfilippo Bagnato
- Department of Clinical and Experimental Medicine, Division of Rheumatology,
University of Messina, Via Consolare Valeria n°1, 98100, Messina,
Italy
| | - Antonino Saitta
- Department of Clinical and Experimental Medicine, Division of Internal
Medicine, University of Messina, Via Consolare Valeria n°1, 98100,
Messina, Italy
| |
Collapse
|
35
|
Er A, Yazar E. Effects of tylosin, tilmicosin and tulathromycin on inflammatory mediators in bronchoalveolar lavage fluid of lipopolysaccharide-induced lung injury. Acta Vet Hung 2012; 60:465-76. [PMID: 23160029 DOI: 10.1556/avet.2012.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of this study was to determine the anti-inflammatory effects of macrolides through kinetic parameters in bronchoalveolar lavage fluid (BALF) of lipopolysaccharide-induced lung injury. Rats were divided into four groups: lipopolysaccharide (LPS), LPS + tylosin, LPS + tilmicosin and LPS + tulathromycin. BALF samples were collected at sampling times. TNF, IL-1β, IL-6, IL-10 and 13,14-dihydro-15-keto-prostaglandin F2α (PGM) and C-reactive protein (CRP) were analysed. Area under the curve (AUC) and maximum plasma concentration (Cmax) values of inflammatory mediators were determined by a pharmacokinetic computer programme. When inflammatory mediator concentrations were compared between the LPS group and other groups for each sampling time, the three macrolides had no pronounced depressor effect on cytokine levels, but they depressed PGM and CRP levels. In addition, tylosin and tilmicosin decreased the AUC0-24 level of TNF, while tilmicosin decreased the AUC0-24 level of IL-10. Tylosin and tulathromycin decreased the AUC0-24 of PGM, and all three macrolides decreased the AUC0-24 of CRP. Especially tylosin and tulathromycin may have more expressed anti-inflammatory effects than tilmicosin, via depressing the production of inflammatory mediators in the lung. The AUC may be used for determining the effects of drugs on inflammation. In this study, the antiinflammatory effects of these antibiotics were evaluated with kinetic parameters as a new and different approach.
Collapse
Affiliation(s)
- Ayse Er
- 1 University of Selcuk, Campus Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine 42075 Konya Turkey
| | - Enver Yazar
- 1 University of Selcuk, Campus Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine 42075 Konya Turkey
| |
Collapse
|
36
|
Flume PA, Van Devanter DR. State of progress in treating cystic fibrosis respiratory disease. BMC Med 2012; 10:88. [PMID: 22883684 PMCID: PMC3425089 DOI: 10.1186/1741-7015-10-88] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 08/10/2012] [Indexed: 12/12/2022] Open
Abstract
Since the discovery of the gene associated with cystic fibrosis (CF), there has been tremendous progress in the care of patients with this disease. New therapies have entered the market and are part of the standard treatment of patients with CF, and have been associated with marked improvement in survival. Now there are even more promising therapies directed at different components of the pathophysiology of this disease. In this review, our current knowledge of the pathophysiology of lung disease in patients with CF is described, along with the current treatment of CF lung disease, and the therapies in development that offer great promise to our patients.
Collapse
|
37
|
Bosnar M, Kragol G, Koštrun S, Vujasinović I, Bošnjak B, Bencetić Mihaljević V, Marušić Ištuk Z, Kapić S, Hrvačić B, Brajša K, Tavčar B, Jelić D, Glojnarić I, Verbanac D, Čulić O, Padovan J, Alihodžić S, Eraković Haber V, Spaventi R. N′-Substituted-2′-O,3′-N-carbonimidoyl Bridged Macrolides: Novel Anti-inflammatory Macrolides without Antimicrobial Activity. J Med Chem 2012; 55:6111-23. [DOI: 10.1021/jm300356u] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Martina Bosnar
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Goran Kragol
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Sanja Koštrun
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Ines Vujasinović
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Berislav Bošnjak
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | | | - Zorica Marušić Ištuk
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Samra Kapić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Boška Hrvačić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Karmen Brajša
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Branka Tavčar
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Dubravko Jelić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Ines Glojnarić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Donatella Verbanac
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Ognjen Čulić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Jasna Padovan
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Sulejman Alihodžić
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Vesna Eraković Haber
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| | - Radan Spaventi
- GlaxoSmithKline Research Centre
Zagreb, Prilaz baruna Filipovića 29, Zagreb, Croatia
| |
Collapse
|
38
|
Dong XS, Hu XB, Liu W, Sun YQ, Liu Z. Effects of RNA interference-induced Smad3 gene silencing on pulmonary fibrosis caused by paraquat in mice. Exp Biol Med (Maywood) 2012; 237:548-55. [PMID: 22581813 DOI: 10.1258/ebm.2011.011280] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Paraquat (PQ) poisoning induces many physiological and histological changes in the human body, but PQ-induced pulmonary fibrosis is most often associated with death. The signaling pathway associated with pulmonary fibrosis is reliant on transforming growth factor-beta 1 (tgf-β(1)) activation of Smad3, as evidenced by Smad3-deficient mice being resistant to tgf-β(1)-induced pulmonary fibrosis. Thus, we sought to determine whether targeted silencing of Smad3 gene expression could inhibit PQ-induced pulmonary fibrosis in mice. We developed an RNA interference (RNAi) method using short hairpin RNAs (shRNAs) targeting Smad3. The shRNA expression cassettes capable of effectively silencing Smad3 in L929 mouse fibroblasts were transferred to an adenovirus vector and intratracheally administered into mouse lung. Treated mice presented with inhibited Smad3 mRNA and protein and were resistant to PQ-induced pulmonary fibrosis, as evidenced by suppressed expressions of procollagen type I mRNA and hydroxyproline amino acid. Thus, silencing of Smad3 appears to be a promising alternative strategy for the treatment of PQ-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Xue-Song Dong
- Department of Emergency, The First Affiliated Hospital, China Medical University, Shenyang 110001, China.
| | | | | | | | | |
Collapse
|
39
|
Wang X, Wu G, Gou L, Liu Z, Wang X, Fan X, Wu L, Liu N. A novel single-chain-Fv antibody against connective tissue growth factor attenuates bleomycin-induced pulmonary fibrosis in mice. Respirology 2011; 16:500-7. [PMID: 21265950 DOI: 10.1111/j.1440-1843.2011.01938.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Connective tissue growth factor (CTGF) has been identified as playing critical roles in fibrosis and is a promising therapeutic target. In a previous study, we used a phage display library to develop a humanized single-chain variable fragment antibody (scFv) against CTGF. In the present study, the protective effect of anti-CTGF scFv against bleomycin (BL)-induced pulmonary fibrosis was investigated in mice. METHODS The expression of α-smooth muscle actin in human embryonic lung fibroblast (HELF) cells was analysed by western blotting. A mouse model of pulmonary fibrosis was established by tracheal injection of BL (5 mg/kg). Mice received anti-CTGF scFv (4 mg/kg, three times a week) by i.v. injection. The effects of anti-CTGF scFv were evaluated by leukocyte counts in BAL fluid, hydroxyproline measurements in lung tissue and pathological examination. RESULTS α-Smooth muscle actin expression was decreased in HELF cells treated with anti-CTGF scFv. Anti-CTGF scFv significantly reduced the numbers of inflammatory leukocytes (total and differential count) in BAL fluid, as well as the hydroxyproline content of lung tissue. The severity of alveolitis and fibrosis in the mouse model was markedly attenuated by treatment with anti-CTGF scFv. CONCLUSIONS Anti-CTGF scFv may potentially be developed as a useful inhibitor of pulmonary fibrosis.
Collapse
Affiliation(s)
- Xihua Wang
- Respiratory Department, Institute of Biotechnology and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Arribillaga L, Dotor J, Basagoiti M, Riezu-Boj JI, Borrás-Cuesta F, Lasarte JJ, Sarobe P, Cornet ME, Feijoó E. Therapeutic effect of a peptide inhibitor of TGF-β on pulmonary fibrosis. Cytokine 2010; 53:327-33. [PMID: 21185199 DOI: 10.1016/j.cyto.2010.11.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Revised: 07/21/2010] [Accepted: 11/23/2010] [Indexed: 12/17/2022]
Abstract
Pulmonary fibrosis encompasses several respiratory diseases characterized by epithelial cell injury, inflammation and fibrosis. Transforming growth factor (TGF)-β1 is one of the main profibrogenic cytokines involved in the pathogenesis of lung fibrosis. It induces fibroblast differentiation into myofibroblasts, which produce high levels of collagen and concomitantly loss of lung elasticity and reduction of the respiratory function. In the present study, we have investigated the effects of P17 (a TGF-β inhibitor peptide) on IMR-90 lung fibroblast differentiation in vitro, as well as on the inhibition of the development of bleomycin-induced pulmonary fibrosis in mice. It was found that in IMR-90 cells, P17 inhibited TGF-β1-induced expression of connective tissue growth factor and α-smooth muscle actin. In vivo, treatment of mice with P17 2days after bleomycin administration decreased lung fibrosis, areas of myofibroblast-like cells and lymphocyte infiltrate. P17 also reduced mRNA expression of collagen type I, fibronectin and the fibronectin splice isoform EDA in the lung, and increased the expression of IFN-γ mRNA. Finally, therapeutic treatment with P17 in mice with already established fibrosis was able to significantly attenuate the progression of lung fibrosis. These results suggest that P17 may be useful in the treatment of pulmonary fibrosis.
Collapse
|
41
|
Vaishnav P, Demain AL. Unexpected applications of secondary metabolites. Biotechnol Adv 2010; 29:223-9. [PMID: 21130862 DOI: 10.1016/j.biotechadv.2010.11.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 11/23/2010] [Accepted: 11/24/2010] [Indexed: 11/16/2022]
Abstract
Secondary metabolites have been found to have interesting applications over and above their well-known medical uses, e.g., as antimicrobials, etc. These alternative applications include antitumor, cholesterol-lowering, immunosuppressant, antiprotozoal, antihelminth, antiviral and anti-ageing activities. Polyene antibiotics, such as amphotericin B, are of use as antiprion agents, antitumor drugs and against leishmaniasis. Other microbial natural products that show antibiotic activity are used against cancer e.g., doxorubicin, neomycin, β-lactams, bleomycin and rapamycin. Macrolide antibiotics, such as erythromycin, clarithromycin and azithromycin, improve pulmonary function in patients suffering from panbioncholitis. Pigments like prodigiosin and shikonin have antitumor activity, while violacein has anti-ulcer and antitumor activity and also acts as an antiprotozoal agent. Statins, in addition to lowering cholesterol and LDL levels, also decrease elevated C-reactive protein (CRP) levels independent of their cholesterol effects. Immunosuppressants have many alternative effects: (i) Cyclosporin is proving useful in treatment of inflammatory disease such as asthma and muscular dystrophy. (ii) Rapamycin is extremely useful in preventing restenosis of stents grafted in balloon angioplasty. (iii) Tacrolimus and ascomycin help in treating inflammatory skin disease such as allergic contact dermatitis and psoriasis. Artemisinin, an antimalarial agent, is also showing antitumor activity. Other natural products, including those from plants (betulinic acid and shikonin), animals (bryostatins) and microbes (squalestatin and sophorolipids) have a multiplicity of potentially useful actions. Unexpected functions of known secondary metabolites are continuously being unraveled, and are fulfilling some of the needs of present day medicine and show great promise for the future.
Collapse
Affiliation(s)
- Preeti Vaishnav
- 206 Akshardeepp Apts., near New Jain Temple, GIDC, Ankleshwar, Gujarat, India
| | | |
Collapse
|
42
|
Furuya A, Asano K, Shoji N, Hirano K, Hamasaki T, Suzaki H. Suppression of nitric oxide production from nasal fibroblasts by metabolized clarithromycin in vitro. JOURNAL OF INFLAMMATION-LONDON 2010; 7:56. [PMID: 21092318 PMCID: PMC3003651 DOI: 10.1186/1476-9255-7-56] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 11/23/2010] [Indexed: 11/29/2022]
Abstract
Background Low-dose and long-term administration of 14-membered macrolide antibiotics, so called macrolide therapy, has been reported to favorably modify the clinical conditions of chronic airway diseases. Since there is growing evidence that macrolide antibiotic-resistant bacteria's spreaders in the populations received macrolide therapy, it is strongly desired to develop macrolide antibiotics, which showed only anti-inflammatory action. The present study was designed to examine the influence of clarithromycin (CAM) and its metabolized materials, M-1, M-4 and M-5, on free radical generation from nasal polyp fibroblasts (NPFs) through the choice of nitric oxide (NO), which is one of important effector molecule in the development of airway inflammatory disease in vitro. Methods NPFs (5 × 105 cells/ml) were stimulated with 1.0 μg/ml lipopolysaccharide (LPS) in the presence of agents for 24 hours. NO levels in culture supernatants were examined by the Griess method. We also examined the influence of agents on the phosphorylation of MAPKs, NF-κB activation, iNOS mRNA expression and iNOS production in NPFs cultured for 2, 4, 8, and 12 hours, respectively. Results The addition of CAM (> 0.4 μg/ml) and M-4 (> 0.04 μg/ml) could suppress NO production from NPFs after LPS stimulation through the suppression of iNOS mRNA expression and NF-κB activation. CAM and M-4 also suppressed phosphorylation of MAPKs, ERK and p38 MAPK, but not JNK, which are increased LPS stimulation. On the other hand, M-1 and M-5 could not inhibit the NO generation, even when 0.1 μg/ml of the agent was added to cell cultures. Conclusion The present results may suggest that M-4 will be a good candidate for the agent in the treatment of chronic airway inflammatory diseases, since M-4 did not have antimicribiological effects on gram positive and negative bacteria.
Collapse
Affiliation(s)
- Ayako Furuya
- Division of Physiology, School of Nursing and Rehabilitation Sciences, Showa University, Yokohama, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Mechanisms of action and clinical application of macrolides as immunomodulatory medications. Clin Microbiol Rev 2010; 23:590-615. [PMID: 20610825 DOI: 10.1128/cmr.00078-09] [Citation(s) in RCA: 442] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Macrolides have diverse biological activities and an ability to modulate inflammation and immunity in eukaryotes without affecting homeostatic immunity. These properties have led to their long-term use in treating neutrophil-dominated inflammation in diffuse panbronchiolitis, bronchiectasis, rhinosinusitis, and cystic fibrosis. These immunomodulatory activities appear to be polymodal, but evidence suggests that many of these effects are due to inhibition of extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation and nuclear factor kappa B (NF-kappaB) activation. Macrolides accumulate within cells, suggesting that they may associate with receptors or carriers responsible for the regulation of cell cycle and immunity. A concern is that long-term use of macrolides increases the emergence of antimicrobial resistance. Nonantimicrobial macrolides are now in development as potential immunomodulatory therapies.
Collapse
|
44
|
Wuyts WA, Willems S, Vos R, Vanaudenaerde BM, De Vleeschauwer SI, Rinaldi M, Vanhooren HM, Geudens N, Verleden SE, Demedts MG, Thomeer M, Verbeken EK, Verleden GM. Azithromycin reduces pulmonary fibrosis in a bleomycin mouse model. Exp Lung Res 2010; 36:602-14. [PMID: 20874225 DOI: 10.3109/01902148.2010.492895] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating disease without proper treatment. Despite intensive research, the exact underlying pathogenesis remains elusive. It is regarded as a continuous injury, resulting in inflammation, infiltration, and proliferation of fibroblasts and extracellular matrix deposition, leading to an irreversible restrictive lung function deterioration and death. In this study the effect of azithromycin, a macrolide antibiotic on bleomycin-induced pulmonary fibrosis was investigated. C57BL/6 mice were intratracheally instilled with bleomycin (0.5 mg/kg) or saline. In the bleomycin group, half of the animals received azithromycin every other day from day 1 on. Bronchoalveolar lavage and histology were performed at days 7 and 35, and pulmonary function tests on day 35. At day 35, fibrotic lesions (spindle cell proliferation/collagen I deposition) were paralleled by a restrictive lung function pattern. Alterations were found in neutrophils and macrophages (innate immunity) and in T(H)2, T(H)17, and Treg cytokines (adaptive immunity). Azithromycin significantly reduced both fibrosis and the restrictive lung function pattern. This study demonstrated a beneficial effect of azithromycin on bleomycin-induced pulmonary fibrosis. A possible mechanism could be a modulation of both innate immunity and adaptive immunity. These findings might suggest a potential role for azithromycin in the treatment of IPF.
Collapse
Affiliation(s)
- W A Wuyts
- Laboratory of Pneumology, Katholieke Universiteit Leuven and University Hospitals Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bioscreening of phage display antibody library and expression of a humanized single-chain variable fragment antibody against human connective tissue growth factor (CTGF/CCN2). Biotechnol Appl Biochem 2010; 56:95-102. [PMID: 20491654 DOI: 10.1042/ba20100031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Excessive expression of CTGF (connective tissue growth factor)/CCN2 has been observed in many fibrotic diseases. The inhibition of the CTGF/CCN2 by antibody has been shown to be clinically useful for the management of fibrosis. A phage display humanized single-chain Fv antibody library was screened using CTGF/C (CTGF/CCN2 C-terminal domain) as the target. A phage ELISA was performed after four rounds of biopanning, and ten positive clones were further evaluated by ELISA and were chosen for DNA sequencing. The DNA encoding scFv (single-chain variable fragment) containing a full-length variable domain fragment of heavy chain and light chain of human immunoglobulin was inserted into pET-32(a)+ vector, and the fusion protein (TrxA-scFv) containing a thrombin cleavage site was expressed mainly in soluble form. The scFv was obtained by purified fusion protein digested with thrombin and then separated from the fusion partner TrxA by gel-filtration chromatography. An immunological assay showed that the purified scFv reacted with CTGF/CCN2 in a concentration-dependent manner. The result of the cell migration assay demonstrated that the scFv at 100 ng/ml could effectively inhibit the migration of HUVEC (human umbilical-vein endothelial cells) caused by CTGF/C. The number of migratory cells was significantly decreased as compared with the negative control (1062+/-92 versus 3269+/-288, P<0.001) and the inhibition rate was 90.5%.
Collapse
|
46
|
[Liver growth factor improves pulmonary fibrosis secondary to cadmium administration in rats]. Arch Bronconeumol 2010; 46:20-6. [PMID: 20034722 DOI: 10.1016/j.arbres.2009.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 09/04/2009] [Accepted: 10/13/2009] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Liver growth factor (LGF) is a liver mitogen with regenerating and anti-fibrotic activity even at extrahepatic sites. We used LGF in a lung fibrosis model induced by cadmium chloride (CdCl(2)), to study its antifibrotic capacity. METHODS Forty-two male Wistar rats were administered a single dose of 0.5ml/rat of CdCl2 0.025% (n=21) or the same volume of saline (control group, n=21). After 35 days, once a lesion was established, we started a 3 week treatment with LGF, after which we determined lung function--inspiratory capacity (IC), lung compliance (LC), forced vital capacity (FVC) and expiratory flow at 75% (FEF75%)-, lung morphometry--alveolar internal area (AIA), mean linear intersection (LM)-, and collagen (both by Sirius red and hydroxyproline residues) and elastin contents. RESULTS Pulmonary fibrosis in CdCl(2) rats was characterized by a marked decrease in pulmonary function with respect to healthy controls -reductions of 28% in IC, 38% in CL, 31% in FVC, and 54% in FEF75%- which was partially recovered after LGF injection -18% IC, 27% CL, 19% FVC and 35% FEF75%-; increase in collagen and elastin contents -165% and 76%, respectively, in CdCl2 rats, versus 110% and 34% after LGF injection-; and increases in AIA and LM, partially reverted by LGF. CONCLUSIONS Together, these data seem to demonstrate that LGF is able to improve lung function and partially reverts the increase in lung matrix proteins produced by CdCl(2) instillation.
Collapse
|
47
|
Martínez-Galán L, del Puerto-Nevado L, Pérez-Rial S, Díaz-Gil JJ, González-Mangado N, Peces-Barba G. Liver Growth Factor Improves Pulmonary Fibrosis Secondary to Cadmium Administration in Rats. ACTA ACUST UNITED AC 2010. [DOI: 10.1016/s1579-2129(10)70005-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
48
|
Abstract
Tobacco smoking is the dominant risk factor for chronic obstructive pulmonary disease (COPD), but viral and bacterial infections are the major causes of exacerbations in later stages of disease. Reactive oxygen species (ROS), pathogen-associated molecular patterns (PAMPs), and damage-associated molecular patterns (DAMPs) activate families of pattern recognition receptors (PRRs) that include the toll-like receptors (TLRs). This understanding has led to the hypothesis that COPD is an archetypal disease of innate immunity. COPD is characterised by abnormal response to injury, with altered barrier function of the respiratory tract, an acute phase reaction, and excessive activation of macrophages, neutrophils, and fibroblasts in the lung. The activated non-specific immune system then mediates the processes of inflammation and repair, fibrosis, and proteolysis. COPD is also associated with corticosteroid resistance, abnormal macrophage and T-cell populations in the airway, autoinflammation and autoimmunity, aberrant fibrosis, accelerated ageing, systemic and concomitant disease, and defective regeneration. Such concepts have been used to generate a range of molecular targets, and clinical trials are taking place to identify effective drugs for the prevention and treatment of COPD exacerbations.
Collapse
Affiliation(s)
- Trevor T Hansel
- National Heart and Lung Institute, Imperial College, London, UK.
| | | |
Collapse
|
49
|
|
50
|
Abstract
Aim To approach the effects of luteolin on proliferation of rat lung fibroblasts. Methods The lung fibroblasts were isolated from the lungs of Sprague-Dawley rats at postnatal day 3, and were then passaged to 96-well culture plates. All cells were divide into 6 groups including normal control group, positive control group, 4×10 time, the cytomorphology of LFb, drew growth curve, determined superoxide dismutase (SOD) activity were observed. Results Luteolin at the concentration of 4×10−4, 4×10−5 mol·L−1 inhibited evidently the growth and proliferation of lung fibroblasts, and enhanced SOD activity. The density, interval and volume of lung fibroblasts became decrescent. The effects, dose-dependently, had obvious statistical significance compared with normal control group. Conclusion Luteolin can inhibit the proliferation of the lung fibroblasts in rat.
Collapse
|