51
|
Xie L, Zeng Y. Therapeutic Potential of Exosomes in Pulmonary Fibrosis. Front Pharmacol 2020; 11:590972. [PMID: 33343360 PMCID: PMC7746877 DOI: 10.3389/fphar.2020.590972] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis is closely associated with the recruitment of fibroblasts from capillary vessels with damaged endothelial cells, the epithelial mesenchymal transition (EMT) of type II alveolar epithelial cells, and the transformation of fibroblasts to myofibroblasts. Recent studies suggest that EMT is a key factor in the pathogenesis of pulmonary fibrosis, as the disruption of EMT-related effector molecules can inhibit the occurrence and development of PF. With the numerous advancements made in molecular biology in recent years, researchers have discovered that exosomes and their cargos, such as miRNAs, lncRNAs, and proteins, can promote or inhibit the EMT, modulate the transformation of fibroblasts into myofibroblasts, contribute to the proliferation of fibroblasts and promote immunoregulatory and mitochondrial damage during pulmonary fibrosis. Exosomes are key factors regulating the differentiation of bone marrow mesenchymal stem cells (BMSCs) into myofibroblasts. Interestingly, exosomes derived from BMSCs under pathological and physiological conditions may promote or inhibit the EMT of type II alveolar epithelial cells and the transformation of fibroblasts into myofibroblasts to regulate pulmonary fibrosis. Thus, exosomes may become a new direction in the study of drugs for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
52
|
The therapeutic potential of galectin-3 inhibition in fibrotic disease. Int J Biochem Cell Biol 2020; 130:105881. [PMID: 33181315 DOI: 10.1016/j.biocel.2020.105881] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/20/2022]
Abstract
Galectin-3 is a beta-galactoside-binding mammalian lectin and part of the 15 member galectin family that are evolutionarily highly conserved. It is the only chimeric protein with a C-terminal carbohydrate recognition domain (CRD) linked to a proline, glycine, and tyrosine rich additional N-terminal domain. Galectin-3 binds several cell surface glycoproteins via its CRD domain as well as undergoing oligomerization, via binding at the N-terminal or the CRD, resulting in the formation of a galectin-3 lattice on the cell surface. The galectin-3 lattice has been regarded as being a crucial mechanism whereby extracellular galectin-3 modulates cellular signalling by prolonging retention time or retarding lateral movement of cell surface receptors in the plasma membrane. As such galectin-3 can regulate various cellular functions such as diffusion, compartmentalization and endocytosis of plasma membrane glycoproteins and glycolipids and the functionality of membrane receptors. In multiple models of organ fibrosis, it has been demonstrated that galectin-3 is potently pro-fibrotic and modulates the activity of fibroblasts and macrophages in chronically inflamed organs. Increased galectin-3 expression also activates myofibroblasts resulting in scar formation and may therefore impact common fibrotic pathways leading to fibrosis in multiple organs. Over the last decade there has been a marked increase in the scientific literature investigating galectin-3 in a range of fibrotic diseases as well as the clinical development of new galectin-3 inhibitors. In this review we will examine the role of galectin-3 in fibrosis, the therapeutic strategies for inhibiting galectin-3 in fibrotic disease and the clinical landscape to date.
Collapse
|
53
|
Liu YL, Chen BY, Nie J, Zhao GH, Zhuo JY, Yuan J, Li YC, Wang LL, Chen ZW. Polydatin prevents bleomycin-induced pulmonary fibrosis by inhibiting the TGF-β/Smad/ERK signaling pathway. Exp Ther Med 2020; 20:62. [PMID: 32952652 PMCID: PMC7485305 DOI: 10.3892/etm.2020.9190] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, irreversible interstitial lung disease, with no effective cure. Polydatin is a resveratrol glucoside with strong antioxidant, anti-inflammatory and anti-apoptotic properties, which is used for treating health-related disorders such as cardiac disabilities, various types of carcinoma, hepatitis and hepatic fibrosis. The present study aimed to investigate the protective effect of polydatin against bleomycin-induced IPF and the possible underlying mechanism. A549 cells were treated with transforming growth factor-β1 (TGF-β1) and polydatin to observe phenotypic transformation and the related gene expression was detected. Sprague-Dawley rats were divided into seven groups and intratracheally infused with bleomycin to establish a pulmonary fibrosis model (the sham control group received saline). The rats were given pirfenidone (50 mg/kg), resveratrol (40 mg/kg) and polydatin (10, 40 and 160 mg/kg) for 28 days. The results demonstrated that polydatin had low toxicity to A549 cells and inhibited TGF-β1-induced phenotypic transformation as determined by MTS assay or observed using a light microscope. It also decreased the gene expression levels of α-smooth muscle actin and collagen I and increased the gene expression levels of epithelial cell cadherin in vitro and in vivo by reverse transcription-quantitative PCR. Furthermore, polydatin ameliorated the pathological damage and fiber production in lung tissues found by hematoxylin and eosin staining and Masson trichrome staining. Polydatin administration markedly reduced the levels of hydroxyproline, tumor necrosis factor-α, interleukin (IL)-6, IL-13, myeloperoxidase and malondialdehyde and promoted total superoxide dismutase activity in lung tissues as determined using ELISA kits or biochemical reagent kits. It inhibited TGF-β1 expression and phosphorylation of Smad 2 and 3 and ERK-1 and -2 in vivo as determined by western blot assays. These results suggest that polydatin protects against IPF via its anti-inflammatory, antioxidant and antifibrotic activities, and the mechanism may be associated with its regulatory effect on the TGF-β pathway.
Collapse
Affiliation(s)
- Yan-Lu Liu
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Bao-Yi Chen
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Juan Nie
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Guang-Hui Zhao
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Jian-Yi Zhuo
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Jie Yuan
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Yu-Cui Li
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Ling-Li Wang
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Zhi-Wei Chen
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
54
|
MiR-200a inversely correlates with Hedgehog and TGF-β canonical/non-canonical trajectories to orchestrate the anti-fibrotic effect of Tadalafil in a bleomycin-induced pulmonary fibrosis model. Inflammopharmacology 2020; 29:167-182. [PMID: 32914382 DOI: 10.1007/s10787-020-00748-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022]
Abstract
Few reports have documented the ability of phosphodiesterase-5 inhibitors (PDE-5-Is) to ameliorate idiopathic pulmonary fibrosis (IPF) mainly by their anti-inflammatory/antioxidant capacities, without unveiling the possible molecular mechanisms involved. Because of the recent role of miR-200 family and Sonic Hedgehog (SHH) trajectory in IPF, we have studied their impact on the anti-fibrotic potential of tadalafil against bleomycin-induced pulmonary fibrosis. Animals were allocated into normal-control, bleomycin-fibrotic control, and bleomycin post-treated with tadalafil or dexamethasone, as the reference drug. On the molecular level, tadalafil has reverted the bleomycin effect on all the assessed parameters. Tadalafil upregulated the gene expression of miR-200a, but decreased the smoothened (SMO) and the transcription factors glioma-associated oncogene homolog (Gli-1, Gli-2), members of SHH pathway. Additionally, tadalafil ebbed transforming growth factor (TGF)-β, its canonical (SMAD-3/alpha smooth muscle actin [α-SMA] and Snail), and non-canonical (p-Akt/p-Forkhead box O3 (FOXO3) a) pathways. Besides, a strong negative correlation between miR-200a and the analyzed pathways was proved. The effect of tadalafil was further confirmed by the improved lung structure and the reduced Ashcroft score/collagen deposition. The results were comparable to that of dexamethasone. In conclusion, our study has highlighted the involvement of miR-200a in the anti-fibrotic effect of tadalafil with the inhibition of SHH hub and the pro-fibrotic pathways (TGF-β/ SMAD-3/α-SMA, Snail and p-AKT/p-FOXO3a). Potential anti-fibrotic effect of tadalafil. Modulation of miR200a/SHH/canonical and non-canonical TGF-β trajectories. → : stimulatory effect; ┴: inhibitory effect.
Collapse
|
55
|
Louzada RA, Corre R, Ameziane El Hassani R, Meziani L, Jaillet M, Cazes A, Crestani B, Deutsch E, Dupuy C. NADPH oxidase DUOX1 sustains TGF-β1 signalling and promotes lung fibrosis. Eur Respir J 2020; 57:13993003.01949-2019. [DOI: 10.1183/13993003.01949-2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 07/22/2020] [Indexed: 01/20/2023]
Abstract
Interstitial lung fibroblast activation coupled with extracellular matrix production is a pathological signature of pulmonary fibrosis, and is governed by transforming growth factor (TGF)-β1/Smad signalling. TGF-β1 and oxidative stress cooperate to drive fibrosis. Cells can produce reactive oxygen species through activation and/or induction of NADPH oxidases, such as dual oxidase (DUOX1/2). Since DUOX enzymes, as extracellular hydrogen peroxide (H2O2)-generating systems, are involved in extracellular matrix formation and in wound healing in different experimental models, we hypothesised that DUOX-based NADPH oxidase plays a role in the pathophysiology of pulmonary fibrosis.Our in vivo data (idiopathic pulmonary fibrosis patients and mouse models of lung fibrosis) showed that the NADPH oxidase DUOX1 is induced in response to lung injury. DUOX1-deficient mice (DUOX1+/− and DUOX1−/−) had an attenuated fibrotic phenotype. In addition to being highly expressed at the epithelial surface of airways, DUOX1 appears to be well expressed in the fibroblastic foci of remodelled lungs. By using primary human and mouse lung fibroblasts, we showed that TGF-β1 upregulates DUOX1 and its maturation factor DUOXA1 and that DUOX1-derived H2O2 promoted the duration of TGF-β1-activated Smad3 phosphorylation by preventing phospho-Smad3 degradation. Analysis of the mechanism revealed that DUOX1 inhibited the interaction between phospho-Smad3 and the ubiquitin ligase NEDD4L, preventing NEDD4L-mediated ubiquitination of phospho-Smad3 and its targeting for degradation.These findings highlight a role for DUOX1-derived H2O2 in a positive feedback that amplifies the signalling output of the TGF-β1 pathway and identify DUOX1 as a new therapeutic target in pulmonary fibrosis.
Collapse
|
56
|
Kim S, Lim JH, Woo CH. Therapeutic potential of targeting kinase inhibition in patients with idiopathic pulmonary fibrosis. Yeungnam Univ J Med 2020; 37:269-276. [PMID: 32693446 PMCID: PMC7606966 DOI: 10.12701/yujm.2020.00458] [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: 06/08/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Fibrosis is characterized by excessive accumulation of extracellular matrix components. The fibrotic process ultimately leads to organ dysfunction and failure in chronic inflammatory and metabolic diseases such as pulmonary fibrosis, advanced kidney disease, and liver cirrhosis. Idiopathic pulmonary fibrosis (IPF) is a common form of progressive and chronic interstitial lung disease of unknown etiology. Pathophysiologically, the parenchyma of the lung alveoli, interstitium, and capillary endothelium becomes scarred and stiff, which makes breathing difficult because the lungs have to work harder to transfer oxygen and carbon dioxide between the alveolar space and bloodstream. The transforming growth factor beta (TGF-β) signaling pathway plays an important role in the pathogenesis of pulmonary fibrosis and scarring of the lung tissue. Recent clinical trials focused on the development of pharmacological agents that either directly or indirectly target kinases for the treatment of IPF. Therefore, to develop therapeutic targets for pulmonary fibrosis, it is essential to understand the key factors involved in the pathogenesis of pulmonary fibrosis and the underlying signaling pathway. The objective of this review is to discuss the role of kinase signaling cascades in the regulation of either TGF-β-dependent or other signaling pathways, including Rho-associated coiled-coil kinase, c-jun N-terminal kinase, extracellular signal-regulated kinase 5, and p90 ribosomal S6 kinase pathways, and potential therapeutic targets in IPF.
Collapse
Affiliation(s)
- Suji Kim
- Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea.,Department of Pharmacology, Yeungnam University College of Medicine, Daegu, Korea
| | - Jae Hyang Lim
- Department of Microbiology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Chang-Hoon Woo
- Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea.,Department of Pharmacology, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
57
|
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: 2.0] [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
|
58
|
Lipid Mediators Regulate Pulmonary Fibrosis: Potential Mechanisms and Signaling Pathways. Int J Mol Sci 2020; 21:ijms21124257. [PMID: 32549377 PMCID: PMC7352853 DOI: 10.3390/ijms21124257] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease of unknown etiology characterized by distorted distal lung architecture, inflammation, and fibrosis. The molecular mechanisms involved in the pathophysiology of IPF are incompletely defined. Several lung cell types including alveolar epithelial cells, fibroblasts, monocyte-derived macrophages, and endothelial cells have been implicated in the development and progression of fibrosis. Regardless of the cell types involved, changes in gene expression, disrupted glycolysis, and mitochondrial oxidation, dysregulated protein folding, and altered phospholipid and sphingolipid metabolism result in activation of myofibroblast, deposition of extracellular matrix proteins, remodeling of lung architecture and fibrosis. Lipid mediators derived from phospholipids, sphingolipids, and polyunsaturated fatty acids play an important role in the pathogenesis of pulmonary fibrosis and have been described to exhibit pro- and anti-fibrotic effects in IPF and in preclinical animal models of lung fibrosis. This review describes the current understanding of the role and signaling pathways of prostanoids, lysophospholipids, and sphingolipids and their metabolizing enzymes in the development of lung fibrosis. Further, several of the lipid mediators and enzymes involved in their metabolism are therapeutic targets for drug development to treat IPF.
Collapse
|
59
|
Li X, Yu H, Liang L, Bi Z, Wang Y, Gao S, Wang M, Li H, Miao Y, Deng R, Ma L, Luan J, Li S, Liu M, Lin J, Zhou H, Yang C. Myricetin ameliorates bleomycin-induced pulmonary fibrosis in mice by inhibiting TGF-β signaling via targeting HSP90β. Biochem Pharmacol 2020; 178:114097. [PMID: 32535102 DOI: 10.1016/j.bcp.2020.114097] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 01/06/2023]
Abstract
Idiopathic pulmonary fibrosis is a progressive-fibrosing lung disease with high mortality and limited therapy, which characterized by myofibroblasts proliferation and extracellular matrix deposition. Myricetin, a natural flavonoid, has been shown to possess a variety of biological characteristics including anti-inflammatory and anti-tumor. In this study we explored the potential effect and mechanisms of myricetin on pulmonary fibrosis in vivo and vitro. The in vivo studies showed that myricetin effectively alleviated bleomycin (BLM)-induced pulmonary fibrosis. KEGG analysis of RNA-seq data indicated that myricetin could regulate the transforming growth factor (TGF)-β signaling pathway. In vitro studies indicated that myricetin could dose-dependently suppress TGF-β1/Smad signaling and attenuate TGF-β1-induced fibroblast activation and epithelial-mesenchymal transition (EMT). Molecular docking indicated that heat shock protein (HSP) 90β may be a potential target of myricetin, and MST assay demonstrated that the dissociation constant (Kd) of myricetin and HSP90β was 331.59 nM. We demonstrated that myricetin interfered with the binding of HSP90β and TGF-β receptor II and impeded fibroblast activation and EMT. In conclusion, myricetin impedes TGF-β1-induced lung fibroblast activation and EMT via targeting HSP90β and attenuates BLM-induced pulmonary fibrosis in mice.
Collapse
Affiliation(s)
- Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Haiyan Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Lu Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Zhun Bi
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Yanhua Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Mukuo Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Hailong Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Yang Miao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China
| | - Ruxia Deng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Ling Ma
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Jiaoyan Luan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Shuangling Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Menghan Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China
| | - Jianping Lin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China.
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, 300457, People's Republic of China.
| |
Collapse
|
60
|
Conditional deletion of Nedd4-2 in lung epithelial cells causes progressive pulmonary fibrosis in adult mice. Nat Commun 2020; 11:2012. [PMID: 32332792 PMCID: PMC7181726 DOI: 10.1038/s41467-020-15743-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by patchy scarring of the distal lung with limited therapeutic options and poor prognosis. Here, we show that conditional deletion of the ubiquitin ligase Nedd4-2 (Nedd4l) in lung epithelial cells in adult mice produces chronic lung disease sharing key features with IPF including progressive fibrosis and bronchiolization with increased expression of Muc5b in peripheral airways, honeycombing and characteristic alterations in the lung proteome. NEDD4-2 is implicated in the regulation of the epithelial Na+ channel critical for proper airway surface hydration and mucus clearance and the regulation of TGFβ signaling, which promotes fibrotic remodeling. Our data support a role of mucociliary dysfunction and aberrant epithelial pro-fibrotic response in the multifactorial disease pathogenesis. Further, treatment with the anti-fibrotic drug pirfenidone reduced pulmonary fibrosis in this model. This model may therefore aid studies of the pathogenesis and therapy of IPF. Idiopathic pulmonary fibrosis (IPF) is a devastating disease with poor prognosis. Here, the authors show that deficiency of the E3 ubiqutin-protein ligase Nedd4-2 in airway epithelial cells causes IPF-like disease in adult mice. This model may aid studies of the pathogenesis and therapy of IPF.
Collapse
|
61
|
Motz KM, Gelbard A. The role of inflammatory cytokines in the development of idiopathic subglottic stenosis. Transl Cancer Res 2020; 9:2102-2107. [PMID: 35117565 PMCID: PMC8797976 DOI: 10.21037/tcr.2019.12.37] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022]
Abstract
Idiopathic subglottic stenosis (iSGS) is a debilitating extrathoracic obstruction involving the lower laryngeal and upper tracheal airway. It arises without a known antecedent injury or associated disease process. iSGS is a fibrotic disease marked histologically by excessive accumulation of fibrous connective tissue components of the extracellular matrix (ECM, i.e., collagen and fibronectin) in inflamed tissue, which leads to airway obstruction and clinical dyspnea. Diverse diseases in divergent organ systems are associated with fibrosis, suggesting common pathogenic pathways. One of the most common is sustained host inflammation. Recent investigations focusing on the inflammatory response associated with iSGS have sought to characterize the immunophenotype and cytokine profile of the airway scar in iSGS. While the role of the immune response as inciting event in iSGS remains unresolved, the centrality of an active immune response to the observed subglottic tissue remodeling is becoming more defined.
Collapse
Affiliation(s)
- Kevin M Motz
- Department of Otolaryngology & Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Alexander Gelbard
- Department of Otolaryngology & Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
62
|
Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med 2020; 217:e20190103. [PMID: 32997468 PMCID: PMC7062524 DOI: 10.1084/jem.20190103] [Citation(s) in RCA: 676] [Impact Index Per Article: 135.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
TGF-β is extensively implicated in the pathogenesis of fibrosis. In fibrotic lesions, spatially restricted generation of bioactive TGF-β from latent stores requires the cooperation of proteases, integrins, and specialized extracellular matrix molecules. Although fibroblasts are major targets of TGF-β, some fibrogenic actions may reflect activation of other cell types, including macrophages, epithelial cells, and vascular cells. TGF-β–driven fibrosis is mediated through Smad-dependent or non-Smad pathways and is modulated by coreceptors and by interacting networks. This review discusses the role of TGF-β in fibrosis, highlighting mechanisms of TGF-β activation and signaling, the cellular targets of TGF-β actions, and the challenges of therapeutic translation.
Collapse
Affiliation(s)
- Nikolaos Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
63
|
Bisserier M, Milara J, Abdeldjebbar Y, Gubara S, Jones C, Bueno-Beti C, Chepurko E, Kohlbrenner E, Katz MG, Tarzami S, Cortijo J, Leopold J, Hajjar RJ, Sassi Y, Hadri L. AAV1.SERCA2a Gene Therapy Reverses Pulmonary Fibrosis by Blocking the STAT3/FOXM1 Pathway and Promoting the SNON/SKI Axis. Mol Ther 2020; 28:394-410. [PMID: 31879190 PMCID: PMC7001085 DOI: 10.1016/j.ymthe.2019.11.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022] Open
Abstract
Inhibition of pulmonary fibrosis (PF) by restoring sarco/endoplasmic reticulum calcium ATPase 2a isoform (SERCA2a) expression using targeted gene therapy may be a potentially powerful new treatment approach for PF. Here, we found that SERCA2a expression was significantly decreased in lung samples from patients with PF and in the bleomycin (BLM) mouse model of PF. In the BLM-induced PF model, intratracheal aerosolized adeno-associated virus serotype 1 (AAV1) encoding for human SERCA2a (AAV1.hSERCA2a) reduces lung fibrosis and associated vascular remodeling. SERCA2a gene therapy also decreases right ventricular pressure and hypertrophy in both prevention and curative protocols. In vitro, we observed that SERCA2a overexpression inhibits fibroblast proliferation, migration, and fibroblast-to-myofibroblast transition induced by transforming growth factor β (TGF-β1). Thus, pro-fibrotic gene expression is prevented by blocking nuclear factor κB (NF-κB)/interleukin-6 (IL-6)-induced signal transducer and activator of transcription 3 (STAT3) activation. This effect is signaled toward an inhibitory mechanism of small mother against decapentaplegic (SMAD)/TGF-β signaling through the repression of OTU deubiquitinase, ubiquitin aldehyde binding 1 (OTUB1) and Forkhead box M1 (FOXM1). Interestingly, this cross-inhibition leads to an increase of SKI and SnoN expression, an auto-inhibitory feedback loop of TGF-β signaling. Collectively, our results demonstrate that SERCA2a gene transfer attenuates bleomycin (BLM)-induced PF by blocking the STAT3/FOXM1 pathway and promoting the SNON/SKI Axis. Thus, SERCA2a gene therapy may be a potential therapeutic target for PF.
Collapse
Affiliation(s)
- Malik Bisserier
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Javier Milara
- Health Research Institute INCLIVA, Valencia, Spain; Pharmacy Unit, University Clinic Hospital, Valencia, Spain; CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Yassine Abdeldjebbar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah Gubara
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carly Jones
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carlos Bueno-Beti
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elena Chepurko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erik Kohlbrenner
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael G Katz
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sima Tarzami
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, USA
| | - Julio Cortijo
- Health Research Institute INCLIVA, Valencia, Spain; Pharmacy Unit, University Clinic Hospital, Valencia, Spain; CIBERES, Health Institute Carlos III, Valencia, Spain
| | - Jane Leopold
- Cardiovascular Medicine Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Yassine Sassi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lahouaria Hadri
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
64
|
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
|
65
|
Bugyei-Twum A, Ford C, Civitarese R, Seegobin J, Advani SL, Desjardins JF, Kabir G, Zhang Y, Mitchell M, Switzer J, Thai K, Shen V, Abadeh A, Singh KK, Billia F, Advani A, Gilbert RE, Connelly KA. Sirtuin 1 activation attenuates cardiac fibrosis in a rodent pressure overload model by modifying Smad2/3 transactivation. Cardiovasc Res 2019; 114:1629-1641. [PMID: 29800064 DOI: 10.1093/cvr/cvy131] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022] Open
Abstract
Aims Transforming growth factor β1 (TGF-β1) is a prosclerotic cytokine involved in cardiac remodelling leading to heart failure (HF). Acetylation/de-acetylation of specific lysine residues in Smad2/3 has been shown to regulate TGF-β signalling by altering its transcriptional activity. Recently, the lysine de-acetylase sirtuin 1 (SIRT1) has been shown to have a cardioprotective effect; however, SIRT1 expression and activity are paradoxically reduced in HF. Herein, we investigate whether pharmacological activation of SIRT1 would induce cardioprotection in a pressure overload model and assess the impact of SIRT1 activation on TGF-β signalling and the fibrotic response. Methods and results Eight weeks old male C57BL/6 mice were randomized to undergo sham surgery or transverse aortic constriction (TAC) to induce pressure overload. Post-surgery, animals were further randomized to receive SRT1720 or vehicle treatment. Echocardiography, pressure-volume loops, and histological analysis revealed an impairment in cardiac function and deleterious left ventricular remodelling in TAC-operated animals that was improved with SRT1720 treatment. Genetic ablation and cell culture studies using a Smad-binding response element revealed SIRT1 to be a specific target of SRT1720 and identified Smad2/3 as a SIRT1 specific substrate. Conclusion Overall, our data demonstrate that Smad2/3 is a specific SIRT1 target and suggests that pharmacological activation of SIRT1 may be a novel therapeutic strategy to prevent/reverse HF via modifying Smad activity.
Collapse
Affiliation(s)
- Antoinette Bugyei-Twum
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| | - Christopher Ford
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Robert Civitarese
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jessica Seegobin
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jean-Francois Desjardins
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Golam Kabir
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Yanling Zhang
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Jennifer Switzer
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Kerri Thai
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Vanessa Shen
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Armin Abadeh
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Krishna K Singh
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Filio Billia
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Advani
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Richard E Gilbert
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada
| | - Kim A Connelly
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, 209 Victoria Street, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada
| |
Collapse
|
66
|
Kou X, Sun Y, Li S, Bian W, Liu Z, Zhang D, Jiang J. Pharmacology Study of the Multiple Angiogenesis Inhibitor RC28-E on Anti-Fibrosis in a Chemically Induced Lung Injury Model. Biomolecules 2019; 9:biom9110644. [PMID: 31652997 PMCID: PMC6920960 DOI: 10.3390/biom9110644] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 11/30/2022] Open
Abstract
Background: Disease-related injury in any organ triggers a complex cascade of cellular and molecular responses that culminate in tissue fibrosis, inflammation, and angiogenesis simultaneously. Multiple cell angiogenesis is an essential part of the tissue damage response, which is involved in fibrosis development. RC28-E is a novel recombinant dual decoy receptor lgG1 Fc-fusion protein that can block vascular endothelial growth factor (VEGFA), platelet-derived growth factor (PDGF), and fibroblast growth factor-2 (FGF-2) simultaneously. This protein has stepped into clinical trials (NCT03777254) for the treatment of pathological neovascularization-related diseases. Here, we report on the role of RC28-E during anti-fibrosis and its potential multitarget function in regulating fibrosis. Methods: A bleomycin-induced pulmonary fibrosis C57BL/6 mouse model was established. Hematoxylin and eosin staining (HE) and Masson staining (Masson’s) were performed to evaluate the pulmonary fibrosis based on the scoring from, Ashcroft score. Fibrosis related factors and inflammatory cytokines including HYP, α-SMA, procollagen, ICAM, IL-6, IL-1, and TNF-α were also determined at the protein and mRNA levels to characterize the fibrosis. Both mRNA and protein levels of VEGF, FGF, and transforming growth factor (TGF)-β were detected by quantitative real-time PCR (qRT-PCR) and immunohistochemical (IHC) analysis, respectively. Pulmonary fibrosis and related cytokines were re-evaluated in vivo after 3 doses of RC28-E (5 mg/kg, 15 mg/kg, and 50 mg/kg, ip. Tiw × 9) in comparison with a mono-target antagonist treatment (VEGF or FGF blocking). RC28-E attenuated the activation of TGF-β induced fibroblasts in vitro. Expression levels of α-SMA and collagen I, as well as proliferation and migration, were determined with the human skin fibroblast cell line Detroit 551 and primary murine pulmonary fibroblast cells. The mechanism of RC28-E via the TGF-β/Smad pathway was also investigated. Results: RC28-E exhibits significant anti-fibrosis effects on Idiopathic pulmonary fibrosis (IPF) in vivo. Moreover, TGF-β induced fibroblast activation in vitro via the inhibition of the TGF-β downstream Smad pathway, thus providing potential therapeutics for clinical disease-related fibrosis-like IPF as well as chemotherapy-induced fibrosis in cancer therapy.
Collapse
Affiliation(s)
- Xiangying Kou
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, China.
| | - Yeying Sun
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, China.
| | - Shenjun Li
- RemeGen Co., Ltd., Yantai 264006, China.
| | - Weihua Bian
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, China.
| | - Zhihao Liu
- RemeGen Co., Ltd., Yantai 264006, China.
| | - Daolai Zhang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, China.
| | - Jing Jiang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, China.
| |
Collapse
|
67
|
Fang X, Hu X, Zheng Z, Tao K, Wang H, Guan H, Shi J, Ji P, Cai W, Bai X, Zhu X, Han J, Liu J, Hu D. Smad interacting protein 1 influences transforming growth factor-β 1/Smad signaling in extracellular matrix protein production and hypertrophic scar formation. J Mol Histol 2019; 50:503-514. [PMID: 31595443 DOI: 10.1007/s10735-019-09844-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/23/2018] [Indexed: 02/07/2023]
Abstract
The transforming growth factor (TGF)-β/Smad signal transduction pathway is closely associated with hypertrophic scar (HS) formation. Smad interacting protein 1 (SIP1) is a cytoplasmic protein that efficiently regulates Smad2-/3-dependent signaling within the TGF-β1 pathway. SIP1 influences collagen synthesis in the HS through a heretofore unknown mechanism. This study investigated the role of the SIP1-mediated TGF-β1/Smad signaling pathway in extracellular matrix (ECM) protein production and hypertrophic scarring. SIP1 expression was markedly lower in HS vs. normal skin (NS) tissue, and α-smooth muscle actin (α-SMA) content and collagen I/III (Col I/III) synthesis were inversely correlated with SIP1 expression. Furthermore, SIP1 inhibited Smad2/3 phosphorylation in vitro, and improved the collagen-based architecture of the scar while reducing collagen expression and overall scar formation in a rabbit ear model of HS. Based on these findings, we propose that SIP1 acts as a molecular modulator capable of altering Smad2-/3-facilitated signaling through the control of Smad phosphorylation, thus inhibiting α-SMA and collagen upregulation in fibroblasts and, ultimately, HS formation. The low SIP1 content in scar tissue also suggests that SIP1 (and positive regulation thereof) is a prospective target for selective HS drug therapy.
Collapse
Affiliation(s)
- Xiaobing Fang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Xiaolong Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Ke Tao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Hongtao Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Peng Ji
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Xiongxiang Zhu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China
| | - Jiaqi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
68
|
Zhou Z, Kandhare AD, Kandhare AA, Bodhankar SL. Hesperidin ameliorates bleomycin-induced experimental pulmonary fibrosis via inhibition of TGF-beta1/Smad3/AMPK and IkappaBalpha/NF-kappaB pathways. EXCLI JOURNAL 2019; 18:723-745. [PMID: 31611754 PMCID: PMC6785776 DOI: 10.17179/excli2019-1094] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022]
Abstract
Bleomycin (BLM) is a chemotherapeutic agent which is associated with Idiopathic pulmonary fibrosis (IPF) due to its chronic administration. Hesperidin, a bioflavonoid has been reported to possess antioxidant, anti-inflammatory, wound healing, and antiapoptotic potential. To evaluate the therapeutic potential of hesperidin against BLM-induced pulmonary fibrosis and decipher its possible mechanism of action. Intraperitoneal administration of BLM (6 IU/kg) caused induction of IPF in Sprague-Dawley rats. Rats were treated with hesperidin (25, 50, and 100 mg/kg, p.o.) for 28 days, followed by estimation of various parameters in bronchoalveolar lavage fluid (BALF) and lung. Hesperidin (50 and 100 mg/kg) administration significantly ameliorated (p < 0.05) alterations induced by BLM in lung index, percent oxygen saturation, serum ALP and LDH levels, BALF differential cell count, and lung function test. Elevated levels of oxido-nitrosative stress, hydroxyproline, and myeloperoxidase levels in BALF and lung were significantly decreased by hesperidin on day 14. Hesperidin significantly inhibited BLM-induced down-regulated lung Nrf2 and HO-1 as well as up-regulated TNF-α, IL-1β, IL-6, collagen-1, TGF-β, and Smad-3 mRNA expressions. Western blot analysis showed that alteration in lung NF-κB, IκBα, AMPK, and PP2C-α protein expressions were ameliorated by hesperidin on day 28. Furthermore, BLM induced histological and ultrastructural aberrations in the lung which were attenuated by hesperidin treatment. Hesperidin alleviates BLM-induced IPF via inhibition of TGF-β1/Smad3/AMPK and IκBα/NF-κB pathways which in turn ameliorate the modulation of oxido-inflammatory markers (Nrf2 and HO-1) and pro-inflammatory markers (TNF-α, IL-1β, and IL-6) to reduce collagen deposition during pulmonary fibrosis. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Zheng Zhou
- Department of Respiratory Medicine, the Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, Henan Province, 450014, China
| | - Amit D Kandhare
- Department of Pharmacology, Center for Advanced Research in Pharmaceutical Sciences, Bharati Vidyapeeth Deemed University, Poona College of Pharmacy, Pune-411 038, India
| | - Anwesha A Kandhare
- Department of Pharmacology, Center for Advanced Research in Pharmaceutical Sciences, Bharati Vidyapeeth Deemed University, Poona College of Pharmacy, Pune-411 038, India
| | - Subhash L Bodhankar
- Department of Pharmacology, Center for Advanced Research in Pharmaceutical Sciences, Bharati Vidyapeeth Deemed University, Poona College of Pharmacy, Pune-411 038, India
| |
Collapse
|
69
|
Meng M, Tan J, Chen W, Du Q, Xie B, Wang N, Zhu H, Wang K. The Fibrosis and Immunological Features of Hypochlorous Acid Induced Mouse Model of Systemic Sclerosis. Front Immunol 2019; 10:1861. [PMID: 31481954 PMCID: PMC6710365 DOI: 10.3389/fimmu.2019.01861] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/23/2019] [Indexed: 01/22/2023] Open
Abstract
Fibrotic animal models are critical for the pathogenesis investigations and drug explorations in systemic sclerosis (SSc). The bleomycin (BLM)-induced mouse model is the classical and most widely used fibrosis model. However, traditional subcutaneous injection of BLM rarely induced diffuse skin and lung lesions. Hypochlorous acid (HOCl)-induced mice are a more representative model that have diffuse cutaneous lesions, lung fibrosis and renal involvement. However, the fibrotic and immunological features of this model are not fully elucidated. Here, we injected BALB/c mice subcutaneously with HOCl used at different concentrations of HOCl (1:55, 1:70, and 1:110 NaClO: KH2PO4, hereafter named HOCl55, HOCl70, and HOCl110, respectively) for 6 weeks to induce fibrosis, and also used HOCl110 at different time course (4, 5, and 6 weeks). Morphological changes were observed via HE and Masson's trichrome staining. Immunohistochemistry or real-time PCR was used to detect inflammatory infiltrates, important fibrosis pathways and pro-inflammatory mediator expression. Flow cytometry was used to detect the alteration of immune cells in mouse spleen. Skin and lung fibrosis were most obvious in the HOCl55 group compared to lower concentration groups. In the HOCl110 group, dominant inflammatory infiltrates were found after 5 weeks, and significant fibrosis was found after 6 weeks. Then we explored the fibrosis and immunological profiles in the HOCl110 (6 weeks) group. Important fibrosis pathway proteins such as TGF-β, NF-κB, Smad3, p-Smad3, STAT3, and p-STAT3 were significantly elevated at week 6 in the HOCl110 group. Increased infiltration of CD4+T cells, CD8+T cells, CD20+B cells, and myofibroblasts was found both in skin and lung tissues. However, decreased CD4+T cells, CD8+T cells, monocytes and macrophages and increased CD19+B cells were found in the spleen tissues. The mRNA expression of fibrosis mediators such as IL-1β, IL-6, IL-17, IL-33, TNF-α, and CTGF was also upregulated in skin and lung tissues. In conclusion, HOCl induced fibrosis mouse model displayed systemic immune cell infiltration, pro-inflammatory mediator release, vasculopathy and fibrosis, which better mimicked human SSc than BLM-induced mice.
Collapse
Affiliation(s)
- Meng Meng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jieqiong Tan
- The Center for Medical Genetics, School of Life Science, Central South University, Changsha, China
| | - Weilin Chen
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Du
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, China.,Department of Laboratory Animals, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
70
|
Bang IJ, Kim HR, Jeon Y, Jeong MH, Park YJ, Kwak JH, Chung KH. β-Peltoboykinolic Acid from Astilbe rubra Attenuates TGF-β1-Induced Epithelial-to-Mesenchymal Transitions in Lung Alveolar Epithelial Cells. Molecules 2019; 24:molecules24142573. [PMID: 31311194 PMCID: PMC6680586 DOI: 10.3390/molecules24142573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/09/2019] [Accepted: 07/14/2019] [Indexed: 01/04/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is increasingly recognized as contributing to the pathogenesis of idiopathic pulmonary fibrosis. Therefore, novel plant-based natural, active compounds have been sought for the treatment of fibrotic EMT. The aim of the present study was to investigate the inhibitory effects of Astilbe rubra on TGF-β1-induced EMT in lung alveolar epithelial cells (A549). A. rubra was subjected to extraction using 70% ethanol (ARE), and ethanol extracts of the aerial part and that of the rhizome were further partitioned using various solvents. Protein expression and cell motility were investigated to evaluate the inhibitory effects of ARE on EMT. EMT occurred in A549 cells treated with TGF-β1, but was prevented by co-treatment with ARE. The dichloromethane fractions showed the strongest inhibitory effect on TGF-β1-induced EMT. β-Peltoboykinolic acid was isolated from the dichloromethane fractions of A. rubra by activity-oriented isolation. β-Peltoboykinolic acid not only attenuated TGF-β1-induced EMT, but also the overproduction of extracellular matrix components including type I collagen and fibronectin. The Smad pathway activated by TGF-β1 was inhibited by co-treatment with β-peltoboykinolic acid. Taken together, these results indicate that β-peltoboykinolic acid from A. rubra and dichloromethane fractions shows potential as an antifibrotic agent in A549 cells treated with TGF-β1.
Collapse
Affiliation(s)
- In Jae Bang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Ha Ryong Kim
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Korea
| | - Yukyoung Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Mi Ho Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Yong Joo Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jong Hwan Kwak
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Kyu Hyuck Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| |
Collapse
|
71
|
Surfactant dysfunction and alveolar collapse are linked with fibrotic septal wall remodeling in the TGF-β1-induced mouse model of pulmonary fibrosis. J Transl Med 2019; 99:830-852. [PMID: 30700849 DOI: 10.1038/s41374-019-0189-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 11/20/2018] [Accepted: 12/17/2018] [Indexed: 11/08/2022] Open
Abstract
In human idiopathic pulmonary fibrosis (IPF), collapse of distal airspaces occurs in areas of the lung not (yet) remodeled. Mice lungs overexpressing transforming growth factor-β1 (TGF-β1) recapitulate this abnormality: surfactant dysfunction results in alveolar collapse preceding fibrosis and loss of alveolar epithelial type II (AE2) cells' apical membrane surface area. Here we examined whether surfactant dysfunction-related alveolar collapse due to TGF-β1 overexpression is linked to septal wall remodeling and AE2 cell abnormalities. Three and 6 days after gene transfer of TGF-β1, mice received either intratracheal surfactant (Surf-groups: Curosurf®, 100 mg/kg bodyweight) or 0.9% NaCl (Saline-groups). On days 7 (D7) and 14 (D14), lung mechanics were assessed followed by design-based stereology at light and electron microscopic level to quantify structures. Compared with Saline, Surf showed significantly improved tissue elastance, increased numbers of open alveoli, as well as reduced alveolar size heterogeneity on D7. Deterioration in lung mechanics was highly correlated to the loss of open alveoli. On D14, lung mechanics, number of open alveoli, and alveolar size heterogeneity remained significantly improved in the Surf-group. Volumes of extracellular matrix and collagen fibrils in septal walls were significantly reduced, whereas the apical membrane surface area of AE2 cells was increased in Surf compared with Saline. In remodeled tissue with collapsed alveoli, three-dimensional reconstruction of AE2 cells based on scanning electron microscopy array tomography revealed that AE2 cells were trapped without contact to airspaces in the TGF-β1 mouse model. Similar observations were made in human IPF. Based on correlation analyses, the number of open alveoli and of alveolar size heterogeneity were highly linked with the loss of apical membrane surface area of AE2 cells and deposition of collagen fibrils in septal walls on D14. In conclusion, surfactant replacement therapy stabilizes alveoli and prevents extracellular matrix deposition in septal walls in the TGF-β1 model.
Collapse
|
72
|
Sundarakrishnan A, Zukas H, Coburn J, Bertini BT, Liu Z, Georgakoudi I, Baugh L, Dasgupta Q, Black LD, Kaplan DL. Bioengineered in Vitro Tissue Model of Fibroblast Activation for Modeling Pulmonary Fibrosis. ACS Biomater Sci Eng 2019; 5:2417-2429. [PMID: 33405750 DOI: 10.1021/acsbiomaterials.8b01262] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a complex disease of unknown etiology with no current curative treatment. Modeling pulmonary fibrotic (PF) tissue has the potential to improve our understanding of IPF disease progression and treatment. Rodent animal models do not replicate human fibroblastic foci (Hum-FF) pathology, and current iterations of in vitro model systems (e.g., collagen hydrogels, polyacrylamide hydrogels, and fibrosis-on-chip systems) are unable to replicate the three-dimensional (3D) complexity and biochemical composition of human PF tissue. Herein, we fabricated a 3D bioengineered pulmonary fibrotic (Eng-PF) tissue utilizing cell laden silk collagen type I dityrosine cross-linked hydrogels and Flexcell bioreactors. We show that silk collagen type I hydrogels have superior stability and mechanical tunability compared to other hydrogel systems. Using customized Flexcell bioreactors, we reproduced Hum-FF-like pathology with airway epithelial and microvascular endothelial cells. Eng-PF tissues can model myofibroblast differentiation and permit evaluation of antifibrotic drug treatments. Further, Eng-PF tissues could be used to model different facets of IPF disease, including epithelial injury with the addition of bleomycin and cellular recruitment by perfusion of cells through the hydrogel microchannel.
Collapse
Affiliation(s)
- Aswin Sundarakrishnan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Heather Zukas
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Jeannine Coburn
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Worcester, Massachusetts 01605, United States
| | - Brian T Bertini
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Zhiyi Liu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, Massachusetts 02114, United States
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Lauren Baugh
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Queeny Dasgupta
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States.,Department of Cell, Molecular & Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, 136 Harrison Avenue, Boston, Massachusetts 02111, United States
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
73
|
Xue K, Zhang J, Li C, Li J, Wang C, Zhang Q, Chen X, Yu X, Sun L, Yu X. The role and mechanism of transforming growth factor beta 3 in human myocardial infarction-induced myocardial fibrosis. J Cell Mol Med 2019; 23:4229-4243. [PMID: 30983140 PMCID: PMC6533491 DOI: 10.1111/jcmm.14313] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/11/2019] [Accepted: 03/04/2019] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor beta (TGFβ) plays a crucial role in tissue fibrosis. A number of studies have shown that TGFβ3 significantly attenuated tissue fibrosis. However, the mechanism involved in this effect is poorly understood. In this study we found that the expression level of TGFβ3 was higher in human myocardial infarction (MI) tissues than in normal tissues, and interestingly, it increased with the development of fibrosis post‐myocardial infarction (post‐MI). In vitro, human cardiac fibroblasts (CFs) were incubated with angiotensin II (Ang II) to mimic the ischaemic myocardium microenvironment and used to investigate the anti‐fibrotic mechanism of TGFβ3. Then, fibrosis‐related proteins were detected by Western blot. It was revealed that TGFβ3 up‐regulation attenuated the proliferation, migration of human CFs and the expression of collagens, which are the main contributors to fibrosis, promoted the phenotype shift and the cross‐linking of collagens. Importantly, the expression of collagens was higher in the si‐smad7 groups than in the control groups, while silencing smad7 increased the phosphorylation level of the TGFβ/smad signalling pathway. Collectively, these results indicated that TGFβ3 inhibited fibrosis via the TGFβ/smad signalling pathway, possibly attributable to the regulation of smad7, and that TGFβ3 might serve as a potential therapeutic target for myocardial fibrosis post‐MI.
Collapse
Affiliation(s)
- Ke Xue
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jun Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Cong Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jing Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Cong Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xianlu Chen
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaotang Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lei Sun
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiao Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
74
|
Kinoshita T, Goto T. Molecular Mechanisms of Pulmonary Fibrogenesis and Its Progression to Lung Cancer: A Review. Int J Mol Sci 2019; 20:ijms20061461. [PMID: 30909462 PMCID: PMC6471841 DOI: 10.3390/ijms20061461] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is defined as a specific form of chronic, progressive fibrosing interstitial pneumonia of unknown cause, occurring primarily in older adults, and limited to the lungs. Despite the increasing research interest in the pathogenesis of IPF, unfavorable survival rates remain associated with this condition. Recently, novel therapeutic agents have been shown to control the progression of IPF. However, these drugs do not improve lung function and have not been tested prospectively in patients with IPF and coexisting lung cancer, which is a common comorbidity of IPF. Optimal management of patients with IPF and lung cancer requires understanding of pathogenic mechanisms and molecular pathways that are common to both diseases. This review article reflects the current state of knowledge regarding the pathogenesis of pulmonary fibrosis and summarizes the pathways that are common to IPF and lung cancer by focusing on the molecular mechanisms.
Collapse
Affiliation(s)
- Tomonari Kinoshita
- Division of General Thoracic Surgery, Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 1608582, Japan.
| | - Taichiro Goto
- Lung Cancer and Respiratory Disease Center, Yamanashi Central Hospital, Kofu, Yamanashi 4008506, Japan.
| |
Collapse
|
75
|
Hoegl S, Burns N, Angulo M, Francis D, Osborne CM, Mills TW, Blackburn MR, Eltzschig HK, Vohwinkel CU. Capturing the multifactorial nature of ARDS - "Two-hit" approach to model murine acute lung injury. Physiol Rep 2019; 6:e13648. [PMID: 29595879 PMCID: PMC5875538 DOI: 10.14814/phy2.13648] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/12/2018] [Accepted: 02/14/2018] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory distress syndrome (ARDS) presents typically with an initializing event, followed by the need for mechanical ventilation. Most animal models of ALI are limited by the fact that they focus on a singular cause of acute lung injury (ALI) and therefore fail to mimic the complex, multifactorial pathobiology of ARDS. To better capture this scenario, we provide a comprehensive characterization of models of ALI combining two injuries: intra tracheal (i.t.) instillation of LPS or hypochloric acid (HCl) followed by ventilator‐induced lung injury (VILI). We hypothesized, that mice pretreated with LPS or HCl prior to VILI and thus receiving a (“two‐hit injury”) will sustain a superadditive lung injury when compared to VILI. Mice were allocated to following treatment groups: control with i.t. NaCl, ventilation with low peak inspiratory pressure (PIP), i.t. HCl, i.t. LPS, VILI (high PIP), HCl i.t. followed by VILI and LPS i.t. followed by VILI. Severity of injury was determined by protein content and MPO activity in bronchoalveolar lavage (BAL), the expression of inflammatory cytokines and histopathology. Mice subjected to VILI after HCl or LPS instillation displayed augmented lung injury, compared to singular lung injury. However, mice that received i.t. LPS prior to VILI showed significantly increased inflammatory lung injury compared to animals that underwent i.t. HCl followed by VILI. The two‐hit lung injury models described, resulting in additive but differential acute lung injury recaptures the clinical relevant multifactorial etiology of ALI and could be a valuable tool in translational research.
Collapse
Affiliation(s)
- Sandra Hoegl
- Organ Protection Program, School of Medicine, Department of Anesthesiology, University of Colorado, Aurora, Colorado.,Developmental Lung Biology, Cardio Vascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatric Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Nana Burns
- Feinberg School of Medicine, Division of Pulmonary and Critical Care, Northwestern University, Chicago, Illinois
| | - Martín Angulo
- Department of Respiratory Therapy, Colorado Children's Hospital, Aurora, Colorado
| | - Daniel Francis
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center Houston, Houston, Texas
| | - Christopher M Osborne
- Feinberg School of Medicine, Division of Pulmonary and Critical Care, Northwestern University, Chicago, Illinois
| | - Tingting W Mills
- Department of Anesthesiology, University Hospital of Ludwig-Maximilians-University, Munich, Germany
| | - Michael R Blackburn
- Department of Anesthesiology, University Hospital of Ludwig-Maximilians-University, Munich, Germany
| | - Holger K Eltzschig
- Organ Protection Program, School of Medicine, Department of Anesthesiology, University of Colorado, Aurora, Colorado
| | - Christine U Vohwinkel
- Organ Protection Program, School of Medicine, Department of Anesthesiology, University of Colorado, Aurora, Colorado.,Feinberg School of Medicine, Division of Pulmonary and Critical Care, Northwestern University, Chicago, Illinois
| |
Collapse
|
76
|
Yun SM, Kim SH, Kim EH. The Molecular Mechanism of Transforming Growth Factor-β Signaling for Intestinal Fibrosis: A Mini-Review. Front Pharmacol 2019; 10:162. [PMID: 30873033 PMCID: PMC6400889 DOI: 10.3389/fphar.2019.00162] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
Inflammatory bowel disease is known as the most chronic inflammatory disorder in colon, which subsequently progresses to intestinal obstruction and fistula formation. Many studies to date for the treatment of IBD have been focused on inflammation. However, most of the anti-inflammatory agents do not have anti-fibrotic effects and could not relieve intestinal stricture in IBD patients. Because preventing or reversing intestinal fibrosis in IBD is a major therapeutic target, we analyzed the papers focusing on TGF-β signaling in intestinal fibrosis. TGF-β is a good candidate to treat the intestinal fibrosis in IBD which involves TGF-β signaling pathway, EMT, EndMT, ECM, and other regulators. Understanding the mechanism involved in TGF-β signaling will contribute to the treatment and diagnosis of intestinal fibrosis occurring in IBD as well as the understanding of the molecular mechanisms underlying the pathogenesis.
Collapse
Affiliation(s)
- Sun-Mi Yun
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Seok-Ho Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam, South Korea
| |
Collapse
|
77
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
78
|
An C, Jia L, Wen J, Wang Y. Targeting Bone Marrow-Derived Fibroblasts for Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:305-322. [DOI: 10.1007/978-981-13-8871-2_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
79
|
LncRNAs in TGF-β-Driven Tissue Fibrosis. Noncoding RNA 2018; 4:ncrna4040026. [PMID: 30287731 PMCID: PMC6315857 DOI: 10.3390/ncrna4040026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/22/2018] [Accepted: 09/28/2018] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a crucial mediator in tissue fibrosis that promotes accumulation of extracellular matrix (ECM), myofibroblasts to epithelial–mesenchymal transition (EMT), endothelial-mesenchymal transition (EndoMT), and apoptosis via canonical and noncanonical signaling pathways. In the past decades, a number of microRNAs have been reported to participate in TGF-β-mediated tissue scarring; however, the roles of long noncoding RNAs (lncRNAs) in fibrogenesis remain largely unknown. Recently, emerging evidence has shown that lncRNAs are involved in the development of different diseases, including cancer, autoimmune diseases, cardiovascular diseases, and fibrotic diseases. In this review, we summarize the current updates of lncRNAs in TGF-β1-driven tissue fibrosis and discuss their therapeutic potential for the treatment of chronic fibrotic diseases.
Collapse
|
80
|
Li X, Lu C, Liu S, Shuaishuai Liu, Su C, Xiao T, Bi Z, Sheng P, Huang M, Liu X, Wei Y, Zhao L, Miao S, Mao J, Huang K, Gao S, Liu N, Qi M, Liu T, Qin S, Wei L, Sun T, Ning W, Yang G, Zhou H, Yang C. Synthesis and discovery of a drug candidate for treatment of idiopathic pulmonary fibrosis through inhibition of TGF-β1 pathway. Eur J Med Chem 2018; 157:229-247. [PMID: 30096654 DOI: 10.1016/j.ejmech.2018.07.074] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 10/28/2022]
Abstract
In this study, anti-IPF lead compounds 42 and 44, derived from natural sesquiterpene lactones Isoalantolactone and alantolactone, were discovered by screening from a high-throughput TGF-β1 reporter luciferase assay. Notably, they could reduce the myofibroblast activation and extracellular matrix deposition both in vitro and in vivo. Additionally, compounds 42 and 44 could significantly attenuate bleomycin-induced pulmonary fibrosis in mice. Further validation of pharmacokinetics study and toxicity evaluation indicated that compound 44 might be a promising anti-IPF drug candidate.
Collapse
Affiliation(s)
- Xiaohe Li
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Cheng Lu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Shuangwei Liu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Shuaishuai Liu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Chengcheng Su
- Department of Respiratory and Critical Care Medicine, Pingjin Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin, 300162, People's Republic of China
| | - Ting Xiao
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Zhun Bi
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Pengzhen Sheng
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Mengying Huang
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Xinhua Liu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Yujiao Wei
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Lin Zhao
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Shengxiang Miao
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Jiahe Mao
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Kai Huang
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Shaoyan Gao
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Ning Liu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Min Qi
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Tongtong Liu
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Shuanglin Qin
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Luqing Wei
- Department of Respiratory and Critical Care Medicine, Pingjin Hospital, Logistics University of Chinese People's Armed Police Forces, Tianjin, 300162, People's Republic of China
| | - Tao Sun
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Wen Ning
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China
| | - Guang Yang
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China.
| | - Honggang Zhou
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China.
| | - Cheng Yang
- College of Pharmacy, The State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, People's Republic of China.
| |
Collapse
|
81
|
Saito A, Horie M, Nagase T. TGF-β Signaling in Lung Health and Disease. Int J Mol Sci 2018; 19:ijms19082460. [PMID: 30127261 PMCID: PMC6121238 DOI: 10.3390/ijms19082460] [Citation(s) in RCA: 338] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/18/2018] [Accepted: 08/18/2018] [Indexed: 01/05/2023] Open
Abstract
Transforming growth factor (TGF)-β is an evolutionarily conserved pleiotropic factor that regulates a myriad of biological processes including development, tissue regeneration, immune responses, and tumorigenesis. TGF-β is necessary for lung organogenesis and homeostasis as evidenced by genetically engineered mouse models. TGF-β is crucial for epithelial-mesenchymal interactions during lung branching morphogenesis and alveolarization. Expression and activation of the three TGF-β ligand isoforms in the lungs are temporally and spatially regulated by multiple mechanisms. The lungs are structurally exposed to extrinsic stimuli and pathogens, and are susceptible to inflammation, allergic reactions, and carcinogenesis. Upregulation of TGF-β ligands is observed in major pulmonary diseases, including pulmonary fibrosis, emphysema, bronchial asthma, and lung cancer. TGF-β regulates multiple cellular processes such as growth suppression of epithelial cells, alveolar epithelial cell differentiation, fibroblast activation, and extracellular matrix organization. These effects are closely associated with tissue remodeling in pulmonary fibrosis and emphysema. TGF-β is also central to T cell homeostasis and is deeply involved in asthmatic airway inflammation. TGF-β is the most potent inducer of epithelial-mesenchymal transition in non-small cell lung cancer cells and is pivotal to the development of tumor-promoting microenvironment in the lung cancer tissue. This review summarizes and integrates the current knowledge of TGF-β signaling relevant to lung health and disease.
Collapse
Affiliation(s)
- Akira Saito
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
- Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| | - Masafumi Horie
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
82
|
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: 8.0] [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]
|
83
|
Gao J, Ye J, Ying Y, Lin H, Luo Z. Negative regulation of TGF-β by AMPK and implications in the treatment of associated disorders. Acta Biochim Biophys Sin (Shanghai) 2018; 50:523-531. [PMID: 29873702 DOI: 10.1093/abbs/gmy028] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 01/18/2023] Open
Abstract
Transforming growth factor beta (TGF-β) regulates a large number of biological processes, including proliferation, differentiation, immune response, and development. In addition, TGF-β plays important roles in some pathological processes, for instance, it is upregulated and activated in fibrosis and advanced cancer. Adenosine monophosphate-activated protein kinase (AMPK) acts as a fuel gauge that is activated when cells sense shortage of ATP and increase in AMP or AMP:ATP ratio. Activation of AMPK slows down anabolic processes and stimulates catabolic processes, leading to increased production of ATP. Furthermore, the functions of AMPK have been extended beyond energy homeostasis. In fact, AMPK has been shown to exert a tumor suppressive effect. Recent studies have demonstrated negative impacts of AMPK on TGF-β function. Therefore, in this review, we will discuss the differences in the biological functions of TGF-β and AMPK, and some pathological processes such as fibrosis, epithelial-mesenchymal transition (EMT) and cancer metastasis, as well as angiogenesis and heterotopic ossifications where TGF-β and AMPK exert opposite effects.
Collapse
Affiliation(s)
- Jiayu Gao
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
- Department of Pathology, Schools of Basic Sciences, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Jinhui Ye
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| | - Zhijun Luo
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University Jiangxi Medical College, Nanchang 330000, China
- Department of Pathology, Schools of Basic Sciences, Nanchang University Jiangxi Medical College, Nanchang 330000, China
| |
Collapse
|
84
|
Divya T, Velavan B, Sudhandiran G. Regulation of Transforming Growth Factor-β/Smad-mediated Epithelial-Mesenchymal Transition by Celastrol Provides Protection against Bleomycin-induced Pulmonary Fibrosis. Basic Clin Pharmacol Toxicol 2018; 123:122-129. [PMID: 29394529 DOI: 10.1111/bcpt.12975] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/24/2018] [Indexed: 12/21/2022]
Abstract
The respiratory disease pulmonary fibrosis (PF), which is characterized by scar formation throughout the lung, imposes a serious health burden. No effective drug without side effects has been proven to prevent this fatal lung disease. In this context, this study was undertaken to elucidate the protective effect of celastrol, a quinine methide pentacyclic triterpenoid from a Chinese medicinal plant 'thunder god vine' against bleomycin (BLM)-induced PF. We also attempted to study how the cytokine transforming growth factor-β (TGF-β) stimulates fibrosis through the induction of epithelial-mesenchymal transition (EMT) and the role of celastrol in regulating EMT. TGF-β (5 ng/ml) was administered to human alveolar epithelial adenocarcinoma A549 cells to induce fibrotic response in cells. Induction of EMT was analysed in cells through morphological analysis and expression of epithelial and mesenchymal markers by Western blotting. Bleomycin at a concentration of 3 U/Kg b.w was used to induce fibrosis in adult male rat lungs. Celastrol (5 mg/kg b.w) was given to rats twice a week after BLM administration for a period of 28 days. Western blot and immunofluorescence analyses were performed with lung tissue sample to find out the potential of celastrol in regulating EMT during the progression of fibrosis. TGF-β induces EMT in A549 cells as demonstrated by changes in epithelial cell morphology and expression of epithelial and mesenchymal marker proteins. The expressions of epithelial marker proteins E-cadherin and claudin were found to be reduced in the BLM-induced group of rats. Expression of mesenchymal markers, such as N-cadherin, snail, slug, vimentin and β-catenin, was enhanced in BLM-induced rat lungs. Celastrol reverts these cellular changes in rat lungs, and it was found that celastrol regulates EMT through the inhibition of heat shock protein 90 (HSP 90). Together, the results indicate that EMT is a crucial phenomenon for the progression of fibrosis, and celastrol provides protection against PF through the regulation of EMT.
Collapse
Affiliation(s)
- Thomas Divya
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Chennai, India
| | | | - Ganapasam Sudhandiran
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Chennai, India
| |
Collapse
|
85
|
Izzo R, Bevivino G, De Simone V, Sedda S, Monteleone I, Marafini I, Di Giovangiulio M, Rizzo A, Franzè E, Colantoni A, Ortenzi A, Monteleone G. Knockdown of Smad7 With a Specific Antisense Oligonucleotide Attenuates Colitis and Colitis-Driven Colonic Fibrosis in Mice. Inflamm Bowel Dis 2018; 24:1213-1224. [PMID: 29668937 DOI: 10.1093/ibd/izy062] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND In Crohn's disease (CD), the pathogenic immune response is associated with high Smad7, an inhibitor of TGF-β1 signaling. Smad7 knockdown with Mongersen, a specific antisense oligonucleotide-containing compound, restores TGF-β1 activity leading to inhibition of inflammatory signals and associates with clinical benefit in CD patients. As TGF-β1 is pro-fibrogenic, it remains unclear whether Mongersen-induced Smad7 inhibition increases the risk of intestinal fibrosis. We assessed the impact of Smad7 inhibition on the course of colitis-driven intestinal fibrosis in mice. METHODS BALB/c mice were rectally treated with increasing doses of trinitrobenzene sulfonic acid (TNBS) for 8 or 12 weeks. The effect of oral Smad7 antisense or control oligonucleotide, administered to mice starting from week 5 or week 8, respectively, on mucosal inflammation and colitis-associated colonic fibrosis was assessed. Mucosal samples were analyzed for Smad7 by immunoblotting and immunohistochemistry, TGF-β1 by enzyme-linked immunosorbent assay, and collagen by immunohistochemistry. RESULTS TNBS-induced chronic colitis was associated with colonic deposition of collagen I and fibrosis, which were evident at week 8 and became more pronounced at week 12. TNBS treatment enhanced Smad7 in both colonic epithelial and lamina propria mononuclear cells. Colitic mice treated with Smad7 antisense oligonucleotide exhibited reduced signs of colitis, less collagen deposition, and diminished fibrosis. These findings were associated with diminished synthesis of TGF-β1 and reduced p-Smad3 protein expression. CONCLUSION Attenuation of colitis with Smad7 antisense oligonucleotide limits development of colonic fibrosis.
Collapse
Affiliation(s)
- Roberta Izzo
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Gerolamo Bevivino
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Veronica De Simone
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Silvia Sedda
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Ivan Monteleone
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy.,Dipartimento di Biomedicina e Prevenzione, Università Tor Vergata, Rome, Italy
| | - Irene Marafini
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | | | - Angelamaria Rizzo
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Eleonora Franzè
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Alfredo Colantoni
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | - Angela Ortenzi
- Dipartimento di Medicina dei Sistemi, Università Tor Vergata, Rome, Italy
| | | |
Collapse
|
86
|
Misa K, Tanino Y, Wang X, Nikaido T, Kikuchi M, Sato Y, Togawa R, Tanino M, Tanaka S, Kadomatsu K, Munakata M. Involvement of midkine in the development of pulmonary fibrosis. Physiol Rep 2018; 5:5/16/e13383. [PMID: 28811360 PMCID: PMC5582267 DOI: 10.14814/phy2.13383] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/12/2017] [Accepted: 07/14/2017] [Indexed: 12/20/2022] Open
Abstract
Midkine is a low‐molecular‐weight heparin‐binding protein that is strongly expressed mainly in the midgestation period and has various physiological activities such as in development and cell migration. Midkine has been reported to be strongly expressed in cancer cells and in inflammation and repair processes, and to be involved in the pathogenesis of various diseases. However, its role in the lung is poorly understood. In this study, we analyzed the clinical characteristics of idiopathic pulmonary fibrosis patients in relation to midkine expression and used a mouse bleomycin‐induced pulmonary fibrosis model to investigate the role of midkine in pulmonary fibrosis. In the idiopathic pulmonary fibrosis patients, the serum midkine level was significantly higher than in healthy subjects, and midkine levels in the serum and bronchoalveolar lavage (BAL) fluid correlated positively with the percentage of inflammatory cells in the BAL fluid. In wild‐type mice, intratracheal bleomycin administration increased midkine expression in lung tissue. Additionally, compared with wild‐type mice, midkine‐deficient mice showed low expression of both collagen and α‐smooth muscle actin, as well as a low value for the pathological lung fibrosis score after bleomycin administration. Furthermore, the total cell count and lymphocyte percentage in the BAL fluid, as well as TNF‐α and transforming growth factor‐β expression in lung tissue, were significantly lower in the midkine‐deficient mice compared with wild‐type mice. These results suggest that midkine is involved in the development of pulmonary fibrosis by regulating inflammatory cell migration into the lung, and TNF‐α and transforming growth factor‐β expression.
Collapse
Affiliation(s)
- Kenichi Misa
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yoshinori Tanino
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Xintao Wang
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takefumi Nikaido
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masami Kikuchi
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuki Sato
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Ryuichi Togawa
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Mishie Tanino
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University School of Medicine, Nagoya, Japan
| | - Mitsuru Munakata
- Department of Pulmonary Medicine, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
87
|
Pattarayan D, Sivanantham A, Bethunaickan R, Palanichamy R, Rajasekaran S. Tannic acid modulates fibroblast proliferation and differentiation in response to pro‐fibrotic stimuli. J Cell Biochem 2018; 119:6732-6742. [DOI: 10.1002/jcb.26866] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 03/15/2018] [Indexed: 12/25/2022]
Affiliation(s)
| | - Ayyanar Sivanantham
- Department of BiotechnologyAnna UniversityBIT‐CampusTiruchirappalliTamil NaduIndia
| | - Ramalingam Bethunaickan
- Department of ImmunologyNational Institute for Research in TuberculosisChennaiTamil NaduIndia
| | - Rajaguru Palanichamy
- Department of BiotechnologyAnna UniversityBIT‐CampusTiruchirappalliTamil NaduIndia
| | - Subbiah Rajasekaran
- Department of BiotechnologyAnna UniversityBIT‐CampusTiruchirappalliTamil NaduIndia
| |
Collapse
|
88
|
Zhang YG, Singhal M, Lin Z, Manzella C, Kumar A, Alrefai WA, Dudeja PK, Saksena S, Sun J, Gill RK. Infection with enteric pathogens Salmonella typhimurium and Citrobacter rodentium modulate TGF-beta/Smad signaling pathways in the intestine. Gut Microbes 2018; 9:326-337. [PMID: 29381406 PMCID: PMC6219646 DOI: 10.1080/19490976.2018.1429878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Salmonella and Citrobacter are gram negative, members of Enterobacteriaceae family that are important causative agents of diarrhea and intestinal inflammation. TGF-β1 is a pleiotropic multifunctional cytokine that has been implicated in modulating the severity of microbial infections. How these pathogens alter the TGF-β1 signaling pathways in the intestine is largely unknown. Streptomycin-pretreated C57BL/6J mouse model colonized with S. typhimurium for 8 hours (acute) and 4 days (chronic) infection and FVB/N mice infected with C. rodentium for 6 days were utilized. Results demonstrated an increase in TGF-β1 receptor I expression (p<0.05) in S. typhimurium infected mouse ileum at both acute and chronic post-infection vs control. This was associated with activation of Smad pathways as evidenced by increased phosphorylated (p)-Smad2 and p-Smad3 levels in the nucleus. The inhibitory Smad7 mRNA levels showed a significant up regulation during acute phase of Salmonella infection but no change at 4d post-infection. In contrast to Salmonella, infection with Citrobacter caused drastic downregulation of TGF receptor I and II concomitant with a decrease in levels of Smad 2, 3, 4 and 7 expression in the mouse colon. We speculate that increased TGF-β1 signaling in response to Salmonella may be a host compensatory response to promote mucosal healing; while C. rodentium decreases TGF-β1 signaling pathways to promote inflammation and contribute to disease pathogenesis. These findings increase our understanding of how enteric pathogens subvert specific aspects of the host-cellular pathways to cause disease.
Collapse
Affiliation(s)
- Yong-Guo Zhang
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Megha Singhal
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Zhijie Lin
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Christopher Manzella
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Anoop Kumar
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA
| | - Waddah A. Alrefai
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA,Department of Research, Jesse brown VA Medical Center, Chicago, IL, USA
| | - Pradeep K. Dudeja
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA,Department of Research, Jesse brown VA Medical Center, Chicago, IL, USA
| | - Seema Saksena
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA,Department of Research, Jesse brown VA Medical Center, Chicago, IL, USA
| | - Jun Sun
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA,Jun Sun, Ph.D., AGAF, Associate Professor Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB Chicago, IL 60612
| | - Ravinder K. Gill
- Department of Medicine, Division of Gastroenterology & Hepatology, University of Illinois at Chicago, Chicago, IL, USA,CONTACT Ravinder K. Gill, Ph.D., Associate Professor Division of Gastroenterology & Hepatology, Department of Medicine, University of Illinois at Chicago, 820 South Damen Avenue Chicago, IL 60612
| |
Collapse
|
89
|
Bale S, Venkatesh P, Sunkoju M, Godugu C. An Adaptogen: Withaferin A Ameliorates in Vitro and in Vivo Pulmonary Fibrosis by Modulating the Interplay of Fibrotic, Matricelluar Proteins, and Cytokines. Front Pharmacol 2018; 9:248. [PMID: 29623041 PMCID: PMC5874319 DOI: 10.3389/fphar.2018.00248] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022] Open
Abstract
Pulmonary fibrosis (PF) is chronic lung disease with only two FDA approved clinically available drugs, with limited safety profile. Inadequate therapy motivated us to explore the effect of vimentin inhibitor Withaferin A, as an anti-fibrotic agent against TGF-β1-induced in vitro fibrotic events and Bleomycin induced in vivo fibrosis with an emphasis on epithelial to mesenchymal transition (EMT), extracellular matrix deposition (ECM), inflammation, and angiogenesis. In vitro EMT and fibrotic events were induced by TGF-β1 in alveolar epithelial cells and human fetal lung fibroblasts followed by treatment with Withaferin A (0.25, 0.5, and 1 μM concentrations) to explore its anti-fibrotic effects. In vivo potential of Withaferin A (2 and 4 mg/kg) was assessed in murine model of Bleomycin induced PF. All the parameters and molecular studies related to PF were performed at the end of treatment period. Withaferin A treatment reduced the progression of PF by modulating the EMT related cell markers both in vivo and in vitro. Withaferin A ameliorated the expression of inflammatory cytokines including NF-κB p65, IL-1β and TNF-α, as well as attenuated the expression of pro-fibrotic proteins including CTGF, collagen 1A2, collagen 3A1, and fibronectin. Expression of angiogenic factors like VEGF, FAK, p38 MAPK, and PLC-γ1 were also inhibited by Withaferin A. Phosphorylation of Smad 2/3 induced by TGF-β1 and Bleomycin were significantly inhibited. Withaferin A suppressed expression of pro-inflammatory, pro-fibrotic, and pro-angiogenic mediators and also reduced the ECM deposition. In a nutshell, Withaferin A could probably prove as an efficient and potential therapeutic against PF.
Collapse
Affiliation(s)
- Swarna Bale
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Pooladanda Venkatesh
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Manoj Sunkoju
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| |
Collapse
|
90
|
Fibrosis: Lessons from OMICS analyses of the human lung. Matrix Biol 2018; 68-69:422-434. [PMID: 29567123 DOI: 10.1016/j.matbio.2018.03.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 12/30/2022]
Abstract
In recent decades there has been a significant shift in our understanding of idiopathic pulmonary fibrosis (IPF), a progressive and lethal disorder. While initially much of the mechanistic understanding was derived from hypotheses generated from animal models of disease, in recent decades new insights derived from humans with IPF have taken precedence. This is mainly because of the establishment of large collections of IPF lung tissues and patient cohorts, and the emergence of high throughput profiling technologies collectively termed 'omics' technologies based on their shared suffix. In this review we describe impacts of 'omics' analyses of human IPF samples on our understanding of the disease. In particular, we discuss the results of genomics and transcriptomics studies, as well as proteomics, epigenomics and metabolomics. We then describe how these findings can be integrated in a modified paradigm of human idiopathic pulmonary fibrosis, that introduces the 'hallmarks of aging' as a central theme in the IPF lung. This allows resolution of all the disparate cellular and molecular features in IPF, from the central role of epithelial cells, through the dramatic phenotypic alterations observed in fibroblasts and the numerous aberrations that inflammatory cells exhibit. We end with reiterating a call for renewed efforts to collect and analyze carefully characterized human tissues, in ways that would facilitate implementation of novel technologies for high resolution single cell omics profiling.
Collapse
|
91
|
Zhou XM, Wang GL, Wang XB, Liu L, Zhang Q, Yin Y, Wang QY, Kang J, Hou G. GHK Peptide Inhibits Bleomycin-Induced Pulmonary Fibrosis in Mice by Suppressing TGFβ1/Smad-Mediated Epithelial-to-Mesenchymal Transition. Front Pharmacol 2017; 8:904. [PMID: 29311918 PMCID: PMC5733019 DOI: 10.3389/fphar.2017.00904] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022] Open
Abstract
Objective: Idiopathic pulmonary fibrosis is an irreversible and progressive fibrotic lung disease that leads to declines in pulmonary function and, eventually, respiratory failure and has no effective treatment. Gly-His-Lys (GHK) is a tripeptide involved in the processes of tissue regeneration and wound healing and has significant inhibitory effects on transforming growth factor (TGF)-β1 secretion. The effect of GHK on fibrogenesis in pulmonary fibrosis and the exact underlying mechanism have not been studied previously. Thus, this study investigated the effects of GHK on bleomycin (BLM)-induced fibrosis and identified the pathway that is potentially responsible for these effects. Methods: Intratracheal injections of 3 mg/kg BLM were administered to induce pulmonary fibrosis in C57BL/6 mice. GHK was administered intraperitoneally at doses of 2.6, 26, and 260 μg/ml/day every other day from the 4th to the 21st day after BLM instillation. Three weeks after BLM instillation, pulmonary injury and pulmonary fibrosis was evaluated by the hematoxylin-eosin (HE) and Masson’s trichrome (MT) staining. Chronic inflammation index was used for the histological assessments by two pathologists blindly to each other. Tumor necrosis factor (TNF)-α and IL-6 levels in BALF and myeloperoxidase (MPO) activity in lung extracts were measured. For the pulmonary fibrosis evaluation, the fibrosis index calculated based on MT staining, collagen deposition and active TGF-β1 expression detected by ELISA, and the expression of TGF-β1, α-smooth muscle actin (SMA), fibronectin, MMP-9, and TIMP-1 by western blotting. The epithelial mesenchymal transition index, E-cadherin, and vimentin was also detected by western blot. The statistical analysis was performed by one-way ANOVA and the comparison between different groups were performed. Results: Treatment with GHK at all three doses reduced inflammatory cell infiltration and interstitial thickness and attenuated BLM-induced pulmonary fibrosis in mice. GHK treatment significantly improved collagen deposition, and MMP-9/TIMP-1 imbalances in lung tissue and also reduced TNF-α, IL-6 expression in bronchoalveolar lavage fluid (BALF) and MPO in lung extracts. Furthermore, GHK reversed BLM-induced increases in TGF-β1, p-Smad2, p-Smad-3 and insulin-like growth factor-1 (IGF-1) expression. Conclusion: GHK inhibits BLM-induced fibrosis progression, the inflammatory response and EMT via the TGF-β1/Smad 2/3 and IGF-1 pathway. Thus, GHK may be a potential treatment for pulmonary fibrosis.
Collapse
Affiliation(s)
- Xiao-Ming Zhou
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Gui-Liang Wang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xiao-Bo Wang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Li Liu
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Qin Zhang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yan Yin
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Qiu-Yue Wang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Gang Hou
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
92
|
Sanchez CG, Molinski SV, Gongora R, Sosulski M, Fuselier T, MacKinnon SS, Mondal D, Lasky JA. The Antiretroviral Agent Nelfinavir Mesylate. Arthritis Rheumatol 2017; 70:115-126. [DOI: 10.1002/art.40326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/13/2017] [Indexed: 12/28/2022]
Affiliation(s)
| | | | - Rafael Gongora
- Tulane University Health Sciences Center New Orleans Louisiana
| | | | - Taylor Fuselier
- Tulane University Health Sciences Center New Orleans Louisiana
| | | | - Debasis Mondal
- Tulane University School of Medicine New Orleans Louisiana
| | - Joseph A. Lasky
- Tulane University Health Sciences Center New Orleans Louisiana
| |
Collapse
|
93
|
Lawrance IC, Rogler G, Bamias G, Breynaert C, Florholmen J, Pellino G, Reif S, Speca S, Latella G. Cellular and Molecular Mediators of Intestinal Fibrosis. J Crohns Colitis 2017. [PMID: 25306501 DOI: 10.1016/j.crohns.2014.09.00] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Intestinal fibrosis is a major complication of the inflammatory bowel diseases (IBD) and although inflammation is necessary for its development, it would appear that it plays a minor role in its progression as anti-inflammatory treatments in IBD do not prevent fibrosis once it has started. The processes that regulate fibrosis would thus appear to be distinct from those regulating inflammation and, therefore, a detailed understanding of these pathways is vital to the development of anti-fibrogenic strategies. There have been several recent reviews exploring what is known, and what remains unknown, about the development of intestinal fibrosis. This review is designed to add to this literature but with a focus on the cellular components that are involved in the development of fibrogenesis and the major molecular mediators that impact on these cells. The aim is to heighten the understanding of the factors involved in intestinal fibrogenesis so that detailed research can be encouraged in order to advance the processes that could lead to effective treatments.
Collapse
Affiliation(s)
- Ian C Lawrance
- Centre for Inflammatory Bowel Diseases, Fremantle Hospital, Fremantle, WA, Australia.,University Department of Medicine and Pharmacology, University of Western Australia, Fremantle Hospital, Freemantle, WA, Australia
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Ethnikon and Kapodistriakon University of Athens, Laikon Hospital, Athens, Greece
| | - Christine Breynaert
- Department of Immunology and Microbiology, Laboratory of Clinical Immunology, KU Leuven, Leuven, Belgium.,Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jon Florholmen
- Research Group of Gastroenterology and Nutrition, Institute of Clinical Medicine, Artic University of Norway and University Hospital of Northern Norway, Tromsø, Norway
| | - Gianluca Pellino
- General Surgery Unit, Second University of Naples, Naples, Italy
| | - Shimon Reif
- Department of Pediatrics, Tel-Aviv Souraski Medical Center, Tel-Aviv, Israel
| | - Silvia Speca
- National Institute of Health and Medical Research-INSERM, Unit U995, Lille, France
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, Gastroenterology Unit, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
94
|
Lawrance IC, Rogler G, Bamias G, Breynaert C, Florholmen J, Pellino G, Reif S, Speca S, Latella G. Cellular and Molecular Mediators of Intestinal Fibrosis. J Crohns Colitis 2017; 11:1491-1503. [PMID: 25306501 PMCID: PMC5885809 DOI: 10.1016/j.crohns.2014.09.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal fibrosis is a major complication of the inflammatory bowel diseases (IBD) and although inflammation is necessary for its development, it would appear that it plays a minor role in its progression as anti-inflammatory treatments in IBD do not prevent fibrosis once it has started. The processes that regulate fibrosis would thus appear to be distinct from those regulating inflammation and, therefore, a detailed understanding of these pathways is vital to the development of anti-fibrogenic strategies. There have been several recent reviews exploring what is known, and what remains unknown, about the development of intestinal fibrosis. This review is designed to add to this literature but with a focus on the cellular components that are involved in the development of fibrogenesis and the major molecular mediators that impact on these cells. The aim is to heighten the understanding of the factors involved in intestinal fibrogenesis so that detailed research can be encouraged in order to advance the processes that could lead to effective treatments.
Collapse
Affiliation(s)
- Ian C Lawrance
- Centre for Inflammatory Bowel Diseases, Fremantle Hospital, Fremantle, WA, Australia
- University Department of Medicine and Pharmacology, University of Western Australia, Fremantle Hospital, Freemantle, WA, Australia
| | - Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Ethnikon and Kapodistriakon University of Athens, Laikon Hospital, Athens, Greece
| | - Christine Breynaert
- Department of Immunology and Microbiology, Laboratory of Clinical Immunology, KU Leuven, Leuven, Belgium
- Department of General Internal Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Jon Florholmen
- Research Group of Gastroenterology and Nutrition, Institute of Clinical Medicine, Artic University of Norway and University Hospital of Northern Norway, Tromsø, Norway
| | - Gianluca Pellino
- General Surgery Unit, Second University of Naples, Naples, Italy
| | - Shimon Reif
- Department of Pediatrics, Tel-Aviv Souraski Medical Center, Tel-Aviv, Israel
| | - Silvia Speca
- National Institute of Health and Medical Research-INSERM, Unit U995, Lille, France
| | - Giovanni Latella
- Department of Life, Health and Environmental Sciences, Gastroenterology Unit, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
95
|
Jin L, Piao ZH, Sun S, Liu B, Ryu Y, Choi SY, Kim GR, Kim HS, Kee HJ, Jeong MH. Gallic acid attenuates pulmonary fibrosis in a mouse model of transverse aortic contraction-induced heart failure. Vascul Pharmacol 2017; 99:74-82. [PMID: 29097327 DOI: 10.1016/j.vph.2017.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/26/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023]
Abstract
Gallic acid, a trihydroxybenzoic acid found in tea and other plants, attenuates cardiac hypertrophy, fibrosis, and hypertension in animal models. However, the role of gallic acid in heart failure remains unknown. In this study, we show that gallic acid administration prevents heart failure-induced pulmonary fibrosis. Heart failure induced in mice, 8weeks after transverse aortic constriction (TAC) surgery, was confirmed by echocardiography. Treatment for 2weeks with gallic acid but not furosemide prevented cardiac dysfunction in mice. Gallic acid significantly inhibited TAC-induced pathological changes in the lungs, such as increased lung mass, pulmonary fibrosis, and damaged alveolar morphology. It also decreased the expression of fibrosis-related genes, including collagen types I and III, fibronectin, connective tissue growth factor (CTGF), and phosphorylated Smad3. Further, it inhibited the expression of epithelial-mesenchymal transition (EMT)-related genes, such as N-cadherin, vimentin, E-cadherin, SNAI1, and TWIST1. We suggest that gallic acid has therapeutic potential for the treatment of heart failure-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Li Jin
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea; Jilin Hospital Affiliated with Jilin University, 4 Nanjing street, Chuanying, Jilin 132011, China
| | - Zhe Hao Piao
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Simei Sun
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Bin Liu
- The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Yuhee Ryu
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Sin Young Choi
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Gwi Ran Kim
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hae Jin Kee
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea.
| | - Myung Ho Jeong
- Heart Research Center of Chonnam National University Hospital, Gwangju 501-757, Republic of Korea.
| |
Collapse
|
96
|
TGF-β-Induced Endothelial-Mesenchymal Transition in Fibrotic Diseases. Int J Mol Sci 2017; 18:ijms18102157. [PMID: 29039786 PMCID: PMC5666838 DOI: 10.3390/ijms18102157] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/06/2017] [Accepted: 10/13/2017] [Indexed: 12/22/2022] Open
Abstract
Fibrotic diseases are characterized by net accumulation of extracellular matrix proteins in affected organs leading to their dysfunction and ultimate failure. Myofibroblasts have been identified as the cells responsible for the progression of the fibrotic process, and they originate from several sources, including quiescent tissue fibroblasts, circulating CD34⁺ fibrocytes and the phenotypic conversion of various cell types into activated myofibroblasts. Several studies have demonstrated that endothelial cells can transdifferentiate into mesenchymal cells through a process termed endothelial- mesenchymal transition (EndMT) and that this can give rise to activated myofibroblasts involved in the development of fibrotic diseases. Transforming growth factor β (TGF-β) has a central role in fibrogenesis by modulating the fibroblast phenotype and function, inducing myofibroblast transdifferentiation and promoting matrix accumulation. In addition, TGF-β by inducing EndMT may further contribute to the development of fibrosis. Despite extensive investigation of the pathogenesis of fibrotic diseases, no effective treatment strategies are available. Delineation of the mechanisms responsible for initiation and progression of fibrotic diseases is crucial for the development of therapeutic strategies for the treatment of the disease. In this review, we summarize the role of the TGF-β signaling pathway and EndMT in the development of fibrotic diseases and discuss their therapeutic potential.
Collapse
|
97
|
Beigh S, Rashid H, Sharma S, Parvez S, Raisuddin S. Bleomycin-induced pulmonary toxicopathological changes in rats and its prevention by walnut extract. Biomed Pharmacother 2017; 94:418-429. [DOI: 10.1016/j.biopha.2017.07.124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 01/22/2023] Open
|
98
|
The Preventive Role of Gemfibrozil on Bleomycin-Induced Lung Injury and Fibrosis in Rats. Jundishapur J Nat Pharm Prod 2017. [DOI: 10.5812/jjnpp.64314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
99
|
Pattarayan D, Sivanantham A, Krishnaswami V, Loganathan L, Palanichamy R, Natesan S, Muthusamy K, Rajasekaran S. Tannic acid attenuates TGF-β1-induced epithelial-to-mesenchymal transition by effectively intervening TGF-β signaling in lung epithelial cells. J Cell Physiol 2017; 233:2513-2525. [DOI: 10.1002/jcp.26127] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Dhamotharan Pattarayan
- Department of Biotechnology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| | - Ayyanar Sivanantham
- Department of Biotechnology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| | - Venkateshwaran Krishnaswami
- Laboratory for Lipid Based Systems; Department of Pharmaceutical Technology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| | - Lakshmanan Loganathan
- Pharmacogenomics and CADD Lab; Department of Bioinformatics; Alagappa University; Karaikudi Tamil Nadu India
| | - Rajaguru Palanichamy
- Department of Biotechnology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| | - Subramanian Natesan
- Laboratory for Lipid Based Systems; Department of Pharmaceutical Technology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| | - Karthikeyan Muthusamy
- Pharmacogenomics and CADD Lab; Department of Bioinformatics; Alagappa University; Karaikudi Tamil Nadu India
| | - Subbiah Rajasekaran
- Department of Biotechnology; Anna University; BIT-Campus; Tiruchirappalli Tamil Nadu India
| |
Collapse
|
100
|
Yue H, Zhao Y, Wang H, Ma F, Liu F, Shen S, Hou Y, Dou H. Anti-fibrosis effect for Hirsutella sinensis mycelium based on inhibition of mTOR p70S6K phosphorylation. Innate Immun 2017; 23:615-624. [DOI: 10.1177/1753425917726361] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hirsutella sinensis, cultured in vitro, is an attractive substitute for Cordyceps sinensis as health supplement. The aim of this study was to demonstrate whether H. sinensis mycelium (HSM) attenuates murine pulmonary fibrosis induced by bleomycin and to explore the underlying molecular mechanisms. Using lung fibrosis modle induced by intratracheal instillation of bleomycin (BLM; 4 mg/kg), we observed that the administration of HSM reduced HYP, TGF-β1 and the production of several pro-fibrosis cytokines (α-smooth muscle actin, fibronectin and vimentin) in fibrotic mice lung sections. Histopathological examination of lung tissues also demonstrated that HSM improved BLM-induced pathological damage. Concurrently, HSM supplementation markedly reduced the chemotaxis of alveolar macrophages and potently suppressed the expression of inflammatory cytokines. Also, HSM influenced Th1/Th2 and Th17/Treg imbalance and blocked the phosphorylation of mTOR pathway in vivo. Alveolar epithelial A549 cells acquired a mesenchymal phenotype and an increased expression of myofibroblast markers of differentiation (vimentin and fibronectin) after treatment with TGF-β1. HSM suppressed these markers and blocked the phosphorylation of mTOR pathway in vitro. The results provide evidence supporting the use of HSM in the intervention of pulmonary fibrosis and suggest that HSM is a potential therapeutic agent for lung fibrosis.
Collapse
Affiliation(s)
- Huimin Yue
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Yarong Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Haining Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Feiya Ma
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Fei Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|