1
|
Sangamesh VC, Jayaswamy PK, Krishnaraj VM, Kuriakose NK, Hosmane GB, Shetty JK, Patil P, Shetty S, Bhandary R, Shetty P. AnxA2-EGFR pro-inflammatory signaling potentiates EMT-induced fibrotic stress and its modulation by short-chain fatty acid butyrate in idiopathic pulmonary fibrosis. Toxicol Appl Pharmacol 2025; 499:117342. [PMID: 40239744 DOI: 10.1016/j.taap.2025.117342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating lung disease characterized by excessive extracellular matrix deposition, leading to irreversible lung scarring. This study explores the underlying molecular mechanisms of IPF and delves into membrane-anchored synergism between EGFR and AnxA2, which amplifies fibrotic stress and plays a pivotal role in promoting pulmonary fibroblast activation and fibrosis. Indeed, these interactions create a synergistic effect that promotes the loss of epithelial traits and the transition to a mesenchymal phenotype, thereby contributing to fibrotic stress and disease progression. In addition, this study also explores the potential of butyrate, a short-chain fatty acid, as a therapeutic agent in reducing fibrotic stress by modulating AnxA2-EGFR signaling. Pre-treatment with butyrate significantly dampens AnxA2-EGFR signaling and Galectin-3 expression, effectively curbing prolonged EGFR phosphorylation. The suppression of upstream signaling leads to a reduction in the angiogenic marker VEGF and a decrease in pro-inflammatory mediators such as TNF-α and IL-6. Collectively, our findings highlight the critical role of EGFR-AnxA2 signaling and Galectin 3 in the pathogenesis of IPF, and highlight butyrate as a potential therapeutic agent for alleviating fibrotic stress.
Collapse
Affiliation(s)
- Vinay C Sangamesh
- NITTE (Deemed to be University), Nitte University Center for Science Education and Research, Deralakatte, Mangalore 575018, Karnataka, India
| | - Pavan K Jayaswamy
- NITTE (Deemed to be University), Central Research Laboratory, KS. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Vijay M Krishnaraj
- NITTE (Deemed to be University), Central Research Laboratory, KS. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Nithin K Kuriakose
- NITTE (Deemed to be University), Nitte University Center for Science Education and Research, Deralakatte, Mangalore 575018, Karnataka, India
| | - Giridhar B Hosmane
- NITTE (Deemed to be University), Department of Pulmonary Medicine, KS. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Jayaprakash K Shetty
- NITTE (Deemed to be University), Department of Pathology, K. S. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Prakash Patil
- NITTE (Deemed to be University), Central Research Laboratory, KS. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Sukanya Shetty
- NITTE (Deemed to be University), Department of Biochemistry, K.S. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Roopa Bhandary
- NITTE (Deemed to be University), Department of Biochemistry, K.S. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India
| | - Praveenkumar Shetty
- NITTE (Deemed to be University), Central Research Laboratory, KS. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India; NITTE (Deemed to be University), Department of Biochemistry, K.S. Hegde Medical Academy, Deralakatte, Mangalore 575018, Karnataka, India.
| |
Collapse
|
2
|
Naiel S, Dowdall N, Zhou Q, Ali P, Hayat A, Vierhout M, Wong EY, Couto R, Yépez B, Seifried B, Moquin P, Kolb MR, Ask K, Hoare T. Modulating pro-fibrotic macrophages using yeast beta-glucan microparticles prepared by Pressurized Gas eXpanded liquid (PGX) Technology®. Biomaterials 2025; 313:122816. [PMID: 39250864 DOI: 10.1016/j.biomaterials.2024.122816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Pro-fibrotic M2-like macrophages are widely implicated in the pathogenesis and progression of lung fibrosis due to their production of pro-fibrotic growth factors and cytokines. Yeast beta-glucan (YBG) microparticles have shown potential as immunomodulators that can convert macrophage polarization from a pro-fibrotic phenotype to an anti-fibrotic phenotype through the engagement of the Dectin-1 receptor. However, the processing conditions used to fabricate YBG microparticles can lead to unpredictable immunomodulatory effects. Herein, we report the use of Pressurized Gas eXpanded liquids (PGX) Technology® to fabricate YBG (PGX-YBG) microparticles with higher surface areas, lower densities, and smaller and more uniform size distributions compared to commercially available spray-dried YBGs. PGX-YBG is shown to activate Dectin-1 more efficiently in vitro while avoiding significant TLR 2/4 activation. Furthermore, PGX-YBG microparticles effectively modulate M2-like fibrosis-inducing murine and human macrophages into fibrosis-suppressing macrophages both in vitro as well as in ex vivo precision-cut murine lung slices, suggesting their potential utility as a therapeutic for addressing a broad spectrum of fibrotic end-point lung diseases.
Collapse
Affiliation(s)
- S Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - N Dowdall
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Q Zhou
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - P Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - A Hayat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - M Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada
| | - E Y Wong
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - R Couto
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - B Yépez
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - B Seifried
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - P Moquin
- Ceapro Inc., 7824 51 Ave NW, Edmonton, AB, T6E 6W2, Canada
| | - M R Kolb
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - K Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 50 Charlton Avenue East, L314-5, Hamilton, ON, L8N 4A6, Canada.
| | - T Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
3
|
Machahua C, Marti TM, Dorn P, Funke-Chambour M. Fibrosis in PCLS: comparing TGF-β and fibrotic cocktail. Respir Res 2025; 26:44. [PMID: 39875887 PMCID: PMC11776118 DOI: 10.1186/s12931-025-03110-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION Fibrotic cocktail (FC) is a combination of pro-fibrotic and pro-inflammatory mediators that induces early fibrotic changes in organotypic lung models. We hypothesised that transforming growth factor beta 1 (TGF-β1) alone induces a pro-fibrotic effect similar to FC. Our aim was to compare the pro-fibrotic effects of TGF-β1 with FC in human precision-cut lung slices (PCLS). METHODS PCLS from "healthy" lung tissue of cancer patients undergoing surgery (n = 7) were incubated with TGF-β1, FC or control for 72 h. Gene expression markers for myofibroblasts differentiation, extracellular matrix (ECM), as well as TGF-β receptors were assessed (RT-qPCR). ECM proteins expression in lysates and supernatant was assessed by ELISA and immunofluorescence. RESULTS We found that TGF-β1 significantly increased gene expression of ACTA2, COL1A1, CCN2, and VIM compared to control but also compared to FC. FC showed a significant increase of matrix metalloproteinase (MMP) 7 and 1 compared to control, while TGF-β receptor 2 was lower after FC compared to TGF-β1 or control. FC or TGF-β1 showed similar fibronectin protein expression in lysates and supernatants, while type I collagen protein expression in lysates was significantly greater with TGF-β1 compared to control. CONCLUSIONS Our findings show that TGF-β1 induces consistent pro-fibrotic changes in PCLS after 72 h. Compared to TGF-β1, FC treatment resulted in reduced gene expression of TGF-β receptor 2 and increased MMPs expression, potentially mitigating the early pro-fibrotic effects. Selecting specific pro-fibrotic stimuli may be preferable depending on the research question and time point of interest in lung fibrosis studies using PCLS.
Collapse
Affiliation(s)
- Carlos Machahua
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBRM), University of Bern, Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBRM), University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Woo S, Gandhi S, Ghincea A, Saber T, Lee CJ, Ryu C. Targeting the NLRP3 inflammasome and associated cytokines in scleroderma associated interstitial lung disease. Front Cell Dev Biol 2023; 11:1254904. [PMID: 37849737 PMCID: PMC10577231 DOI: 10.3389/fcell.2023.1254904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023] Open
Abstract
SSc-ILD (scleroderma associated interstitial lung disease) is a complex rheumatic disease characterized in part by immune dysregulation leading to the progressive fibrotic replacement of normal lung architecture. Because improved treatment options are sorely needed, additional study of the fibroproliferative mechanisms mediating this disease has the potential to accelerate development of novel therapies. The contribution of innate immunity is an emerging area of investigation in SSc-ILD as recent work has demonstrated the mechanistic and clinical significance of the NLRP3 inflammasome and its associated cytokines of TNFα (tumor necrosis factor alpha), IL-1β (interleukin-1 beta), and IL-18 in this disease. In this review, we will highlight novel pathophysiologic insights afforded by these studies and the potential of leveraging this complex biology for clinical benefit.
Collapse
Affiliation(s)
| | | | | | | | | | - Changwan Ryu
- Department of Internal Medicine, Yale School of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, New Haven, CT, United States
| |
Collapse
|
5
|
Gredic M, Karnati S, Ruppert C, Guenther A, Avdeev SN, Kosanovic D. Combined Pulmonary Fibrosis and Emphysema: When Scylla and Charybdis Ally. Cells 2023; 12:1278. [PMID: 37174678 PMCID: PMC10177208 DOI: 10.3390/cells12091278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Combined pulmonary fibrosis and emphysema (CPFE) is a recently recognized syndrome that, as its name indicates, involves the existence of both interstitial lung fibrosis and emphysema in one individual, and is often accompanied by pulmonary hypertension. This debilitating, progressive condition is most often encountered in males with an extensive smoking history, and is presented by dyspnea, preserved lung volumes, and contrastingly impaired gas exchange capacity. The diagnosis of the disease is based on computed tomography imaging, demonstrating the coexistence of emphysema and interstitial fibrosis in the lungs, which might be of various types and extents, in different areas of the lung and several relative positions to each other. CPFE bears high mortality and to date, specific and efficient treatment options do not exist. In this review, we will summarize current knowledge about the clinical attributes and manifestations of CPFE. Moreover, we will focus on pathophysiological and pathohistological lung phenomena and suspected etiological factors of this disease. Finally, since there is a paucity of preclinical research performed for this particular lung pathology, we will review existing animal studies and provide suggestions for the development of additional in vivo models of CPFE syndrome.
Collapse
Affiliation(s)
- Marija Gredic
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, 35392 Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany
| | - Clemens Ruppert
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, 35392 Giessen, Germany
- UGMLC Giessen Biobank & European IPF Registry/Biobank, 35392 Giessen, Germany
| | - Andreas Guenther
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus Liebig University, 35392 Giessen, Germany
- UGMLC Giessen Biobank & European IPF Registry/Biobank, 35392 Giessen, Germany
- Institute for Lung Health (ILH), 35392 Giessen, Germany
- Lung Clinic, Evangelisches Krankenhaus Mittelhessen, 35398 Giessen, Germany
| | - Sergey N. Avdeev
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Djuro Kosanovic
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
6
|
Novak CM, Sethuraman S, Luikart KL, Reader BF, Wheat JS, Whitson B, Ghadiali SN, Ballinger MN. Alveolar macrophages drive lung fibroblast function in cocultures of IPF and normal patient samples. Am J Physiol Lung Cell Mol Physiol 2023; 324:L507-L520. [PMID: 36791050 PMCID: PMC10259863 DOI: 10.1152/ajplung.00263.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/19/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by increased collagen accumulation that is progressive and nonresolving. Although fibrosis progression may be regulated by fibroblasts and alveolar macrophage (AM) interactions, this cellular interplay has not been fully elucidated. To study AM-fibroblast interactions, cells were isolated from IPF and normal human lung tissue and cultured independently or together in direct 2-D coculture, direct 3-D coculture, indirect transwell, and in 3-D hydrogels. AM influence on fibroblast function was assessed by gene expression, cytokine/chemokine secretion, and hydrogel contractility. Normal AMs cultured in direct contact with fibroblasts downregulated extracellular matrix (ECM) gene expression whereas IPF AMs had little to no effect. Fibroblast contractility was assessed by encapsulating cocultures in 3-D collagen hydrogels and monitoring gel diameter over time. Both normal and IPF AMs reduced baseline contractility of normal fibroblasts but had little to no effect on IPF fibroblasts. When stimulated with Toll-like receptor (TLR) agonists, IPF AMs increased production of pro-inflammatory cytokines TNFα and IL-1β, compared with normal AMs. TLR ligand stimulation did not alter fibroblast contraction, but stimulation with exogenous TNFα and TGFβ did alter contraction. To determine if the observed changes required cell-to-cell contact, AM-conditioned media and transwell systems were utilized. Transwell culture showed decreased ECM gene expression changes compared with direct coculture and conditioned media from AMs did not alter fibroblast contraction regardless of disease state. Taken together, these data indicate that normal fibroblasts are more responsive to AM crosstalk, and that AM influence on fibroblast behavior depends on cell proximity.
Collapse
Affiliation(s)
- Caymen M Novak
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Shruthi Sethuraman
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States
| | - Kristina L Luikart
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States
| | - Brenda F Reader
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States
| | - Jana S Wheat
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| | - Bryan Whitson
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States
| | - Samir N Ghadiali
- Department of Biomedical Engineering, Ohio State University, Columbus, Ohio, United States
| | - Megan N Ballinger
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, Ohio, United States
| |
Collapse
|
7
|
TNFR1 Mediated Apoptosis Is Protective against Mycobacterium avium in Mice. Microorganisms 2023; 11:microorganisms11030778. [PMID: 36985352 PMCID: PMC10051498 DOI: 10.3390/microorganisms11030778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Mycobacterium avium is an intracellular proliferating pathogen that causes chronic refractory respiratory infection. Although apoptosis induced by M. avium has been reported in vitro, the role of apoptosis against M. avium infection in vivo remains unclear. Here, we investigated the role of apoptosis in mouse models of M. avium infection. Tumor necrosis factor receptor-1 knockout mice (TNFR1-KO) andTNFR2-KO micewere used. M. avium (1 × 107 cfu/body) was administered intratracheally to mice. Apoptosis in lungs was detected by terminal deoxynucleotidyl transferase mediated dUTP nick end labeling and lung histology as well as cell death detection kits using BAL fluids. TNFR1-KO mice were susceptible to M. avium infection compared with TNFR2-KO and wild type mice based on the bacterial number and lung histology. Higher numbers of apoptotic cells were detected in the lungs of TNFR2-KO and wild-type mice were compared with TNFR1-KO mice. The inhalation of Z-VAD-FMK deteriorated M. avium infection compared with vehicle-inhaled controls. Overexpression of Iκ-B alpha by adenovirus vector attenuated M. avium infection. Our study showed apoptosis had an important role in innate immunity against M. avium in mice. The induction of apoptosis in M. avium-infected cells might be a new strategy to control M. avium infection.
Collapse
|
8
|
Afarin R, Aslani F, Asadizade S, Jaberian Asl B, Mohammadi Gahrooie M, Shakerian E, Ahangarpour A. The Effect of Lipopolysaccharide-Stimulated Adipose-Derived Mesenchymal Stem Cells on NAFLD Treatment in High-Fat Diet-Fed Rats. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2023; 22:e134807. [PMID: 38116551 PMCID: PMC10728850 DOI: 10.5812/ijpr-134807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 12/21/2023]
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are 2 common liver diseases that currently lack effective treatment options. Objectives This study aimed to investigate the effect of lipopolysaccharide (LPS)-stimulated adipose-derived stem cells (ADSCs) on NAFLD treatment in an animal model. Methods Male Wistar rats were fed a high-fat diet (HFD) to induce NAFLD for 7 weeks. The rats were then categorized into 3 groups: Mesenchymal stem cell (MSC), MSC + LPS, and fenofibrate (FENO) groups. Liver and body weight were measured, and the expression of genes involved in fatty acid biosynthesis, β-oxidation, and inflammatory responses was assessed. Results Lipopolysaccharide-stimulated ADSCs were more effective in regulating liver and body weight gain and reducing liver triglyceride (TG) levels compared to the other groups. Treatment with LPS-stimulated ADSCs effectively corrected liver enzymes, including alanine aminotransferase (ALT) and aspartate aminotransferase (AST), and lipid factors, including low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C) values, better than treatment with both FENO and MSCs. ADSCs + LPS treatment significantly decreased transforming growth factor β (TGF-β) and genes associated with inflammatory responses. Additionally, there was a significant reduction in reactive oxygen species (ROS) levels in the rats treated with ADSCs + LPS. Conclusions Lipopolysaccharide-stimulated ADSCs showed potential in alleviating NAFLD by reducing inflammatory genes and ROS levels in HFD rats, demonstrating better results than treatment with ADSCs and FENO groups alone.
Collapse
Affiliation(s)
- Reza Afarin
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fereshteh Aslani
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahla Asadizade
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahar Jaberian Asl
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mehrnoosh Mohammadi Gahrooie
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elham Shakerian
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Diabetes Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
El-Bestawy EM, Tolba AM, Rashad WA. Morphological, ultrastructural, and biochemical changes induced by sodium fluoride in the tongue of adult male albino rat and the ameliorative effect of resveratrol. Anat Cell Biol 2022; 55:483-496. [PMID: 36168808 PMCID: PMC9747341 DOI: 10.5115/acb.22.088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/05/2022] [Accepted: 06/21/2022] [Indexed: 01/02/2023] Open
Abstract
Little knowledge is available about the effects of fluoride exposure on the tongue. This study evaluated the effects of sodium fluoride (NaF) on the tongue ultrastructure and detected the ameliorative effects of resveratrol. Forty adult albino rats were separated into 4 groups: the control group was given a balanced diet and purified water. The NaF treated group: received 10 mg/kg/d dissolved in 2.5 ml distilled water once daily for 30 days orally. The NaF+resveratrol group: received NaF 10 mg/kg/d orally together with resveratrol in a dose of 30 mg/kg daily for 30 days. The resveratrol group was subjected to resveratrol in a dose of 30 mg/kg/d by oral gavage for 30 days. Sections were stained with hematoxylin & eosin, and Masson's trichrome. Tumor necrosis factor α immunohistochemical study and electron microscopic examinations were done. The oxidative stress markers malondialdehyde, antioxidant reduced glutathione, and the total antioxidant capacity were measured. The NaF group revealed ulceration, necrotic muscle fibers, distorted papillae and a significant increase in malondialdehyde level, and a significant decrease in glutathione and the total antioxidant levels. In the NaF+resveratrol group, pathological changes were less, and the oxidant levels were decreased by the administration of resveratrol with NaF. In conclusion, NaF adversely affects the ultrastructure of the adult rat tongue and resveratrol can ameliorate this effect.
Collapse
Affiliation(s)
- Emtethal M. El-Bestawy
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Sharqia Governorate, Egypt
| | - Asmaa M. Tolba
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Sharqia Governorate, Egypt
| | - Walaa A. Rashad
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Sharqia Governorate, Egypt
| |
Collapse
|
10
|
Iwanishi H, Yamanaka O, Sumioka T, Yasuda S, Miyajima M, Saika S. Delayed regression of laser-induced choroidal neovascularization in TNFα-null mice. J Cell Mol Med 2022; 26:5315-5325. [PMID: 36127870 PMCID: PMC9575074 DOI: 10.1111/jcmm.17562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/30/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022] Open
Abstract
We investigated the effects of lacking TNFα on the development and regression of Argon-laser-induced choroidal neovascularization (CNV) in mice. We lasered ocular fundus for induction of CNV in both wild-type (WT) and TNFα-null (KO) mice. Fluorescence angiography was performed to examine the size of CNV lesions. Gene expression pattern of wound healing-related components was examined. The effects of exogenous TNFα on apoptosis of human retinal microvascular endothelial cells (HRMECs) and on the tube-like structure of the cells were investigated in vitro. The results showed that Argon-laser irradiation-induced CNV was significantly larger in KO mice than WT mice on Day 21, but not at other timepoints. Lacking TNFα increased neutrophil population in the lesion. The distribution of cleaved caspase3-labelled apoptotic cells was more frequently observed in the laser-irradiated tissue in a WT mouse as compared with a KO mouse. Exogenous TNFα induced apoptosis of HRMECs and accelerated regression of tube-like structure of HRMECs in cell culture. Taken together, TNFα gene knockout delays the regression of laser-induced CNV in mice. The mechanism underlying the phenotype might include the augmentation of neutrophil population in the treated tissue and attenuation of vascular endothelial cell apoptosis.
Collapse
Affiliation(s)
- Hiroki Iwanishi
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Shingo Yasuda
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Masayasu Miyajima
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
11
|
Ghosh AJ, Hobbs BD, Yun JH, Saferali A, Moll M, Xu Z, Chase RP, Morrow J, Ziniti J, Sciurba F, Barwick L, Limper AH, Flaherty K, Criner G, Brown KK, Wise R, Martinez FJ, McGoldrick D, Cho MH, DeMeo DL, Silverman EK, Castaldi PJ, Hersh CP. Lung tissue shows divergent gene expression between chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Respir Res 2022; 23:97. [PMID: 35449067 PMCID: PMC9026726 DOI: 10.1186/s12931-022-02013-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) are characterized by shared exposures and clinical features, but distinct genetic and pathologic features exist. These features have not been well-studied using large-scale gene expression datasets. We hypothesized that there are divergent gene, pathway, and cellular signatures between COPD and IPF. METHODS We performed RNA-sequencing on lung tissues from individuals with IPF (n = 231) and COPD (n = 377) compared to control (n = 267), defined as individuals with normal spirometry. We grouped the overlapping differential expression gene sets based on direction of expression and examined the resultant sets for genes of interest, pathway enrichment, and cell composition. Using gene set variation analysis, we validated the overlap group gene sets in independent COPD and IPF data sets. RESULTS We found 5010 genes differentially expressed between COPD and control, and 11,454 genes differentially expressed between IPF and control (1% false discovery rate). 3846 genes overlapped between IPF and COPD. Several pathways were enriched for genes upregulated in COPD and downregulated in IPF; however, no pathways were enriched for genes downregulated in COPD and upregulated in IPF. There were many myeloid cell genes with increased expression in COPD but decreased in IPF. We found that the genes upregulated in COPD but downregulated in IPF were associated with lower lung function in the independent validation cohorts. CONCLUSIONS We identified a divergent gene expression signature between COPD and IPF, with increased expression in COPD and decreased in IPF. This signature is associated with worse lung function in both COPD and IPF.
Collapse
Affiliation(s)
- Auyon J. Ghosh
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Brian D. Hobbs
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Jeong H. Yun
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Aabida Saferali
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Matthew Moll
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Zhonghui Xu
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Robert P. Chase
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Jarrett Morrow
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - John Ziniti
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA
| | - Frank Sciurba
- grid.21925.3d0000 0004 1936 9000Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Lucas Barwick
- grid.280434.90000 0004 0459 5494The Emmes Company, Rockville, MD USA
| | - Andrew H. Limper
- grid.66875.3a0000 0004 0459 167XDivision of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Kevin Flaherty
- grid.214458.e0000000086837370Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Healthy System, Ann Arbor, MI USA
| | - Gerard Criner
- grid.264727.20000 0001 2248 3398Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA USA
| | - Kevin K. Brown
- grid.240341.00000 0004 0396 0728Department of Medicine, National Jewish Health, Denver, CO USA
| | - Robert Wise
- grid.21107.350000 0001 2171 9311Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD USA
| | - Fernando J. Martinez
- grid.5386.8000000041936877XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Daniel McGoldrick
- grid.34477.330000000122986657Northwest Genomics Center, University of Washington, Seattle, WA USA
| | - Michael H. Cho
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Dawn L. DeMeo
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Edwin K. Silverman
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | - Peter J. Castaldi
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| | | | - Craig P. Hersh
- grid.62560.370000 0004 0378 8294Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115 USA ,grid.62560.370000 0004 0378 8294Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA USA ,grid.38142.3c000000041936754XHarvard Medical School, Boston, MA USA
| |
Collapse
|
12
|
Guo X, Sunil C, Qian G. Obesity and the Development of Lung Fibrosis. Front Pharmacol 2022; 12:812166. [PMID: 35082682 PMCID: PMC8784552 DOI: 10.3389/fphar.2021.812166] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Obesity is an epidemic worldwide and the obese people suffer from a range of respiratory complications including fibrotic changes in the lung. The influence of obesity on the lung is multi-factorial, which is related to both mechanical injury and various inflammatory mediators produced by excessive adipose tissues, and infiltrated immune cells. Adiposity causes increased production of inflammatory mediators, for example, cytokines, chemokines, and adipokines, both locally and in the systemic circulation, thereby rendering susceptibility to respiratory diseases, and altered responses. Lung fibrosis is closely related to chronic inflammation in the lung. Current data suggest a link between lung fibrosis and diet-induced obesity, although the mechanism remains incomplete understood. This review summarizes findings on the association of lung fibrosis with obesity, highlights the role of several critical inflammatory mediators (e.g., TNF-α, TGF-β, and MCP-1) in obesity related lung fibrosis and the implication of obesity in the outcomes of idiopathic pulmonary fibrosis patients.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| | - Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, The University of Texas at Tyler, Tyler, TX, United States
| |
Collapse
|
13
|
The Role of Cell Proliferation and Extracellular Matrix Accumulation Induced by Food Additive Butylated Hydroxytoluene in Uterine Leiomyoma. Nutrients 2021; 13:nu13093074. [PMID: 34578952 PMCID: PMC8468901 DOI: 10.3390/nu13093074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 11/17/2022] Open
Abstract
Leiomyoma is the most common benign uterine tumor in reproductive-age women. Increasing numbers of studies are focusing on the effects of environmental exposure on the incidence and progression of tumors. One major step taken in the food industry is the addition of food preservatives to maintain freshness. Butylated hydroxytoluene (BHT) is a synthetic phenolic antioxidant, which is widely used as an additive to develop fat-soluble characteristics, as well as in cosmetics and rubber. Previous studies also highlighted that BHT may be related to increased fibrosis capacity and carcinogenic effects. In this study, we explored the effects of the commonly used food additive BHT on leiomyoma progression, and the related mechanism. The exposure of the ELT-3 leiomyoma cell line to BHT for 48 h increased the proliferative effect. Since leiomyoma progression is related to increases in extracellular matrix (ECM) accumulation and matrix metalloproteinase (MMP), BHT could effectively increase ECM-related protein expression, as well as MMP-2 and MMP-9 protein expression. This increase in ECM, in response to BHT, may be linked to the activation of the phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) signaling pathway. Through PI3K inhibition, BHT’s effect on leiomyoma progression could be partially modulated. These results suggest the harmful effect of BHT exposure on leiomyoma progression may relate to PI3K modulation. However, an in vivo study is necessary to confirm these findings.
Collapse
|
14
|
Gonzalez Rodriguez A, Schroeder ME, Grim JC, Walker CJ, Speckl KF, Weiss RM, Anseth KS. Tumor necrosis factor-α promotes and exacerbates calcification in heart valve myofibroblast populations. FASEB J 2021; 35:e21382. [PMID: 33554387 DOI: 10.1096/fj.202002013rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022]
Abstract
Pro-inflammatory cytokines play critical roles in regulating valvular interstitial cell (VIC) phenotypic changes that can cause heart valve fibrosis and calcification. Tumor necrosis factor alpha (TNF-α) is a cytokine known to influence VIC behavior and has been reported at high levels in calcified valves ex vivo. We sought to understand the specific effects of TNF-α on VIC phenotypes (eg, fibroblast, profibrotic activated myofibroblasts) and its link with heart valve disorders. We characterize human aortic valve tissue from patients with valve disorders and identify a high variability of fibrotic and calcific markers between tissues. These results motivated in vitro studies to explore the effects of TNF-α on defined VIC fibroblasts and profibrotic activated myofibroblasts, induced via FGF-2 and TGF-β1 treatment. Using 3D hydrogels to culture VICs, we measure the effect of TNF-α (0.1-10 ng/mL) on key markers of fibrosis (eg, αSMA, COL1A1) and calcification (eg, RUNX2, BMP2, and calcium deposits). We observe calcification in TNF-α-treated VIC activated myofibroblasts and identify the MAPK/ERK signaling cascade as a potential pathway for TNF-α mediated calcification. Conversely, VIC fibroblasts respond to TNF-α with decreased calcification. Treatment of VIC profibrotic activated myofibroblast populations with TNF-α leads to increased calcification. Our in vitro findings correlate with findings in diseased human valves and highlight the importance of understanding the effect of cytokines and signaling pathways on specific VIC phenotypes. Finally, we reveal MAPK/ERK as a potential pathway involved in VIC-mediated matrix calcification with TNF-α treatment, suggesting this pathway as a potential pharmaceutical target for aortic valve disease.
Collapse
Affiliation(s)
- Andrea Gonzalez Rodriguez
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Megan E Schroeder
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Joseph C Grim
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Cierra J Walker
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| | - Kelly F Speckl
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Robert M Weiss
- Department of Internal Medicine, University of Iowa, Iowa City, IA, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA.,BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA.,Materials Science and Engineering Program, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
15
|
Hult EM, Gurczynski SJ, Moore BB. M2 macrophages have unique transcriptomes but conditioned media does not promote profibrotic responses in lung fibroblasts or alveolar epithelial cells in vitro. Am J Physiol Lung Cell Mol Physiol 2021; 321:L518-L532. [PMID: 34231378 DOI: 10.1152/ajplung.00107.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Macrophages are critical regulators of pulmonary fibrosis. Their plasticity, proximity, and ability to cross talk with structural cells of the lung make them a key cell type of interest in the regulation of lung fibrosis. Macrophages can express a variety of phenotypes, which have been historically represented through an "M1-like" to "M2-like" delineation. In this classification, M1-like macrophages are proinflammatory and have increased phagocytic capacity compared with alternatively activated M2-like macrophages that are profibrotic and are associated with wound healing. Extensive evidence in the field in both patients and animal models aligns pulmonary fibrosis with M2 macrophages. In this study, we performed RNA sequencing (RNAseq) to fully characterize M1- vs. M2-skewed bone marrow-derived macrophages (BMDMs) and investigated the profibrotic abilities of M2 BMDM conditioned media (CM) to promote fibroblast migration and proliferation, alveolar epithelial cell (AEC) apoptosis, and mRNA expression of key fibrotic genes in both fibroblasts and AECs. Although M2 CM-treated fibroblasts had increased migration and M2 CM-treated fibroblasts and AECs had increased expression of profibrotic proteins over M1 CM-treated cells, all differences can be attributed to M2 polarization reagents IL-4 and IL-13 also present in the CM. Collectively, these data suggest that the profibrotic effects associated with M2 macrophage CM in vitro are attributable to effects of polarization cytokines rather than additional factors secreted in response to those polarizing cytokines.
Collapse
Affiliation(s)
- Elissa M Hult
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Stephen J Gurczynski
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Bethany B Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
16
|
High Doses of Kefir Accelerate Lung-Injury Progression in Bleomycin-Induced Pneumonitis in Rats. Jundishapur J Nat Pharm Prod 2021. [DOI: 10.5812/jjnpp.111882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Bleomycin-induced pneumonitis (BIP) is a common consequence of bleomycin (BLE) use during chemotherapy. Kefir is a probiotic with many health benefits. Many cancer patients in Indonesia consume kefir as a complementary traditional medicine alongside standard chemotherapy. Objectives: This study aimed to investigate the effects of high-dose kefir consumption on BIP in a rat model. Methods: Wistar male rats were given 0.3 mg of BLE via intranasal inhalation for 6 days with a daily intragastric administration of either phosphate buffered saline (PBS) or kefir at dosages of 2.5 mL, 3.5 mL, and 4.5 mL per day for 30 days. On day 30, lung sections were obtained and stained with hematoxylin and eosin for histological examinations. Immunohistochemistry tests were carried out to determine the activity levels of matrix metalloproteinase (MMP)-1, signal transducer, and activator of transcription (STAT)-3. TNF-α and IL-6 concentrations in plasma were also evaluated. Results: Histological results showed damage to the lung structure by inflammation with diffuse infiltrate, with some areas exhibiting slight fibrosis. The number of alveolar epithelial cells expressing MMP-1 significantly increased with the kefir dosage. Interestingly, only the highest dose of kefir raised IL-6 levels, while TNF-α levels increased at all kefir doses. STAT-3 showed a slight increase in activity level. As MMP-1 works to degrade fibrosis while both TNF-α and Il-6 are correlated with inflammation, these findings might explain the observed histological changes in lung structure in the BLE and kefir groups. Conclusions: The administration of high doses of kefir in rats increased the expression of pro-inflammatory cytokines, which worsened BIP.
Collapse
|
17
|
Qu J, Yang SZ, Zhu Y, Guo T, Thannickal VJ, Zhou Y. Targeting mechanosensitive MDM4 promotes lung fibrosis resolution in aged mice. J Exp Med 2021; 218:e20202033. [PMID: 33688918 PMCID: PMC7953267 DOI: 10.1084/jem.20202033] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/18/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Aging is a strong risk factor and an independent prognostic factor for progressive human idiopathic pulmonary fibrosis (IPF). Aged mice develop nonresolving pulmonary fibrosis following lung injury. In this study, we found that mouse double minute 4 homolog (MDM4) is highly expressed in the fibrotic lesions of human IPF and experimental pulmonary fibrosis in aged mice. We identified MDM4 as a matrix stiffness-regulated endogenous inhibitor of p53. Reducing matrix stiffness down-regulates MDM4 expression, resulting in p53 activation in primary lung myofibroblasts isolated from IPF patients. Gain of p53 function activates a gene program that sensitizes lung myofibroblasts to apoptosis and promotes the clearance of apoptotic myofibroblasts by macrophages. Destiffening of the fibrotic lung matrix by targeting nonenzymatic cross-linking or genetic ablation of Mdm4 in lung (myo)fibroblasts activates the Mdm4-p53 pathway and promotes lung fibrosis resolution in aged mice. These findings suggest that mechanosensitive MDM4 is a molecular target with promising therapeutic potential against persistent lung fibrosis associated with aging.
Collapse
Affiliation(s)
- Jing Qu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shan-Zhong Yang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Yi Zhu
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Ting Guo
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
- The Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Victor J. Thannickal
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Yong Zhou
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
18
|
Interstitial Score and Concentrations of IL-4Rα, PAR-2, and MMP-7 in Bronchoalveolar Lavage Fluid Could Be Useful Markers for Distinguishing Idiopathic Interstitial Pneumonias. Diagnostics (Basel) 2021; 11:diagnostics11040693. [PMID: 33924683 PMCID: PMC8070528 DOI: 10.3390/diagnostics11040693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/04/2021] [Accepted: 04/12/2021] [Indexed: 01/28/2023] Open
Abstract
Idiopathic interstitial pneumonia (IIP) entails a variable group of lung diseases of unknown etiology. Idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, interstitial lung diseases related to connective tissue disease (CTD-ILD), and hypersensitivity pneumonitis (HP) can manifest with similar clinical, radiological, and histopathological features. In a differential diagnosis, biomarkers can play a significant role. We assume that levels of specific cyto- or chemokines or their receptors can signal pathogenetic processes in the lungs. Eighty patients with different types of idiopathic interstitial pneumonia were enrolled in this study. Cell counts and concentrations of tumor necrosis factor (TNF)-α, interleukin-4 receptor α, proteinase-activated receptor (PAR)-2, matrix metalloproteinase (MMP)-7, and B cell-activating factor were measured in bronchoalveolar lavage fluid using commercial ELISA kits. High resolution computer tomography results were evaluated using alveolar and interstitial (IS) score scales. Levels of TNF-α were significantly higher in HP compared to fibrosing IIP (p < 0.0001) and CTD-ILD (p = 0.0381). Concentrations of IL-4Rα, PAR-2, and MMP-7 were positively correlated with IS (p = 0.0009; p = 0.0256; p = 0.0015, respectively). Since TNF-α plays a major role in inflammation, our results suggest that HP is predominantly an inflammatory disease. From the positive correlation with IS we believe that IL-4Rα, PAR-2, and MMP-7 could serve as fibroproliferative biomarkers in differential diagnosis of IIP.
Collapse
|
19
|
Sharma MR, Mitrani R, Werth VP. Effect of TNFα blockade on UVB-induced inflammatory cell migration and collagen loss in mice. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 213:112072. [PMID: 33181431 DOI: 10.1016/j.jphotobiol.2020.112072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 09/25/2020] [Accepted: 10/26/2020] [Indexed: 11/27/2022]
Abstract
UVB irradiation induces pro-inflammatory cytokines including interleukin-1 (IL-1) and tumor necrosis factor-α (TNFα) in the skin. TNFα stimulates the chemotaxis of inflammatory cells to the skin. These cells secrete metalloproteinases (MMPs) and other enzymes that damage the cutaneous matrix. Therefore, blocking TNFα activity could be effective in preventing the influx of inflammatory cells and subsequent collagen degradation in the skin. In addition, TNFα downregulates procollagen mRNA, and thus blockade may be beneficial to production of type I collagen. Female C57BL/6 J mice were treated with etanercept (TNFα blocker, 4 mg/kg/day) for 4 days 1 h prior to UVB irradiation (100 mJ/cm2/day for 5 days). On the 5th day mice were sacrificed 3 h after UVB exposure. Blocking TNFα significantly inhibited UVB-induced recruitment of macrophages, mast cells, and neutrophils. UVB-irradiated mice skin contained more mature collagen compared to etanercept and UVB + etanercept-treated mice. Skin from UVB + etanercept-treated mice had more collagen fragments relative to UVB-irradiated mice. Procollagen protein was lower in UVB-irradiated and UVB + etanercept-treated mice. TNFα blockade decreased decorin and TGF-β1 in UVB-irradiated mice compared to UVB alone. MMP13 was inhibited by etanercept in UVB-irradiated mice (p < 0.01). In conclusion, blockade of TNFα significantly decreased mature collagen in UVB-irradiated mice, while increasing collagen fragmentation and decreasing procollagen.
Collapse
Affiliation(s)
- Meena R Sharma
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States of America; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Robert Mitrani
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States of America; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Victoria P Werth
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States of America; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States of America.
| |
Collapse
|
20
|
Wang C, Zhou J, Wang J, Li S, Fukunaga A, Yodoi J, Tian H. Progress in the mechanism and targeted drug therapy for COPD. Signal Transduct Target Ther 2020; 5:248. [PMID: 33110061 PMCID: PMC7588592 DOI: 10.1038/s41392-020-00345-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is emphysema and/or chronic bronchitis characterised by long-term breathing problems and poor airflow. The prevalence of COPD has increased over the last decade and the drugs most commonly used to treat it, such as glucocorticoids and bronchodilators, have significant therapeutic effects; however, they also cause side effects, including infection and immunosuppression. Here we reviewed the pathogenesis and progression of COPD and elaborated on the effects and mechanisms of newly developed molecular targeted COPD therapeutic drugs. Among these new drugs, we focussed on thioredoxin (Trx). Trx effectively prevents the progression of COPD by regulating redox status and protease/anti-protease balance, blocking the NF-κB and MAPK signalling pathways, suppressing the activation and migration of inflammatory cells and the production of cytokines, inhibiting the synthesis and the activation of adhesion factors and growth factors, and controlling the cAMP-PKA and PI3K/Akt signalling pathways. The mechanism by which Trx affects COPD is different from glucocorticoid-based mechanisms which regulate the inflammatory reaction in association with suppressing immune responses. In addition, Trx also improves the insensitivity of COPD to steroids by inhibiting the production and internalisation of macrophage migration inhibitory factor (MIF). Taken together, these findings suggest that Trx may be the ideal drug for treating COPD.
Collapse
Affiliation(s)
- Cuixue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jiedong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Jinquan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Shujing Li
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China
| | - Atsushi Fukunaga
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, 312000, China.
- Jiaozhimei Biotechnology (Shaoxing) Co, Ltd, Shaoxing, 312000, China.
| |
Collapse
|
21
|
Sun Z, Yang Z, Wang M, Huang C, Ren Y, Zhang W, Gao F, Cao L, Li L, Nie S. Paraquat induces pulmonary fibrosis through Wnt/β-catenin signaling pathway and myofibroblast differentiation. Toxicol Lett 2020; 333:170-183. [PMID: 32795487 DOI: 10.1016/j.toxlet.2020.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022]
Abstract
Paraquat (PQ) poisoning-induced pulmonary fibrosis always results in fatal harm to patients. Our study aimed to investigate the functions of the Wnt/β-catenin pathway in PQ-induced pulmonary fibrosis. By comparing the proteomic profiles of rat lung tissues using protein array in the absence or presence of PQ, the Wnt/β-catenin signaling, as a fibrosis-related pathway, was discovered to be profoundly activated by PQ. The protein levels of Wnt/β-catenin signaling components including MMP-2, β-catenin, Wnt3a, Wnt10b, Cyclin D1, and WISP1 were increased in PQ-treated rat lung tissues. Surprisingly, PQ was found to be able to promote lung epithelial cells and fibroblasts differentiating into myofibroblasts by activating Wnt/β-catenin signaling pathway. Dickkopf-1 (DKK1), an antagonist of Wnt/β-catenin signaling pathway, could inhibit the myofibroblast differentiation and attenuate PQ-induced pulmonary fibrogenesis in vitro and in vivo. The expression levels of fibroblasts markers Vimentin, α-smooth muscle actin (α-SMA) and Collagen I was detected and found to be increased when PQ treated and restored with additional DKK1 treatment. In summary, these assays indicated that Wnt/β-catenin signaling pathway played a regulatory role in the differentiation of lung epithelial cells and fibroblasts, and the pathogenesis of pulmonary fibrosis related to PQ. Inhibition of the Wnt/β-catenin signaling pathway may be investigated further as a potential fibrosis suppressor for pulmonary fibrosis therapy.
Collapse
Affiliation(s)
- Zhaorui Sun
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Zhizhou Yang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Southern Medical University, Nanjing, 210002, PR China.
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Changbao Huang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Yi Ren
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Fei Gao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Liping Cao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Liang Li
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China.
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, PR China; Department of Emergency Medicine, Jinling Hospital, Southern Medical University, Nanjing, 210002, PR China.
| |
Collapse
|
22
|
Antifibrotic effect of curcumin, N-acetyl cysteine and propolis extract against bisphenol A-induced hepatotoxicity in rats: Prophylaxis versus co-treatment. Life Sci 2020; 260:118245. [PMID: 32791144 DOI: 10.1016/j.lfs.2020.118245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022]
Abstract
AIMS Bisphenol A (BPA) has been shown to induce liver fibrosis in rodents. Therefore, this study examined the protective effect of a triple combination of curcumin (Cur), N-acetyl cysteine (NAC) and propolis (Prp) extract against BPA-induced hepatic fibrosis. METHODS 100 Wistar male rats were equally assigned into 10 groups; one group was designated as control. 10 rats were gavaged with BPA (50 mg/kg/day) for 8 wk and left un-treated (BPA group). The remaining 80 rats were divided into 8 groups, distributed in 2 models. Protective model: rats were daily co-treated with BPA and Cur (100 mg/kg, p.o) or NAC (150 mg/kg, p.o) or Prp (200 mg/kg, p.o) or their combination for 8 wk. Preventive model: rats were daily treated with Cur or NAC or Prp or their combination for 4 wk before BPA administration and then in the same manner as protective model. KEY FINDINGS Current treatment interventions significantly alleviated BPA-induced hepatic damage and fibrosis. They also restored pro-oxidant/antioxidant balance, shifted cytokine balance towards the anti-inflammatory side, decreasing interleukin-1β/interleukin-10 ratio. Moreover, these compounds seem to exert anti-apoptotic effects by increasing the immunoexpression of B-cell lymphoma 2 in hepatocytes and decreasing hepatic caspase-3 content. Finally, they ameliorated extracellular matrix turn over through down-regulation of matrix metalloproteinase-9 and up-regulation of tissue inhibitor of matrix metalloproteinase-2 genetic expression. SIGNIFICANCE Current treatments guarded against BPA-induced hepatic fibrosis due to their antioxidant, anti-inflammatory and anti-apoptotic properties, decreasing extracellular matrix turnover. Interestingly, the triple therapy provided hepatoprotection superior to monotherapy. Besides, prophylactic and concurrent treatments seem to be more effective than concurrent treatments.
Collapse
|
23
|
Yanagihara T, Chong SG, Vierhout M, Hirota JA, Ask K, Kolb M. Current models of pulmonary fibrosis for future drug discovery efforts. Expert Opin Drug Discov 2020; 15:931-941. [PMID: 32396021 DOI: 10.1080/17460441.2020.1755252] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Pulmonary fibrosis includes several lung disorders characterized by progressive fibrosis, of which idiopathic pulmonary fibrosis (IPF) is a particularly severe form with a median survival time of 3-5 years after diagnosis. Although numerous compounds have shown efficacy in attenuating pulmonary fibrosis using animal models, only a few compounds have shown their beneficial effects for IPF in clinical trials. Thus, there is an emergent need to improve the preclinical development process to better identify, characterize and select clinically useful targets. AREAS COVERED In this review, the authors extensively describe current models of pulmonary fibrosis, including rodent models, ex vivo models, and in vitro models. EXPERT OPINION Based upon our current understanding, improving the identification and characterization of clinically relevant molecules or pathways responsible for progressive fibrotic diseases and use of the appropriate preclinical model system to test these will likely be required to improve the drug development pipeline for pulmonary fibrosis. Combination with appropriate preclinical models with ex vivo (precision-cut lung slices) or in vitro models would be beneficial for high-throughput drug discovery or validation of drug effects.
Collapse
Affiliation(s)
- Toyoshi Yanagihara
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada.,Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University , Fukuoka, Japan
| | - Sy Giin Chong
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Megan Vierhout
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Jeremy A Hirota
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Research Institute at St Joseph's Healthcare, Department of Medicine, McMaster University , Hamilton, ON, Canada
| |
Collapse
|
24
|
Zhou J, Wang C, Wu J, Fukunaga A, Cheng Z, Wang J, Yamauchi A, Yodoi J, Tian H. Anti-Allergic and Anti-Inflammatory Effects and Molecular Mechanisms of Thioredoxin on Respiratory System Diseases. Antioxid Redox Signal 2020; 32:785-801. [PMID: 31884805 DOI: 10.1089/ars.2019.7807] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: The pathogenesis and progression of allergic inflammation in the respiratory system are closely linked to oxidative stress. Thioredoxin (TRX) is an essential redox balance regulator in organisms and is induced by various oxidative stress factors, including ultraviolet rays, radiation, oxidation, viral infections, ischemia reperfusion, and anticancer agents. Recent Advances: We demonstrated that systemic administration and transgenic overexpression of TRX is useful in a wide variety of in vivo inflammatory respiratory diseases models, such as viral pneumonia, interstitial lung disease, chronic obstructive pulmonary disease, asthma, acute respiratory distress syndrome, and obstructive sleep apnea syndrome, by removing reactive oxygen species, blocking production of inflammatory cytokines, inhibiting migration and activation of neutrophils and eosinophils, and regulating the cellular redox status. In addition, TRX's anti-inflammatory mechanism is different from the mechanisms associated with anti-inflammatory agents, such as glucocorticoids, which regulate the inflammatory reaction in association with suppressing immune responses. Critical Issues: Understanding the molecular mechanism of TRX is very helpful for understanding the role of TRX in respiratory diseases. In this review, we show the protective effect of TRX in various respiratory diseases. In addition, we discuss its anti-allergic and anti-inflammatory molecular mechanism in detail. Future Directions: The application of TRX may be useful for treating respiratory allergic inflammatory disorders. Antioxid. Redox Signal. 32, 785-801.
Collapse
Affiliation(s)
- JieDong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - CuiXue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - JiaLin Wu
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Atsushi Fukunaga
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - ZuSheng Cheng
- Department of Radiology, Shaoxing Seventh People's Hospital, Shaoxing, China
| | - JinQuan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Akira Yamauchi
- Department of Breast Surgery, Nara Prefectural General Medical Center, Nara, Japan
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China.,Jiaozhimei Biotechnology (Shaoxing) Co., Ltd., Shaoxing, China
| |
Collapse
|
25
|
Wu EK, Henkes ZI, McGowan B, Bell RD, Velez MJ, Livingstone AM, Ritchlin CT, Schwarz EM, Rahimi H. TNF-Induced Interstitial Lung Disease in a Murine Arthritis Model: Accumulation of Activated Monocytes, Conventional Dendritic Cells, and CD21 +/CD23 - B Cell Follicles Is Prevented with Anti-TNF Therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2837-2849. [PMID: 31659014 PMCID: PMC6989047 DOI: 10.4049/jimmunol.1900473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/29/2019] [Indexed: 12/29/2022]
Abstract
Interstitial lung disease (ILD) is a well-known extra-articular manifestation of rheumatoid arthritis (RA). RA-associated ILD (RA-ILD) exists on a wide spectrum, with variable levels of inflammatory and fibrotic activity, although all subtypes are regarded as irreversible pathologic conditions. In both articular and pulmonary manifestations, TNF is a significant pathogenic factor. Whereas anti-TNF therapy alleviates joint pathologic conditions, it exacerbates fibrotic RA-ILD. The TNF-transgenic (TNF-Tg) murine model of RA develops both inflammatory arthritis and an ILD that mimics a cellular nonspecific interstitial pneumonia pattern dominated by an interstitial accumulation of inflammatory cells with minimal-to-absent fibrosis. Given the model's potential to elucidate the genesis of inflammatory RA-ILD, we aim to achieve the following: 1) characterize the cellular accumulations in TNF-Tg lungs, and 2) assess the reversibility of inflammatory ILD following anti-TNF therapy known to resolve TNF-Tg inflammatory arthritis. TNF-Tg mice with established disease were randomized to anti-TNF or placebo therapy and evaluated with imaging, histology, and flow cytometric analyses, together with wild-type controls. Flow cytometry of TNF-Tg versus wild-type lungs revealed significant increases in activated monocytes, conventional dendritic cells, and CD21+/CD23- B cells that are phenotypically distinct from the B cells in inflamed nodes, which are known to accumulate in joint-draining lymph nodes. In contrast to human RA-ILD, anti-TNF treatment significantly alleviated both joint and lung inflammation. These results identify a potential role for activated monocytes, conventional dendritic cells, and CD21+/CD23- B cells in the genesis of RA-ILD, which exist in a previously unknown, reversible, prefibrotic stage of the disease.
Collapse
Affiliation(s)
- Emily K Wu
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Zoe I Henkes
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Brion McGowan
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Richard D Bell
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Moises J Velez
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Alexandra M Livingstone
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Christopher T Ritchlin
- Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Edward M Schwarz
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- Department of Orthopaedics and Rehabilitation, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642; and
| | - Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642;
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
26
|
Wu EK, Eliseeva S, Rahimi H, Schwarz EM, Georas SN. Restrictive lung disease in TNF-transgenic mice: correlation of pulmonary function testing and micro-CT imaging. Exp Lung Res 2019; 45:175-187. [PMID: 31318607 PMCID: PMC6812493 DOI: 10.1080/01902148.2019.1636899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 10/26/2022]
Abstract
Purpose: Micro-computed tomography (µCT) is increasingly being used on animal models as a minimally-invasive longitudinal outcome measure of pulmonary disease progression. However, while imaging can elucidate macroscopic structural changes over the whole lung, µCT is unable to describe the mechanical changes and functional impairments imposed by progressive disease, which can only be measured via pulmonary function tests (PFTs). The tumor necrosis factor-transgenic (TNF-Tg) mouse model of rheumatoid arthritis (RA) develops pulmonary pathology that mimics many aspects of the inflammatory interstitial lung disease (ILD) seen in a subset of patients with RA. Prior studies using µCT imaging of these mice found increased pulmonary density, characteristic of restrictive disease; however, there have been conflicting reports in the literature regarding the obstructive versus restrictive phenotype of this model. Our study looks to 1) define the functional impairments and 2) characterize the restrictive/obstructive nature of the disease found in this model. Materials and Methods: In this study, we performed PFTs at end-stage ILD, and paired these findings with µCT results, correlating radiology to functional parameters. TNF-Tg and WT littermates of both sexes underwent µCT imaging and PFT testing at 5.5 months-old. Spearman's correlation analyses were performed comparing lung tissue volume (LTV) to PFT parameters of gas exchange and tissue stiffness. Results: Compared to WT, TNF-Tg mice had impaired gas exchange capacity, increased respiratory resistance, and reduced lung compliance, elastance, and inspiratory capacity, indicating increased tissue stiffness and compromised pulmonary function. LTV was also consistently higher in TNF-Tg lungs. Conclusions: These findings demonstrate that: 1) TNF-Tg mice display a restrictive pathology, and 2) in vivo µCT is a valid outcome measure to infer changes in pulmonary mechanical and functional parameters.
Collapse
Affiliation(s)
- Emily K. Wu
- Department of Microbiology and Immunology..University of Rochester School of Medicine and Dentistry, Rochester, NY
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sophia Eliseeva
- Department of Microbiology and Immunology..University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Homaira Rahimi
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, NY
- Department of Orthopaedics, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Steve N. Georas
- Department of Microbiology and Immunology..University of Rochester School of Medicine and Dentistry, Rochester, NY
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
27
|
Na JD, Choi YJ, Jun DS, Kim YC. Alleviation of paraquat-induced oxidative lung injury by betaineviaregulation of sulfur-containing amino acid metabolism despite the lack of betaine-homocysteine methyltransferase (BHMT) in the lung. Food Funct 2019; 10:1225-1234. [DOI: 10.1039/c8fo01457d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Betaine regulates sulfur-containing amino acid metabolism in the lung despite the lack of BHMT and increases pulmonary antioxidant capacity.
Collapse
Affiliation(s)
- Jong Deok Na
- College of Pharmacy
- Seoul National University
- Seoul 151-742
- Korea
| | - Yeo Jin Choi
- College of Pharmacy
- Seoul National University
- Seoul 151-742
- Korea
| | - Doo Sung Jun
- College of Pharmacy
- Seoul National University
- Seoul 151-742
- Korea
| | - Young Chul Kim
- College of Pharmacy
- Seoul National University
- Seoul 151-742
- Korea
- Research Institute of Pharmaceutical Sciences
| |
Collapse
|
28
|
Sisson TH, Christensen PJ, Muraki Y, Dils AJ, Chibucos L, Subbotina N, Tohyama K, Horowitz JC, Matsuo T, Bailie M, Nikam S, Hazama M. Phosphodiesterase 4 inhibition reduces lung fibrosis following targeted type II alveolar epithelial cell injury. Physiol Rep 2018; 6:e13753. [PMID: 29952109 PMCID: PMC6021279 DOI: 10.14814/phy2.13753] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/18/2018] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Fibrosis of the lung constitutes a major clinical challenge and novel therapies are required to alleviate the associated morbidity and mortality. Investigating the antifibrotic efficacy of drugs that are already in clinical practice offers an efficient strategy to identify new therapies. The phosphodiesterase 4 (PDE4) inhibitors, approved for the treatment of chronic obstructive pulmonary disease, harbor therapeutic potential for pulmonary fibrosis by augmenting the activity of endogenous antifibrotic mediators that signal through cyclic AMP. In this study, we tested the efficacy of several PDE4 inhibitors including a novel compound (Compound 1) in a murine model of lung fibrosis that results from a targeted type II alveolar epithelial cell injury. We also compared the antifibrotic activity of PDE4 inhibition to the two therapies that are FDA-approved for idiopathic pulmonary fibrosis (pirfenidone and nintedanib). We found that both preventative (day 0-21) and therapeutic (day 11-21) dosing regimens of the PDE4 inhibitors significantly ameliorated the weight loss and lung collagen accumulation that are the sequelae of targeted epithelial cell damage. In a therapeutic protocol, the reduction in lung fibrosis with PDE4 inhibitor administration was equivalent to pirfenidone and nintedanib. Treatment with this class of drugs also resulted in a decrease in plasma surfactant protein D concentration, a reduction in the plasma levels of several chemokines implicated in lung fibrosis, and an in vitro inhibition of fibroblast profibrotic gene expression. These results motivate further investigation of PDE4 inhibition as a treatment for patients with fibrotic lung disease.
Collapse
Affiliation(s)
- Thomas H. Sisson
- Pulmonary and Critical Care DivisionDepartment of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMichigan
| | - Paul J. Christensen
- Division of Pulmonary & Critical Care MedicineDepartment of Internal MedicineWilliam Beaumont Medical CenterTroyMichigan
| | - Yo Muraki
- Takeda Pharmaceutical Company LimitedFujisawaJapan
| | - Anthony J. Dils
- Pulmonary and Critical Care DivisionDepartment of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMichigan
| | - Lauren Chibucos
- Pulmonary and Critical Care DivisionDepartment of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMichigan
| | - Natalya Subbotina
- Pulmonary and Critical Care DivisionDepartment of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMichigan
| | | | - Jeffrey C. Horowitz
- Pulmonary and Critical Care DivisionDepartment of Internal MedicineUniversity of Michigan Medical CenterAnn ArborMichigan
| | | | - Marc Bailie
- In Vivo FacilityDepartment of Pharmacology and ToxicologyMichigan State UniversityEast LansingMichigan
| | - Sham Nikam
- Takeda Pharmaceutical Company LimitedFujisawaJapan
| | | |
Collapse
|
29
|
|
30
|
Protective Effect of Infliximab, a Tumor Necrosis Factor-Alfa Inhibitor, on Bleomycin-Induced Lung Fibrosis in Rats. Inflammation 2016; 39:65-78. [PMID: 26253295 DOI: 10.1007/s10753-015-0224-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We aimed to investigate the preventive effect of Infliximab (IFX), a tumor necrosis factor (TNF)-α inhibitor, on bleomycin (BLC)-induced lung fibrosis in rats. Rats were assigned into four groups as follows: I-BLC group, a single intra-tracheal BLC (2.5 mg/kg) was installed; II-control group, a single intra-tracheal saline was installed; III-IFX + BLC group, a single-dose IFX (7 mg/kg) was administered intraperitoneally (i.p.), 72 h before the intra-tracheal BLC installation; IV-IFX group, IFX (7 mg/kg) was administered alone i.p. on the same day with IFX + BLC group. All animals were sacrificed on the 14th day of BLC installation. Levels of tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β, interleukin (IL)-6, periostin, YKL-40, nitric oxide (NO) in rat serum were measured, as well as, myeloperoxidase (MPO), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) activity, and reduced glutathione (GSH), hydroxyproline, malondialdehyde (MDA) content in lung homogenates. Lung tissues were stained with hematoxylin and eosin (H&E) for quantitative histological evaluation. The inducible nitric oxide synthase (iNOS) expression and cell apoptosis in the lung tissues were determined quantitatively by immunohistochemical staining (INOS) and by TUNNEL staining, respectively. BLC installation worsened antioxidant status (such as SOD, CAT, GPx, GSH, MPO), while it increased the serum TNF-α, TGF-β, IL-6, periostin, YKL-40, and lipid peroxidation, and collagen deposition, measured by MDA and hydroxyproline, respectively. IFX pretreatment improved antioxidant status as well as BLC-induced lung pathological changes, while it decreased the TNF-α, TGF-β, IL-6, periostin, YKL-40, lipid peroxidation and collagen deposition. Finally, histological, immunohistochemical, and TUNNEL evidence also supported the ability of IFX to prevent BLC-induced lung fibrosis. The results of the present study indicate that IFX pretreatment can attenuate BLC-induced pulmonary fibrosis.
Collapse
|
31
|
Elswefy SES, Abdallah FR, Atteia HH, Wahba AS, Hasan RA. Inflammation, oxidative stress and apoptosis cascade implications in bisphenol A-induced liver fibrosis in male rats. Int J Exp Pathol 2016; 97:369-379. [PMID: 27925325 DOI: 10.1111/iep.12207] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/15/2016] [Indexed: 12/15/2022] Open
Abstract
Bisphenol A (BPA) is a key monomer in the production of plastics. It has been shown to be hepatotoxic. Inflammation and oxidative stress are closely linked with liver fibrosis, the major contributing factor to hepatic failure. Therefore, the aim of this study was to evaluate the impact of chronic exposure to BPA on the development of hepatic fibrosis in male rats and to determine the cross-talk between the hepatic cytokine network, oxidative stress and apoptosis. For this purpose, 30 male Wistar albino rats were divided into three equal groups as follows: the first group was given no treatment (normal control group); the second group was given corn oil once daily by oral gavage for 8 weeks (vehicle control group); and the third group received BPA (50 mg/kg body weight/day, p.o.) for 8 weeks. BPA administration induced liver fibrosis as reflected in an increase in serum hepatic enzymes activities, hepatic hydroxyproline content and histopathological changes particularly increased collagen fibre deposition around the portal tract. In addition, there was inflammation (as reflected in increase in interleukin-1beta 'IL-1β', decrease in interleukin-10 'IL-10' serum levels and increase in IL-1β/IL-10 ratio), oxidative stress (as reflected in increase in malondialdehyde (MDA) level, reduction in reduced glutathione (GSH) content and inhibition of catalase (CAT) activity) and apoptosis [as reflected in an increase in caspase-3 level and a decrease in numbers of B-cell lymphoma 2 (BCL2)-immunopositive hepatocytes]. Interestingly, BPA had an upregulating effect on an extracellular matrix turnover gene [as reflected in matrix metalloproteinase-9 (MMP-9)] and a downregulating effect on its inhibitor gene [as reflected in tissue inhibitor of matrix metalloproteinase-2 (TIMP-2)] expression. Thus, the mechanism by which BPA induced liver fibrosis seems to be related to stimulation of the inflammatory response, along with oxidative stress, the apoptotic pathway and activation of extracellular matrix turnover.
Collapse
Affiliation(s)
- Sahar El-Sayed Elswefy
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia Governorate, Egypt
| | - Fatma Rizk Abdallah
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia Governorate, Egypt
| | - Hebatallah Husseini Atteia
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia Governorate, Egypt
| | - Alaa Samir Wahba
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Sharkia Governorate, Egypt
| | - Rehab Abdallah Hasan
- Department of Histology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
32
|
Kim HS, Kim HG, Lee HW, Lee SB, Lee JS, Im HJ, Kim WY, Lee DS, Son CG. A Herbal Formula, CGXII, Exerts Antihepatofibrotic Effect in Dimethylnitrosamine-Induced SD Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:5093718. [PMID: 27340416 PMCID: PMC4907344 DOI: 10.1155/2016/5093718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/20/2016] [Accepted: 04/11/2016] [Indexed: 01/20/2023]
Abstract
We aimed to evaluate the antihepatofibrotic effects of CGXII, an aqueous extract which is composed of A. iwayomogi, A. xanthioides, and S. miltiorrhiza, against dimethylnitrosamine- (DMN-) induced hepatofibrosis. Male Sprague Dawley rats were intraperitoneally injected with 10 mg/kg of DMN for 4 weeks (three consecutive days weekly). Rats were orally given distilled water, CGXII (50 or 100 mg/kg), or dimethyl dimethoxy biphenyl dicarboxylate (50 mg/kg) daily. DMN injection caused substantial alteration of total body weight and liver and spleen mass, whereas they were notably normalized by CGXII. CGXII treatment also markedly attenuated the elevation of serum aspartate aminotransferase and alanine aminotransferase levels, hepatic lipid peroxidation, and protein carbonyl contents. Collagen accumulation in hepatic tissue evidenced by histopathological analysis and quantitative assessment of hepatic hydroxyproline was ameliorated by CGXII. Immunohistochemistry analysis revealed decreased α-smooth muscle actin supporting the antihepatofibrotic effect of CGXII. The profibrogenic cytokines transforming growth factor-β, platelet-derived growth factor-β, and connective tissue growth factor were increased by DMN injection. Administration of CGXII normalized the protein and gene expression levels of these cytokines. Our findings suggest that CGXII lowers the levels of profibrogenic cytokines and thereby exerts antifibrotic effects.
Collapse
Affiliation(s)
- Hyo-Seon Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Hyeong-Geug Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Hye-Won Lee
- TKM-Based Herbal Drug Research Group, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Sung-Bae Lee
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Hwi-Jin Im
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Won-Yong Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| | - Dong-Soo Lee
- Department of Internal Medicine, Daejeon St. Mary's Hospital, The Catholic University of Korea, 64 Daeheung-ro, Jung-gu, Daejeon 34943, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Daejeon Oriental Hospital, Oriental Medical College, Daejeon University, 176-9 Daeheung-ro, Jung-gu, Daejeon 34929, Republic of Korea
| |
Collapse
|
33
|
Byrne AJ, Maher TM, Lloyd CM. Pulmonary Macrophages: A New Therapeutic Pathway in Fibrosing Lung Disease? Trends Mol Med 2016; 22:303-316. [PMID: 26979628 DOI: 10.1016/j.molmed.2016.02.004] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 12/20/2022]
Abstract
Pulmonary fibrosis (PF) is a growing clinical problem which can result in breathlessness or respiratory failure and has an average life expectancy of 3 years from diagnosis. Therapeutic options for PF are limited and there is therefore a significant unmet clinical need. The recent resurgent interest in macrophage biology has led to a new understanding of lung macrophage origins, biology, and phenotypes. In this review we discuss fibrotic mechanisms and focus on the role of macrophages during fibrotic lung disease. Data from both human and murine studies are reviewed, highlighting novel macrophage-orientated biomarkers for disease diagnosis and potential targets for future anti-fibrotic therapies.
Collapse
Affiliation(s)
- Adam J Byrne
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK.
| | - Toby M Maher
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK; National Institute for Health Research (NIHR) Respiratory Biomedical Research Unit, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Clare M Lloyd
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK
| |
Collapse
|
34
|
Epigenetic regulation of cyclooxygenase-2 by methylation of c8orf4 in pulmonary fibrosis. Clin Sci (Lond) 2016; 130:575-86. [PMID: 26744410 PMCID: PMC4782165 DOI: 10.1042/cs20150697] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/07/2016] [Indexed: 02/01/2023]
Abstract
The present study demonstrates that hypermethylation and silencing of chromosome 8 open reading frame 4 (thyroid cancer protein 1, TC-1) (c8orf4), a transcriptional regulator of cyclooxygenase-2 (COX-2), is a major contributor to failure of fibroblasts to up-regulate COX-2 in pulmonary fibrosis. DNA methyltransferase (DNMT) inhibition reduces c8orf4 methylation, restores COX-2 expression and normalizes fibroblast function. Fibroblasts derived from the lungs of patients with idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc) produce low levels of prostaglandin (PG) E2, due to a limited capacity to up-regulate cyclooxygenase-2 (COX-2). This deficiency contributes functionally to the fibroproliferative state, however the mechanisms responsible are incompletely understood. In the present study, we examined whether the reduced level of COX-2 mRNA expression observed in fibrotic lung fibroblasts is regulated epigenetically. The DNA methylation inhibitor, 5-aza-2′-deoxycytidine (5AZA) restored COX-2 mRNA expression by fibrotic lung fibroblasts dose dependently. Functionally, this resulted in normalization of fibroblast phenotype in terms of PGE2 production, collagen mRNA expression and sensitivity to apoptosis. COX-2 methylation assessed by bisulfite sequencing and methylation microarrays was not different in fibrotic fibroblasts compared with controls. However, further analysis of the methylation array data identified a transcriptional regulator, chromosome 8 open reading frame 4 (thyroid cancer protein 1, TC-1) (c8orf4), which is hypermethylated and down-regulated in fibrotic fibroblasts compared with controls. siRNA knockdown of c8orf4 in control fibroblasts down-regulated COX-2 and PGE2 production generating a phenotype similar to that observed in fibrotic lung fibroblasts. Chromatin immunoprecipitation demonstrated that c8orf4 regulates COX-2 expression in lung fibroblasts through binding of the proximal promoter. We conclude that the decreased capacity of fibrotic lung fibroblasts to up-regulate COX-2 expression and COX-2-derived PGE2 synthesis is due to an indirect epigenetic mechanism involving hypermethylation of the transcriptional regulator, c8orf4.
Collapse
|
35
|
Miltonprabu S, Nazimabashir, Manoharan V. Hepatoprotective effect of grape seed proanthocyanidins on Cadmium-induced hepatic injury in rats: Possible involvement of mitochondrial dysfunction, inflammation and apoptosis. Toxicol Rep 2015; 3:63-77. [PMID: 28959524 PMCID: PMC5615429 DOI: 10.1016/j.toxrep.2015.11.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/24/2015] [Accepted: 11/28/2015] [Indexed: 01/14/2023] Open
Abstract
The present study was undertaken to evaluate the possible ameliorative role of grape seed proanthocyanidins (GSP) against Cadmium (Cd) induced hepatic inflammation, apoptosis and hepatic mitochondrial toxicity in rats. Male Wistar rats were distributed in four experimental groups: control, GSP, Cd and Cd + GSP. Exposure to a hepatotoxic dose of Cd (5 mg/kg BW) caused liver damage, coupled with enhanced reactive oxygen species (ROS) generation, increased inflammation and apoptosis in liver with increased DNA damage in hepatocytes of rats. Mitochondria were isolated from the hepatic tissues of rats from each group. Our results showed significant decrease in the tri-carboxylic acid cycle enzymes, increased mitochondrial swelling, inhibition of cytochrome c oxidase activity and complex I-III, II-III and IV mediated electron transfer, decreased mitochondrial ATPases, a reduction in calcium content and mitochondrial oxygen consumption in Cd treated rats. All these molecular changes caused by Cd were alleviated by the pre-supplementation with GSP (100 mg/kg BW). The ultra structural changes in the liver also support our findings. From our results, it is clearly indicated that the free radical scavenging, metal chelating and antioxidant potentials of GSP might be the possible reason, responsible for the rescue action against Cd induced mitochondrial damage in the liver of rats.
Collapse
Affiliation(s)
- Selvaraj Miltonprabu
- Department of Zoology, Faculty of Science Annamalai University, Annamalainagar, 608002 Tamilnadu, India
| | - Nazimabashir
- Department of Zoology, Faculty of Science Annamalai University, Annamalainagar, 608002 Tamilnadu, India
| | - Vaihundam Manoharan
- Department of Zoology, Faculty of Science Annamalai University, Annamalainagar, 608002 Tamilnadu, India
| |
Collapse
|
36
|
Kim HG, Han JM, Lee JS, Suk Lee J, Son CG. Ethyl acetate fraction of Amomum xanthioides improves bile duct ligation-induced liver fibrosis of rat model via modulation of pro-fibrogenic cytokines. Sci Rep 2015; 5:14531. [PMID: 26412144 PMCID: PMC4585957 DOI: 10.1038/srep14531] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 07/15/2015] [Indexed: 12/13/2022] Open
Abstract
We investigated anti-hepatofibrotic effects of ethyl acetate fraction of Ammomum xanthoides (EFAX) using bile duct ligation (BDL)-induced hepatic fibrosis in a rat model. Male SD rats (6 weeks old) underwent BDL followed by 15 days of orall administration of EFAX (12.5, 25 or 50 mg/kg) or ursodeoxycholic acid (25 mg/kg). BDL caused animal death, ascites formation, alterations in serum biochemistries, and severe hepatic injury with excessive collagen deposition, whereas EFAX treatment significantly attenuated these effects. BDL markedly increased the pro-fibrogenic cytokines (TGF-β, PDGF-β, and CTGF) and the extracellular matrix indicators α-SMA, TIMP-1 and collagen type 1 in hepatic proteins and gene expression levels, which were notably normalized by EFAX treatment. EFAX also markedly normalized pro-fibrogenic signaling molecules including Smad2/3, Smad7, Akt, p44/42, and p38. We further explored EFAX mechanisms of actions using LX-2 cells (human derived hepatic stellate cell line). Pre-treatment with EFAX drastically attenuated the activation of α-SMA and Smad2/3, which are downstream molecules of TGF-β. These findings suggest that EFAX may be a potent anti-hepatofibrotic agent, and its corresponding mechanisms primarily involve the modulation of pro-fibrogenic cytokines.
Collapse
Affiliation(s)
- Hyeong-Geug Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, South Korea
| | - Jong-Min Han
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, South Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, South Korea
| | - Jong Suk Lee
- GyeongGi Bio-Center, GSTEP, 864-1 Iui-dong, Yeongtong-gu, Suwon, Gyeonggi-do, South Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, South Korea
| |
Collapse
|
37
|
Park HJ, Kim HG, Wang JH, Choi MK, Han JM, Lee JS, Son CG. Comparison of TGF-β, PDGF, and CTGF in hepatic fibrosis models using DMN, CCl4, and TAA. Drug Chem Toxicol 2015; 39:111-118. [PMID: 26045230 DOI: 10.3109/01480545.2015.1052143] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 03/31/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022]
Abstract
Three chemotoxins including dimethylnitrosamine (DMN), carbon tetrachloride (CCl4), and thioacetamide (TAA) are commonly used in hepatofibrotic models. We aimed to draw characteristics of histopathology and pro-fibrogenic cytokines including TGF-β, PDGF and CTGF among three models. Rats were divided into six groups and intra-peritoneally injected with DMN (10 mg/kg, for three weeks, three consecutive days weekly), CCl4 (1.6 g/kg, for 10 weeks, twice weekly), TAA (200 mg/kg, for 12 weeks, twice weekly) or their corresponded treatment for each control group. The liver weights were decreased in DMN model, but not other models. Ascites were occurred as 3-, 2-, and 7-rats in DMN, CCl4, and TAA model, respectively. The lipid peroxidation was highest in CCl4 model, serum levels of liver enzymes were increased as similar severity. The hepatofibrotic alterations were remarkable in DMN and TAA model, but not CCl4 as evidenced by the Masson trichrome staining and hydroxyproline. The immunohistochemistry for α-SAM showed that the DMN model was most severely enhanced than other models. On the other hand, hepatic tissue levels of pro-fibrogenic cytokines including TGF-β, PDGF, and CTGF were generally increased in three models, but totally different among models or measurement resources. Especially, serum levels of three cytokines were remarkably increased by CCl4 injection and CTGF levels in both hepatic tissue and serum were highest in CCl4 group. Our results firstly demonstrated comparative study for features of morphological finding and pro-fibrogenic cytokines in serum and hepatic protein levels among three models. Above results would be a helpful reference for hepatofibrotic studies.
Collapse
Affiliation(s)
- Hye-Jung Park
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Hyeong-Geug Kim
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jing-Hua Wang
- b Laboratory of Medical Science , Dongguk University Medical Center, Dongguk University , Republic of Korea
| | - Min-Kyung Choi
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jong-Min Han
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Jin-Seok Lee
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| | - Chang-Gue Son
- a Department of Liver and Immunology Research Center , Daejeon Oriental Hospital of Daejeon University , Republic of Korea and
| |
Collapse
|
38
|
Wang C, Dai J, Sun Z, Shi C, Cao H, Chen X, Gu S, Li Z, Qian W, Han X. Targeted inhibition of disheveled PDZ domain via NSC668036 depresses fibrotic process. Exp Cell Res 2015; 331:115-122. [DOI: 10.1016/j.yexcr.2014.10.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 12/16/2022]
|
39
|
Arafa MH, Mohamed DA, Atteia HH. Ameliorative effect of N-acetyl cysteine on alpha-cypermethrin-induced pulmonary toxicity in male rats. ENVIRONMENTAL TOXICOLOGY 2015; 30:26-43. [PMID: 23900960 DOI: 10.1002/tox.21891] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 06/22/2013] [Accepted: 06/26/2013] [Indexed: 06/02/2023]
Abstract
Alpha-cypermethrin (α-CYP) is one of the most widely used insecticides. It may become an air pollutant and adversely affect the health. The present study was designed to determine whether treatment with N-acetyl cysteine (NAC), a well-known antioxidant, can be useful for the management of the deleterious effects of α-CYP on lung tissues. For this purpose, thirty two male rats were divided into four different groups (eight rats for each). Group (I) gavaged with corn oil (control group), group (II) gavaged daily with NAC (150 mg kg(-1) body weight), group (III) gavaged with α-CYP (14.5 mg kg(-1) body weight/day, dissolved in corn oil), group (IV) gavaged with NAC then with α-CYP 2 h later for 12 weeks. α-CYP significantly increased serum lactate dehydrogenase (LDH) and pulmonary malondialdehyde (MDA) levels, while decreased the activities of catalase (CAT) and superoxide dismutase (SOD) as well as reduced glutathione (GSH) content in lung. It also provoked higher levels of serum nitric oxide (NO), lung interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), hydroxyproline (Hyp) as well as heme oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS) and nuclear factor-kappa B (NF-К B) gene expression in lung tissues. Histopathological alterations in lung with congestion, cellular infiltration, necrotic changes and thickening of inter-alveolar septa were observed following α-CYP administration. NAC reduced the adverse effects of α-CYP on lung tissues and improved the histological architecture of lung since it showed antioxidant, anti-inflammatory and antifibrotic effects on lung tissues. Our results indicate that NAC exerts a potent protective effect against α-CYP-induced oxidative damage and inflammation in lung tissues.
Collapse
Affiliation(s)
- Manar Hamed Arafa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Sharkia Government, Egypt
| | | | | |
Collapse
|
40
|
Kim HG, Kim JM, Han JM, Lee JS, Choi MK, Lee DS, Park YH, Son CG. Chunggan extract, a traditional herbal formula, ameliorated alcohol-induced hepatic injury in rat model. World J Gastroenterol 2014; 20:15703-15714. [PMID: 25400454 PMCID: PMC4229535 DOI: 10.3748/wjg.v20.i42.15703] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/14/2014] [Accepted: 06/21/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate protective effects of Chunggan extract (CGX), a traditional herbal formula, under 4 wk of alcohol consumption-induced liver injury. METHODS Male Sprague-Dawley Rats were orally administered 30% ethanol daily for 4 wk with or without CGX. The pharmaceutical properties were assessed through liver enzymes, histopathology, fibrogenic cytokines, and alcohol metabolism in hepatic tissues as well as by in vitro experiment using HSC-T6 cells. RESULTS Four weeks of alcohol consumption notably increased liver enzymes and malondialdehyde levels in serum and hepatic tissue. CGX not only prevented the collagen deposition determined by histopathology and hydroxyproline content, but also normalized transforming growth factor-beta, platelet-derived growth factor-beta and connective tissue growth factor at the gene expression and protein levels in liver tissue. Moreover, CGX treatment also significantly normalized the abnormal changes in gene expression profiles of extracellular matrix proteins, matrix metalloproteinase and their inhibitors, alcohol metabolism, and inflammatory reactions. In the acetaldehyde-stimulated HSC-T6 cells, CGX considerably inhibited collagen production and normalized fibrogenic cytokines in both gene expression and protein levels. CONCLUSION The present study evidenced that CGX has hepatoprotective properties via modulation of fibrogenic cytokines and alcohol metabolism in alcoholic liver injury.
Collapse
|
41
|
Arafa MH, Mohammad NS, Atteia HH. Fenugreek seed powder mitigates cadmium-induced testicular damage and hepatotoxicity in male rats. ACTA ACUST UNITED AC 2014; 66:293-300. [DOI: 10.1016/j.etp.2014.04.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/27/2014] [Accepted: 04/07/2014] [Indexed: 12/14/2022]
|
42
|
Arafa MH, Mohammad NS, Atteia HH, Abd-Elaziz HR. Protective effect of resveratrol against doxorubicin-induced cardiac toxicity and fibrosis in male experimental rats. J Physiol Biochem 2014; 70:701-11. [PMID: 24939721 DOI: 10.1007/s13105-014-0339-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 06/02/2014] [Indexed: 11/29/2022]
Abstract
The possible effectiveness of resveratrol, a polyphenol present in different plants comprising berries, grapes and peanuts, on the prevention of doxorubicin-induced cardiac toxicity and fibrosis was investigated. Forty adult male Wistar albino rats were divided into four groups. Group I received normal saline, group II gavaged with resveratrol (20 mg/kg, daily for 4 weeks), group III received doxorubicin (2.5 mg/kg i.p. in six injections for 2 weeks; accumulative dose of 15 mg/kg), and group IV received doxorubicin + resveratrol (starting resveratrol intake 2 weeks before doxorubicin administration). Resveratrol significantly alleviated the increase in left ventricular lipid peroxidation, hydroxyproline, and tumor necrosis factor alpha levels as well as serum creatine kinase-myocardial band (CK-MB) activity and prevented the decrease in body and heart weights in doxorubicin-treated group. However, a marked protection against reduced glutathione content depletion and superoxide dismutase activity reduction was observed in the left ventricles of rats pretreated with resveratrol in combination with doxorubicin. Resveratrol also ameliorated the up-regulation of left ventricular caspase-3 and transforming growth factor-beta1 gene expression as well as left ventricular histopathological changes including necrosis and fibrosis induced by doxorubicin. Collectively, our results suggest that resveratrol provides a significant protection against doxorubicin-induced cardiotoxicity and fibrosis in rats. Therefore, it may be used as a promising cardioprotective agent in patients treated with doxorubicin due to malignant diseases. So, further clinical trials are required to confirm these findings.
Collapse
Affiliation(s)
- Manar Hamed Arafa
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, Sharkia Gov, Egypt,
| | | | | | | |
Collapse
|
43
|
Lin X, Zhang S, Huang R, Wei L, Tan S, Liang S, Tian Y, Wu X, Lu Z, Huang Q. Helenalin attenuates alcohol-induced hepatic fibrosis by enhancing ethanol metabolism, inhibiting oxidative stress and suppressing HSC activation. Fitoterapia 2014; 95:203-13. [PMID: 24704336 DOI: 10.1016/j.fitote.2014.03.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/23/2014] [Accepted: 03/25/2014] [Indexed: 11/21/2022]
Abstract
A compound was isolated from Centipeda minima using bioassay-guided screening. The structure of this compound was elucidated based on its spectral data, and it was identified as helenalin. The hepatoprotective effect of helenalin was evaluated using a liver fibrosis model induced by intragastric administration with alcohol within 24 weeks in rats. The results revealed that helenalin significantly prevented alcohol-induced hepatic injury and fibrogenesis, as evidenced by the decrease in serum aminotransferase, the attenuation of histopathological changes, and the inhibition of the hepatic fibrosis indicators, such as hyaluronic acid, type III precollagen, laminin, hydroxyproline and collagen α type I. Mechanistically, studies showed that helenalin expedited ethanol metabolism by enhancing the alcohol and aldehyde dehydrogenase activities. Furthermore, helenalin alleviated lipid peroxidation, recruited the antioxidative defense system, inhibited CYP2E1 activity, and reduced the inflammatory mediators, including TGF-β1, TNF-α, IL-6 and IL-1β and myeloperoxidase, via down-regulation of NF-κB. Helenalin significantly decreased collagen deposition by reducing the profibrotic cytokines like transforming growth factor-β, platelet-derived growth factor-β and connective tissue growth factor, and promoted extracellular matrix degradation by modulating the levels of tissue inhibitor of matrix metalloproteinase-1 and matrix metalloproteinase-9. In addition, helenalin inhibited HSC activation as evidenced by the down-regulation of α-SMA and TGF-β levels. In conclusion, helenalin had a significant protective effect on chronic ethanol-induced hepatic fibrosis and may be a major bioactive ingredient of C. minima.
Collapse
Affiliation(s)
- Xing Lin
- Guangxi Medical University, Nanning 530021, China
| | - Shijun Zhang
- Guangxi Medical University, Nanning 530021, China
| | - Renbin Huang
- Guangxi Medical University, Nanning 530021, China
| | - Ling Wei
- Guangxi Medical University, Nanning 530021, China
| | - Shimei Tan
- Guangxi Medical University, Nanning 530021, China
| | - Shuang Liang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Yuanchun Tian
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Xiaoyan Wu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Zhongpeng Lu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China
| | - Quanfang Huang
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, China.
| |
Collapse
|
44
|
Turgut B, Eren K, Akın MM, Demir T, Kobat S. Topical infliximab for the suppression of wound healing following experimental glaucoma filtration surgery. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:421-9. [PMID: 24851041 PMCID: PMC4018311 DOI: 10.2147/dddt.s63320] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background The purpose of this work was to look into the effects of infliximab on wound healing in experimental glaucoma filtration surgery and to compare the antifibrotic effects of this agent to that of mitomycin-C (MMC). Methods Twenty-eight male New Zealand White rabbits were randomly assigned to four groups, each including seven rabbits: control group, sham group, MMC group, and infliximab group. The rabbits in the control group were not operated on and did not receive any treatment. The rabbits in the sham group underwent trabeculectomy and had one drop of saline instilled four times a day for 14 days. The rabbits in the MMC treatment group underwent trabeculectomy, and a sponge soaked in 0.4 mg/mL MMC was applied intraoperatively to the scleral surgical site for three minutes. The rabbits in the infliximab treatment group underwent trabeculectomy and one drop of 10 mg/mL infliximab was instilled four times a day for 14 days after surgery. On day 14 of the experiment, the operated and control eyes were enucleated and histologically and immunohistochemically analyzed. Results The mean fibroblast and mononuclear cell (MNC) numbers and the mean immunostaining intensities of transforming growth factor-β (TGF-β), fibroblast growth factor-β (FGF-β), and platelet-derived growth factor (PDGF) in the sham group were higher than those of the control group (P<0.01). The mean fibroblast and MNC numbers and the mean immunostaining intensities of TGF-β, FGF-β, and PDGF in the MMC and infliximab groups were statistically significantly lower than those of the sham group (P<0.01). The mean fibroblast and MNC numbers and the mean TGF-β, FGF-β, and PDGF immunostaining intensities of the MMC and infliximab groups were similar (P>0.05). Conclusion Our study suggests that topical infliximab effectively suppresses the subconjunctival wound healing response after experimental glaucoma filtration surgery, reducing the MNC and fibroblast numbers and immunostaining intensities of TGF-β, FGF-β, and PDGF.
Collapse
Affiliation(s)
- Burak Turgut
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Kenan Eren
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Mehmet Mustafa Akın
- Department of Pathology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Tamer Demir
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| | - Sabiha Kobat
- Department of Ophthalmology, School of Medicine, Fırat University, Elazığ, Turkey
| |
Collapse
|
45
|
Redente EF, Keith RC, Janssen W, Henson PM, Ortiz LA, Downey GP, Bratton DL, Riches DWH. Tumor necrosis factor-α accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages. Am J Respir Cell Mol Biol 2014; 50:825-37. [PMID: 24325577 PMCID: PMC4068926 DOI: 10.1165/rcmb.2013-0386oc] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/04/2013] [Indexed: 01/13/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α(-/-) mice by measuring hydroxyproline levels, static compliance, and Masson's trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α(-/-) mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α-induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Rebecca C. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - William Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - Peter M. Henson
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | - Luis A. Ortiz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh Pennsylvania
| | - Gregory P. Downey
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | | | - David W. H. Riches
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
46
|
Kim JM, Kim HG, Han JM, Lee JS, Lee HW, Choi MK, Son CG. The herbal formula CGX ameliorates the expression of vascular endothelial growth factor in alcoholic liver fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2013; 150:892-900. [PMID: 24095833 DOI: 10.1016/j.jep.2013.09.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 08/23/2013] [Accepted: 09/22/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGIC RELEVANCE The Chunggan extract (CGX) is a traditional herbal formula prescribed for patients suffering from various liver diseases, including alcoholic liver disease, in which the mechanism of CGX action remains unclear. This study aimed to investigate the anti-hepatic fibrosis effects of CGX and its underlying mechanisms in alcohol-induced rat livers. MATERIALS AND METHODS To elucidate the mechanism of action of CGX, we evaluated gene expression profiles in the livers of rats treated with 30% alcohol and anti-fibrotic doses of CGX of 100 and 200 mg/kg/day at 1 day, and 2 and 4 weeks using microarrays. The mRNA and protein expression levels of vascular endothelial growth factor (VEGF), one of the candidate genes selected in this study, in alcohol-induced rat livers were measured by real-time PCR and enzyme-linked immunosorbent assays, respectively. RESULTS We identified 4128 genes as differentially expressed by at least twofold between alcohol-only- and alcohol-CGX-fed rats at various doses and time points, compared to naïve control animals. Twenty-three of these genes were associated with liver fibrosis and oxidative stress based on the GeneCards database, resulting in p<0.05 by ANOVA between the alcohol-only and alcohol-CGX groups. Especially, Vegf was decreased in CGX 200 mg/kg/day-fed rat livers at all time points evaluated, and mRNA and protein levels at the 4-week time point were validated. CONCLUSION These gene expression profiles provide a better understanding of the mechanisms underlying the anti-fibrotic effects of CGX. Suppression of VEGF may play a critical role in anti-fibrotic action of CGX in alcoholic liver injury.
Collapse
Affiliation(s)
- Jung Min Kim
- (a)Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 22-5 Daeheung-dong, Jung-gu, Daejeon 301-724, Republic of Korea; (b)NAR Center, Inc., Daejeon Oriental Hospital of Daejeon University, Daejeon 301-724, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
47
|
Kim HG, Wang JH, Han JM, Hwang SY, Lee DS, Son CG. Chunggan extract (CGX), a traditional Korean herbal medicine, exerts hepatoprotective effects in a rat model of chronic alcohol consumption. Phytother Res 2013; 27:1854-1862. [PMID: 23460575 DOI: 10.1002/ptr.4935] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 12/27/2012] [Accepted: 01/07/2013] [Indexed: 12/25/2022]
Abstract
Chunggan extract, CGX, is a modification of a traditional herbal medicine that has been used for patients suffering from various liver disorders since 2001. Here, we investigated the hepatoprotective effects of CGX and its underlying mechanisms in a rat model of chronic alcohol consumption. Rats were orally administered 30% ethanol solution for 4 weeks with or without CGX (50, 100, 200 mg/kg). The histopathology, biochemistry, oxidative stress/antioxidant biomarkers, hepatofibrogenic cytokines, and serum endotoxin level were analyzed. Alcohol treatment markedly elevated the serum levels of aspartate transaminase, alkaline phosphatase, and total reactive oxygen species, and tissue levels of hydroxyproline and malondialdehyde (MDA), while reducing the total glutathione (GSH) contents and the activities of superoxide dismutase and catalase. These alterations were significantly attenuated by CGX treatment (mainly 100 and 200 mg/kg). CGX treatment normalized the elevation of fibrogenic cytokines, including transforming growth factor-β, platelet derived growth factor-β, and connective tissue growth factor in hepatic tissues and ameliorated the increase in serum endotoxin concentration. These results suggest that CGX protects liver tissue from alcohol injury through antioxidant actions and prevention of endotoxin reflux. .
Collapse
Affiliation(s)
- Hyeong Geug Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Oriental Medical Collage of Daejeon University, 22-5 Daehung-dong, Jung-gu, Daejeon, 301-724, Republic of Korea
| | | | | | | | | | | |
Collapse
|
48
|
Protective role of JAK/STAT signaling against renal fibrosis in mice with unilateral ureteral obstruction. Clin Immunol 2013; 150:78-87. [PMID: 24333535 DOI: 10.1016/j.clim.2013.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 10/20/2013] [Accepted: 11/02/2013] [Indexed: 02/04/2023]
Abstract
Inflammation is involved in renal fibrosis, a final common pathway for kidney diseases. To clarify how JAK/STAT/SOCS system was involved in renal fibrosis, UUO was induced in BALB/c or SOCS3(+/-) mice in the presence or absence of JAK inhibitor-incorporated nanoparticle (pyridine6-PGLA). UUO increased pSTAT3 and subsequently elevated SOCS3 levels in the obstructed kidneys. pSTAT3 levels were further increased in SOCS3(+/-) mice. UUO-induced renal fibrosis was markedly suppressed in SOCS3(+/-) mice, while it was aggravated by pre-treatment with pyridine6-PGLA. Although there were no differences in renal mRNA levels of TGF-β and collagens between wild and SOCS3(+/-) mice, MMP-2 activity was enhanced in SOCS3(+/-) UUO mice. Activated MMP-2 was completely suppressed by pyridine6-PGLA-pre-treatment. TNF-α one of JAK/STAT activators, increased pSTAT3 levels and subsequently induced MMP-2 activation in proximal tubular cells. These results suggest that JAK/STAT3 signaling may play a role in repair process of renal fibrosis in UUO partly via MMP-2 activation.
Collapse
|
49
|
Abstract
Cytokines and growth factors play an integral role in the maintenance of immune homeostasis, the generation of protective immunity, and lung reparative processes. However, the dysregulated expression of cytokines and growth factors in response to infectious or noxious insults can initiate and perpetuate deleterious lung inflammation and fibroproliferation. In this article, we will comprehensively review the contribution of individual cytokines and growth factors and cytokine networks to key pathophysiological events in human and experimental acute lung injury (ALI), including inflammatory cell recruitment and activation, alveolar epithelial injury and repair, angiogenesis, and matrix deposition and remodeling. The application of cytokines/growth factors as prognostic indicators and therapeutic targets in human ALI is explored.
Collapse
Affiliation(s)
- Jane C Deng
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, UCLA Medical Center, Los Angeles, CA, USA
| | | |
Collapse
|
50
|
B Moore B, Lawson WE, Oury TD, Sisson TH, Raghavendran K, Hogaboam CM. Animal models of fibrotic lung disease. Am J Respir Cell Mol Biol 2013; 49:167-79. [PMID: 23526222 DOI: 10.1165/rcmb.2013-0094tr] [Citation(s) in RCA: 315] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Interstitial lung fibrosis can develop as a consequence of occupational or medical exposure, as a result of genetic defects, and after trauma or acute lung injury leading to fibroproliferative acute respiratory distress syndrome, or it can develop in an idiopathic manner. The pathogenesis of each form of lung fibrosis remains poorly understood. They each result in a progressive loss of lung function with increasing dyspnea, and most forms ultimately result in mortality. To better understand the pathogenesis of lung fibrotic disorders, multiple animal models have been developed. This review summarizes the common and emerging models of lung fibrosis to highlight their usefulness in understanding the cell-cell and soluble mediator interactions that drive fibrotic responses. Recent advances have allowed for the development of models to study targeted injuries of Type II alveolar epithelial cells, fibroblastic autonomous effects, and targeted genetic defects. Repetitive dosing in some models has more closely mimicked the pathology of human fibrotic lung disease. We also have a much better understanding of the fact that the aged lung has increased susceptibility to fibrosis. Each of the models reviewed in this report offers a powerful tool for studying some aspect of fibrotic lung disease.
Collapse
Affiliation(s)
- Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| | | | | | | | | | | |
Collapse
|