1
|
Xie K, Tan X, Chen Z, Yao Y, Luo J, Ma H, Feng Y, Jiang W. Exploring the Mechanisms and Preventive Strategies for the Progression from Idiopathic Pulmonary Fibrosis to Lung Cancer: Insights from Transcriptomics and Genetic Factors. Biomedicines 2024; 12:2382. [PMID: 39457694 PMCID: PMC11504276 DOI: 10.3390/biomedicines12102382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) leads to excessive fibrous tissue in the lungs, increasing the risk of lung cancer (LC) due to heightened fibroblast activity. Advances in nucleotide point mutation studies offer insights into fibrosis-to-cancer transitions. Methods: A two-sample Mendelian randomization (TSMR) approach was used to explore the causal relationship between IPF and LC. A weighted gene co-expression network analysis (WGCNA) identified shared gene modules related to immunogenic cell death (ICD) from transcriptomic datasets. Machine learning selected key genes, and a multi-layer perceptron (MLP) model was developed for IPF prediction and diagnosis. SMR and PheWAS were used to assess the expression of key genes concerning IPF risk. The impact of core genes on immune cells in the IPF microenvironment was explored, and in vivo experiments were conducted to examine the progression from IPF to LC. Results: The TSMR approach indicated a genetic predisposition for IPF progressing to LC. The predictive model, which includes eight ICD key genes, demonstrated a strong predictive capability (AUC = 0.839). The SMR analysis revealed that the elevated expression of MS4A4A was associated with an increased risk of IPF (OR = 1.275, 95% CI: 1.029-1.579; p = 0.026). The PheWAS did not identify any significant traits linked to MS4A4A expression. The rs9265808 locus in MS4A4A was identified as a susceptibility site for the progression of IPF to LC, with mutations potentially reprogramming lung neutrophils and increasing the LC risk. In vivo studies suggested MS4A4A as a promising therapeutic target. Conclusions: A causal link between IPF and LC was established, an effective prediction model was developed, and MS4A4A was highlighted as a therapeutic target to prevent IPF from progressing to LC.
Collapse
Affiliation(s)
- Kai Xie
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Xiaoyan Tan
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Zhe Chen
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Yu Yao
- Department of Respiratory Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China;
| | - Jing Luo
- Department of Cardiothoracic Surgery, Medical School of Nanjing University, Nanjing 210002, China;
| | - Haitao Ma
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
- Department of The First Clinical, Medical College of Soochow University, Suzhou 215006, China
| | - Yu Feng
- Department of The First Clinical, Medical College of Soochow University, Suzhou 215006, China
| | - Wei Jiang
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| |
Collapse
|
2
|
Li X, Liu Y, Tang Y, Xia Z. Transformation of macrophages into myofibroblasts in fibrosis-related diseases: emerging biological concepts and potential mechanism. Front Immunol 2024; 15:1474688. [PMID: 39386212 PMCID: PMC11461261 DOI: 10.3389/fimmu.2024.1474688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
Macrophage-myofibroblast transformation (MMT) transforms macrophages into myofibroblasts in a specific inflammation or injury microenvironment. MMT is an essential biological process in fibrosis-related diseases involving the lung, heart, kidney, liver, skeletal muscle, and other organs and tissues. This process consists of interacting with various cells and molecules and activating different signal transduction pathways. This review deeply discussed the molecular mechanism of MMT, clarified crucial signal pathways, multiple cytokines, and growth factors, and formed a complex regulatory network. Significantly, the critical role of transforming growth factor-β (TGF-β) and its downstream signaling pathways in this process were clarified. Furthermore, we discussed the significance of MMT in physiological and pathological conditions, such as pulmonary fibrosis and cardiac fibrosis. This review provides a new perspective for understanding the interaction between macrophages and myofibroblasts and new strategies and targets for the prevention and treatment of MMT in fibrotic diseases.
Collapse
Affiliation(s)
- Xiujun Li
- Health Science Center, Chifeng University, Chifeng, China
| | - Yuyan Liu
- Rehabilitation Medicine College, Shandong Second Medical University, Jinan, China
| | - Yongjun Tang
- Department of Emergency, Affiliated Hospital of Chifeng University, Chifeng, China
| | - Zhaoyi Xia
- Department of Library, Children’s Hospital Affiliated to Shandong University, Jinan, China
- Department of Library, Jinan Children’s Hospital, Jinan, China
| |
Collapse
|
3
|
Moloudi K, Azariasl S, Abrahamse H, George BP, Yasuda H. Expected role of photodynamic therapy to relieve skin damage in nuclear or radiological emergency: Review. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 110:104517. [PMID: 39032581 DOI: 10.1016/j.etap.2024.104517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/02/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Nuclear and radiological accidents can occur due to poor management, in transportation, radiation therapy and nuclear wards in hospitals, leading to extreme radiation exposure and serious consequences for human health. Additionally, in many of previous radiological accidents, skin damage was observed in patients and survivors due to the high radiation exposure. However, as part of a medical countermeasures in a nuclear/radiological emergency, it is critical to plan for the treatment of radiation-induced skin damage. Hence, the new, non-invasive technology of photodynamic therapy (PDT) is projected to be more effectively used for treating skin damage caused by high-dose radiation. PDT plays an important role in treating, repairing skin damage and promoting wound healing as evidenced by research. This review, highlighted and recommended potential impacts of PDT to repair and decrease radiation-induced skin tissue damage. Moreover, we have suggested some photosensitizer (PS) agent as radio-mitigator drugs to decrease radiobiological effects.
Collapse
Affiliation(s)
- Kave Moloudi
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa
| | - Samayeh Azariasl
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Minami-ku 734-8553, Japan
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Science, Doornfontein Campus, University of Johannesburg, Johannesburg 2028, South Africa.
| | - Hiroshi Yasuda
- Department of Radiation Biophysics, Research Institute for Radiation Biology and Medicine, Hiroshima University, Kasumi, Minami-ku 734-8553, Japan
| |
Collapse
|
4
|
Russo RC, Quesniaux VFJ, Ryffel B. Homeostatic chemokines as putative therapeutic targets in idiopathic pulmonary fibrosis. Trends Immunol 2023; 44:1014-1030. [PMID: 37951789 DOI: 10.1016/j.it.2023.10.003] [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: 09/22/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal chronic interstitial lung disease (ILD) that affects lung mechanical functions and gas exchange. IPF is caused by increased fibroblast activity and collagen deposition that compromise the alveolar-capillary barrier. Identifying an effective therapy for IPF remains a clinical challenge. Chemokines are key proteins in cell communication that have functions in immunity as well as in tissue homeostasis, damage, and repair. Chemokine receptor signaling induces the activation and proliferation of lung-resident cells, including alveolar macrophages (AMs) and fibroblasts. AMs are an important source of chemokines and cytokines during IPF. We highlight the complexity of this system and, based on insights from genetic and transcriptomic studies, propose a new role for homeostatic chemokine imbalance in IPF, with implications for putative therapeutic targets.
Collapse
Affiliation(s)
- Remo C Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Valerie F J Quesniaux
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics (INEM), Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7355, University of Orleans, Orleans 45071, France.
| |
Collapse
|
5
|
Zhan JH, Wei J, Liu L, Xu YT, Ji H, Wang CN, Liu YJ, Zhu XY. Investigation of a UPR-Related Gene Signature Identifies the Pro-Fibrotic Effects of Thrombospondin-1 by Activating CD47/ROS/Endoplasmic Reticulum Stress Pathway in Lung Fibroblasts. Antioxidants (Basel) 2023; 12:2024. [PMID: 38136144 PMCID: PMC10740656 DOI: 10.3390/antiox12122024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Unfolded protein response (UPR) signaling and endoplasmic reticulum (ER) stress have been linked to pulmonary fibrosis. However, the relationship between UPR status and pulmonary function and prognosis in idiopathic pulmonary fibrosis (IPF) patients remains largely unknown. Through a series of bioinformatics analyses, we established a correlation between UPR status and pulmonary function in IPF patients. Furthermore, thrombospondin-1 (TSP-1) was identified as a potential biomarker for prognostic evaluation in IPF patients. By utilizing both bulk RNA profiling and single-cell RNA sequencing data, we demonstrated the upregulation of TSP-1 in lung fibroblasts during pulmonary fibrosis. Gene set enrichment analysis (GSEA) results indicated a positive association between TSP-1 expression and gene sets related to the reactive oxygen species (ROS) pathway in lung fibroblasts. TSP-1 overexpression alone induced mild ER stress and pulmonary fibrosis, and it even exacerbated bleomycin-induced ER stress and pulmonary fibrosis. Mechanistically, TSP-1 promoted ER stress and fibroblast activation through CD47-dependent ROS production. Treatment with either TSP-1 inhibitor or CD47 inhibitor significantly attenuated BLM-induced ER stress and pulmonary fibrosis. Collectively, these findings suggest that the elevation of TSP-1 during pulmonary fibrosis is not merely a biomarker but likely plays a pathogenic role in the fibrotic changes in the lung.
Collapse
Affiliation(s)
- Jun-Hui Zhan
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Juan Wei
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Lin Liu
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China;
| | - Yi-Tong Xu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Hui Ji
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Chang-Nan Wang
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; (J.-H.Z.); (J.W.); (Y.-T.X.); (H.J.)
| | - Xiao-Yan Zhu
- Department of Physiology, Navy Medical University, Shanghai 200433, China;
| |
Collapse
|
6
|
Zhang X, Shi X, Xie F, Liu Y, Wei X, Cai Y, Chao J. Dissecting pulmonary fibroblasts heterogeneity in lung development, health and diseases. Heliyon 2023; 9:e19428. [PMID: 37674845 PMCID: PMC10477496 DOI: 10.1016/j.heliyon.2023.e19428] [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: 02/17/2023] [Revised: 08/11/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
Lung fibroblasts are the major components in the connective tissue of the pulmonary interstitium and play essential roles in the developing of postnatal lung, synthesizing the extracellular matrix and maintaining the integrity of the lung architecture. Fibroblasts are activated in various disease conditions and exhibit functional heterogeneities according to their origin, spatial location, activated state and microenvironment. In recent years, advances in technology have enabled researchers to identify fibroblast subpopulations in both mouse and human. Here, we discuss pulmonary fibroblast heterogeneity, focusing on the developing, healthy and pathological lung conditions. We firstly review the expression profiles of fibroblasts during lung development, and then consider fibroblast diversity according to different anatomical sites of lung architecture. Subsequently, we discuss fibroblast heterogeneity in genetic lineage. Finally, we focus on how fibroblast heterogeneity may shed light on different pathological lung conditions such as fibrotic diseases, infectious diseases including COVID-19, and lung cancers. We emphasize the importance of comparative studies to illuminate the overlapping characteristics, expression profiles and signaling pathways of the fibroblast subpopulations across disease conditions, a better characterization of the functional complexity rather than the expression of a particular gene may have important therapeutic applications.
Collapse
Affiliation(s)
- Xinxin Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
- Department of Histology and Embryology, School of Medicine, Southeast University, Nanjing 210009, PR China
| | - Xiaoni Shi
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Feiyan Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yaping Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xinyan Wei
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Yu Cai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
7
|
Werner G, Sanyal A, Mirizio E, Hutchins T, Tabib T, Lafyatis R, Jacobe H, Torok KS. Single-Cell Transcriptome Analysis Identifies Subclusters with Inflammatory Fibroblast Responses in Localized Scleroderma. Int J Mol Sci 2023; 24:9796. [PMID: 37372943 DOI: 10.3390/ijms24129796] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/19/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Localized scleroderma (LS) is an autoimmune disease with both inflammatory and fibrotic components causing an abnormal deposition of collagen in the skin and underlying tissue, often leading to disfigurement and disability. Much of its pathophysiology is extrapolated from systemic sclerosis (SSc) since the histopathology findings in the skin are nearly identical. However, LS is critically understudied. Single-cell RNA sequencing (scRNA seq) technology provides a novel way to obtain detailed information at the individual cellular level, overcoming this barrier. Here, we analyzed the affected skin of 14 patients with LS (pediatric and adult) and 14 healthy controls. Fibroblast populations were the focus, since they are the main drivers of fibrosis in SSc. We identified 12 fibroblast subclusters in LS, which overall had an inflammatory gene expression (IFN and HLA-associated genes). A myofibroblast-like cluster (SFRP4/PRSS23) was more prevalent in LS subjects and shared many upregulated genes expressed in SSc-associated myofibroblasts, though it also had strong expression of CXCL9/10/11, known CXCR3 ligands. A CXCL2/IRF1 cluster identified was unique to LS, with a robust inflammatory gene signature, including IL-6, and according to cell communication analysis are influenced by macrophages. In summary, potential disease-propagating fibroblasts and associated gene signatures were identified in LS skin via scRNA seq.
Collapse
Affiliation(s)
- Giffin Werner
- Department of Pediatrics (Rheumatology), University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Anwesha Sanyal
- Department of Pediatrics (Rheumatology), University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Emily Mirizio
- Department of Pediatrics (Rheumatology), University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Theresa Hutchins
- Department of Pediatrics (Rheumatology), University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Heidi Jacobe
- Department of Dermatology, University of Texas Southwestern, Dallas, TX 75390, USA
| | - Kathryn S Torok
- Department of Pediatrics (Rheumatology), University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
8
|
Grillo E, Ravelli C, Colleluori G, D'Agostino F, Domenichini M, Giordano A, Mitola S. Role of gremlin-1 in the pathophysiology of the adipose tissues. Cytokine Growth Factor Rev 2023; 69:51-60. [PMID: 36155165 DOI: 10.1016/j.cytogfr.2022.09.004] [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: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 02/07/2023]
Abstract
Gremlin-1 is a secreted bone morphogenetic protein (BMP) antagonist playing a pivotal role in the regulation of tissue formation and embryonic development. Since its first identification in 1997, gremlin-1 has been shown to be a multifunctional factor involved in wound healing, inflammation, cancer and tissue fibrosis. Among others, the activity of gremlin-1 is mediated by its interaction with BMPs or with membrane receptors such as the vascular endothelial growth factor receptor 2 (VEGFR2) or heparan sulfate proteoglycans (HSPGs). Growing evidence has highlighted a central role of gremlin-1 in the homeostasis of the adipose tissue (AT). Of note, gremlin-1 is involved in AT dysfunction during type 2 diabetes, obesity and non-alcoholic fatty liver disease (NAFLD) metabolic disorders. In this review we discuss recent findings on gremlin-1 involvement in AT biology, with particular attention to its role in metabolic diseases, to highlight its potential as a prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Georgia Colleluori
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Francesco D'Agostino
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Mattia Domenichini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020 Ancona, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
Filidou E, Kandilogiannakis L, Tarapatzi G, Spathakis M, Steiropoulos P, Mikroulis D, Arvanitidis K, Paspaliaris V, Kolios G. Anti-Inflammatory and Anti-Fibrotic Effect of Immortalized Mesenchymal-Stem-Cell-Derived Conditioned Medium on Human Lung Myofibroblasts and Epithelial Cells. Int J Mol Sci 2022; 23:ijms23094570. [PMID: 35562961 PMCID: PMC9102072 DOI: 10.3390/ijms23094570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is caused by progressive lung tissue impairment due to extended chronic fibrosis, and it has no known effective treatment. The use of conditioned media (CM) from an immortalized human adipose mesenchymal stem cell line could be a promising therapeutic strategy, as it can reduce both fibrotic and inflammatory responses. We aimed to investigate the anti-inflammatory and anti-fibrotic effect of CM on human pulmonary subepithelial myofibroblasts (hPSM) and on A549 pulmonary epithelial cells, treated with pro-inflammatory or pro-fibrotic mediators. CM inhibited the proinflammatory cytokine-induced mRNA and protein production of various chemokines in both hPSMs and A549 cells. It also downregulated the mRNA expression of IL-1α, but upregulated IL-1β and IL-6 mRNA production in both cell types. CM downregulated the pro-fibrotic-induced mRNA expression of collagen Type III and the migration rate of hPSMs, but upregulated fibronectin mRNA production and the total protein collagen secretion. CM's direct effect on the chemotaxis and cell recruitment of immune-associated cells, and its indirect effect on fibrosis through the significant decrease in the migration capacity of hPSMs, makes it a plausible candidate for further development towards a therapeutic treatment for IPF.
Collapse
Affiliation(s)
- Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| | - Leonidas Kandilogiannakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| | - Michail Spathakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| | - Paschalis Steiropoulos
- Department of Pneumonology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Dimitrios Mikroulis
- Department of Cardiac Surgery, Democritus University of Thrace, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece;
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| | - Vasilis Paspaliaris
- Vasilis Paspaliaris, Tithon Biotech Inc., 11440 West Bernardo Court, Suite 300, San Diego, CA 92127, USA
- Correspondence: ; Tel./Fax: +1-88-8780-2639
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (E.F.); (L.K.); (G.T.); (M.S.); (K.A.); (G.K.)
| |
Collapse
|
10
|
Zou M, Zou J, Hu X, Zheng W, Zhang M, Cheng Z. Latent Transforming Growth Factor-β Binding Protein-2 Regulates Lung Fibroblast-to-Myofibroblast Differentiation in Pulmonary Fibrosis via NF-κB Signaling. Front Pharmacol 2022; 12:788714. [PMID: 35002722 PMCID: PMC8740300 DOI: 10.3389/fphar.2021.788714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Despite past extensive studies, the mechanisms underlying pulmonary fibrosis (PF) still remain poorly understood. The aberrantly activated lung myofibroblasts, predominantly emerging through fibroblast-to-myofibroblast differentiation, are considered to be the key cells in PF, resulting in excessive accumulation of extracellular matrix (ECM). Latent transforming growth factor-β (TGFβ) binding protein-2 (LTBP2) has been suggested as playing a critical role in modulating the structural integrity of the ECM. However, its function in PF remains unclear. Here, we demonstrated that lungs originating from different types of patients with PF, including idiopathic PF and rheumatoid arthritis-associated interstitial lung disease, and from mice following bleomycin (BLM)-induced PF were characterized by increased LTBP2 expression in activated lung fibroblasts/myofibroblasts. Moreover, serum LTBP2 was also elevated in patients with COVID-19-related PF. LTBP2 silencing by lentiviral shRNA transfection protected against BLM-induced PF and suppressed fibroblast-to-myofibroblast differentiation in vivo and in vitro. More importantly, LTBP2 overexpression was able to induce differentiation of lung fibroblasts to myofibroblasts in vitro, even in the absence of TGFβ1. By further mechanistic analysis, we demonstrated that LTBP2 silencing prevented fibroblast-to-myofibroblast differentiation and subsequent PF by suppressing the phosphorylation and nuclear translocation of NF-κB signaling. LTBP2 overexpression-induced fibroblast-to-myofibroblast differentiation depended on the activation of NF-κB signaling in vitro. Therefore, our data indicate that intervention to silence LTBP2 may represent a promising therapy for PF.
Collapse
Affiliation(s)
- Menglin Zou
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingfeng Zou
- Department of Respiratory and Critical Care Medicine, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Xingxing Hu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weishuai Zheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mingyang Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenshun Cheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Qiu L, Gong G, Wu W, Li N, Li Z, Chen S, Li P, Chen T, Zhao H, Hu C, Fang Z, Wang Y, Liu H, Cui P, Zhang G. A novel prognostic signature for idiopathic pulmonary fibrosis based on five-immune-related genes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1570. [PMID: 34790776 PMCID: PMC8576669 DOI: 10.21037/atm-21-4545] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/02/2021] [Indexed: 01/04/2023]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a highly fatal lung disease of unknown etiology with a median survival after diagnosis of only 2–3 years. Its poor prognosis is due to the limited therapy options available as well as the lack of effective prognostic indicators. This study aimed to construct a novel prognostic signature for IPF to assist in the personalized management of IPF patients during treatment. Methods Differentially-expressed genes (DEGs) in IPF patients versus healthy individuals were analyzed using the “limma” package of R software. Immune-related genes (IRGs) were obtained from the ImmPort database. Univariate Cox regression analysis was adopted to screen significantly prognostic IRGs for IPF patients. Multiple Cox regression analysis was used to identify optimal prognostic IRGs and construct a prognostic signature. Results Compared with healthy individuals, there were a total of 52 prognosis-related DEGs in the bronchoalveolar lavage (BAL) samples of IPF patients, of which 37 genes were identified as IRGs. Of these, five genes (CXCL14, SLC40A1, RNASE3, CCR3, and RORA) were significantly associated with overall survival (OS) in IPF patients, and were utilized for establishment of the prognostic signature. IPF patients were divided into high- and low-risk groups based on the prognostic signature. Marked differences in the OS probability were observed between high- and low-risk IPF patients. The area under curves (AUCs) of the receiver operating characteristic (ROC) curve for the prognostic signature in the training and validation cohorts were 0.858 and 0.837, respectively. The expression levels between RNASE3 and SLC40A1 (P<0.01, r=0.394), between RORA and CXCL14 (P<0.01, r=−0.355), between CCR3 and CXCL14 (P<0.01, r=0.258), as well as between RNASE3 and CCR3 (P<0.01, r=0.293) were significantly correlated. Conclusions We developed a validated and reproducible IRG-based prognostic signature that should be helpful in the personalized management of patients with IPF, providing new insights into the relationship between the immune system and IPF.
Collapse
Affiliation(s)
- Lingxiao Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China
| | - Gencheng Gong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjuan Wu
- Department of Geriatric Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Nana Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaonan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China
| | - Ping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China.,Zhengzhou Key Laboratory for Chronic Respiratory Disease, Zhengzhou, China
| | - Tengfei Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huasi Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunling Hu
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zeming Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongping Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Panpan Cui
- School of Nursing and Heath, Zhengzhou University, Zhengzhou, China
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China.,Zhengzhou Key Laboratory for Chronic Respiratory Disease, Zhengzhou, China
| |
Collapse
|
12
|
Yu P, Qian T, Gong Q, Fu M, Bian X, Sun T, Zhang Z, Xu X. Inflammatory cytokines levels in aqueous humour and surgical outcomes of trabeculectomy in patients with prior acute primary angle closure. Acta Ophthalmol 2021; 99:e1106-e1111. [PMID: 33438359 DOI: 10.1111/aos.14763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/02/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE To quantify the levels of three inflammatory cytokines in the aqueous humour of patients with prior acute primary angle closure (APAC) and investigate their correlation with surgical outcomes of trabeculectomy. METHODS In this prospective cohort study, aqueous humour samples were collected from 44 prior APAC eyes. Analyte concentrations of monocyte chemoattractant protein-1 (MCP-1), vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6) were measured using multiplexed immunoassay kits. Intraocular pressure was measured using Goldmann application tonometry. RESULTS Forty-four prior APAC eyes were followed up for 24 months after trabeculectomy and divided into success and failure groups according to surgical outcomes. Monocyte chemoattractant protein-1 (MCP-1) levels in the aqueous humour were significantly higher in the failure group (p = 0.0118). Univariate and multivariate analyses showed that MCP-1 level was a significant risk factor for trabeculectomy outcomes (univariate analysis: p = 0.016, odds ratio = 14.538; multivariate analysis: p = 0.023, odds ratio = 13.718). When prior APAC eyes were divided according to MCP-1 levels, the overall success rate was significantly higher in eyes with low MCP-1 levels than eyes with high MCP-1 levels (p = 0.0249). CONCLUSION In prior APAC patients, the MCP-1 level in the aqueous humour predicts trabeculectomy results. Therefore, modulation of MCP-1 expression may have potential clinical applications after filtration surgery.
Collapse
Affiliation(s)
- Ping Yu
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Tianwei Qian
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Qiaoyun Gong
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Mingshui Fu
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Xiaolan Bian
- Department of Pharmacy Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Tao Sun
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Zhihua Zhang
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| | - Xun Xu
- Department of Ophthalmology Shanghai General Hospital Shanghai Jiao Tong University Shanghai China
- National Clinical Research Center for Eye Diseases Shanghai China
- Shanghai Key Laboratory of Ocular Fundus Diseases Shanghai China
- Shanghai Engineering Center for Visual Science and Photomedicine Shanghai China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease Shanghai China
| |
Collapse
|
13
|
Reyes-Jiménez E, Ramírez-Hernández AA, Santos-Álvarez JC, Velázquez-Enríquez JM, Pina-Canseco S, Baltiérrez-Hoyos R, Vásquez-Garzón VR. Involvement of 4-hydroxy-2-nonenal in the pathogenesis of pulmonary fibrosis. Mol Cell Biochem 2021; 476:4405-4419. [PMID: 34463938 DOI: 10.1007/s11010-021-04244-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/12/2021] [Indexed: 01/19/2023]
Abstract
Pulmonary fibrosis is a chronic progressive disease with high incidence, prevalence, and mortality rates worldwide. It is characterized by excessive accumulation of extracellular matrix in the lung parenchyma. The cellular and molecular mechanisms involved in its pathogenesis are complex, and some are still unknown. Several studies indicate that oxidative stress, characterized by overproduction of 4-hydroxy-2-nonenal (4-HNE), is an important player in pulmonary fibrosis. 4-HNE is a highly reactive compound derived from polyunsaturated fatty acids that can react with proteins, phospholipids, and nucleic acids. Thus, many of the altered cellular mechanisms that contribute to this disease can be explained by the participation of 4-HNE. Here, we summarize the current knowledge on the molecular states and signal transduction pathways that contribute to the pathogenesis of pulmonary fibrosis. Furthermore, we describe the participation of 4-HNE in various mechanisms involved in pulmonary fibrosis development, with a focus on the cell populations involved in the initiation, development, and maintenance of the fibrotic process, mainly alveolar cells, endothelial cells, macrophages, and inflammatory cells. Due to its characteristic activity as a second messenger, 4-HNE, in addition to being a consequence of oxidative stress, can support maintenance of the inflammatory and fibrotic process by spreading the effects of reactive oxygen species (ROS). Thus, regulation of 4-HNE levels could be a viable strategy to reduce its effects on the mechanisms involved in pulmonary fibrosis development.
Collapse
Affiliation(s)
- Edilburga Reyes-Jiménez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | | | - Rafael Baltiérrez-Hoyos
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, Mexico
| | | |
Collapse
|
14
|
Hanmandlu A, Zhu L, Mertens TC, Collum S, Bi W, Xiong F, Wang R, Amirthalingam RT, Ren D, Han L, Jyothula SS, Li W, Zheng WJ, Karmouty-Quintana H. Transcriptomic and Epigenetic Profiling of Fibroblasts in Idiopathic Pulmonary Fibrosis (IPF). Am J Respir Cell Mol Biol 2021; 66:53-63. [PMID: 34370624 DOI: 10.1165/rcmb.2020-0437oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), a devastating, fibro-proliferative chronic lung disorder, is associated with expansion of fibroblasts/myofibroblasts, which leads to excessive production and deposition of extracellular matrix (ECM). IPF is typically clinically identified as end-stage lung disease, after fibrotic processes are well-established and advanced. Fibroblasts have been shown to be critically important in the development and progression of IPF. We hypothesize that differential chromatin access can drive genetic differences in IPF fibroblasts relative to healthy fibroblasts. To this end, we performed Assay of Transposase-Accessible Chromatin (ATAC)-sequencing to identify differentially accessible regions within the genomes of fibroblasts from healthy and IPF lungs. Multiple motifs were identified to be enriched in IPF fibroblasts compared to healthy fibroblasts, including binding motifs for TWIST1 and FOXA1. RNA-sequencing identified 93 genes that could be annotated to differentially accessible regions. Pathway analysis of the annotated genes identified cellular adhesion, cytoskeletal anchoring, and cell differentiation as important biological processes. In addition, single nucleotide polymorphisms (SNPs) analysis showed that linkage disequilibrium (LD) blocks of IPF risk SNPs with IPF accessible regions that have been identified to be located in genes which are important in IPF, including MUC5B, TERT and TOLLIP. Validation studies in isolated lung tissue confirmed increased expression for TWIST1 and FOXA1 in addition to revealing SHANK2 and CSPR2 as novel targets. Thus, modulation of differential chromatin access may be an important mechanism in the pathogenesis of lung fibrosis.
Collapse
Affiliation(s)
- Ankit Hanmandlu
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Lisha Zhu
- University of Texas Health Science Center at Houston, 12340, School of Biomedical Informatics, Houston, Texas, United States
| | - Tinne Cj Mertens
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Scott Collum
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Weizhen Bi
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Feng Xiong
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Ruoyu Wang
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | | | - Dewei Ren
- Houston Methodist Hospital, 23534, J.C. Walter Jr. Transplant Center, Houston, Texas, United States
| | - Leng Han
- The University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - Soma Sk Jyothula
- University of Texas Health Science Center at Houston, 12340, Internal Medicine, Houston, Texas, United States
| | - Wenbo Li
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States
| | - W Jim Zheng
- The University of Texas Health Science Center at Houston, 12340, School of Biomedical Informatics, Houston, Texas, United States
| | - Harry Karmouty-Quintana
- University of Texas Health Science Center at Houston, 12340, Biochemistry and Molecular Biology, Houston, Texas, United States;
| |
Collapse
|
15
|
Kreuter M, Lee JS, Tzouvelekis A, Oldham JM, Molyneaux PL, Weycker D, Atwood M, Kirchgaessler KU, Maher TM. Monocyte Count as a Prognostic Biomarker in Patients with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2021; 204:74-81. [PMID: 33434107 PMCID: PMC8437112 DOI: 10.1164/rccm.202003-0669oc] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: There is an urgent need for simple, cost-effective prognostic biomarkers for idiopathic pulmonary fibrosis (IPF); biomarkers that show potential include monocyte count. Objectives: We used pooled data from pirfenidone and IFNγ-1b trials to explore the association between monocyte count and prognosis in patients with IPF. Methods: This retrospective pooled analysis included patients (active and placebo arms) from the following four phase III, randomized, placebo-controlled trials: ASCEND (NCT01366209), CAPACITY (NCT00287729 and NCT00287716), and INSPIRE (NCT00075998). Outcomes included IPF progression (≥10% absolute decline in FVC% predicted, ≥50 m decline in 6-minute-walk distance, or death), all-cause hospitalization, and all-cause mortality over 1 year. The relationship between monocyte count (defined as time-dependent) and outcomes was assessed using bivariate and multivariable models. Measurements and Main Results: This analysis included 2,067 patients stratified by monocyte count (at baseline: <0.60 × 109 cells/L [n = 1,609], 0.60 to <0.95 × 109 cells/L [n = 408], and ≥0.95 × 109 cells/L [n = 50]). In adjusted analyses, a higher proportion of patients with monocyte counts of 0.60 to <0.95 × 109 cells/L or ≥0.95 × 109 cells/L versus <0.60 × 109 cells/L experienced IPF progression (P = 0.016 and P = 0.002, respectively), all-cause hospitalization (P = 0.030 and P = 0.003, respectively), and all-cause mortality (P = 0.005 and P < 0.001, respectively) over 1 year. Change in monocyte count from baseline was not associated with any of the outcomes over 1 year and did not appear to be affected by study treatment. Conclusions: In patients with IPF, elevated monocyte count was associated with increased risks of IPF progression, hospitalization, and mortality. Monocyte count may provide a simple and inexpensive prognostic biomarker in IPF.
Collapse
Affiliation(s)
- Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik, University of Heidelberg, Heidelberg, Germany
- German Center for Lung Research, Heidelberg, Germany
| | - Joyce S Lee
- Department of Medicine, University of Colorado, Denver, Colorado
| | | | - Justin M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of California Davis, Sacramento, California
| | - Philip L Molyneaux
- Interstitial Lung Disease Unit, Royal Brompton Hospital, London, United Kingdom
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Mark Atwood
- Policy Analysis, Inc., Brookline, Massachusetts
| | | | - Toby M Maher
- Interstitial Lung Disease Unit, Royal Brompton Hospital, London, United Kingdom
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care, and Sleep Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
16
|
Duffy L, Henderson J, Brown M, Pryzborski S, Fullard N, Summa L, Distler JHW, Stratton R, O'Reilly S. Bone Morphogenetic Protein Antagonist Gremlin-1 Increases Myofibroblast Transition in Dermal Fibroblasts: Implications for Systemic Sclerosis. Front Cell Dev Biol 2021; 9:681061. [PMID: 34150776 PMCID: PMC8213337 DOI: 10.3389/fcell.2021.681061] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/13/2021] [Indexed: 11/18/2022] Open
Abstract
Objective Systemic Sclerosis is an autoimmune connective tissue disease which results in fibrosis of the skin and lungs. The disease is characterized by activation of myofibroblasts but what governs this is unknown. Gremlin-1 is a BMP antagonist that is developmentally regulated and we sought to investigate its role in Systemic Sclerosis. Methods Dermal fibroblasts were transfected with Grem1pcDNA3.1 expression vectors or empty vectors. Various markers of myofibroblasts were measured at the mRNA and protein levels. Scratch wound assays were also performed. Media Transfer experiments were performed to evaluate cytokine like effects. Various inhibitors of TGF-β signaling and MAPK signaling were used post-transfection. siRNA to Gremlin-1 in SSc dermal fibroblasts were performed to evaluate the role of Gremlin-1. Different cytokines were incubated with fibroblasts and Gremlin-1 measured. Bleomycin was used as model of fibrosis and immunohistochemistry performed. Results Overexpression of Gremlin-1 was achieved in primary dermal fibroblasts and lead to activation of quiescent cells to myofibroblasts indicated by collagen and α-Smooth muscle actin. Overexpression also led to functional effects. This was associated with increased TGF-β1 levels and SBE luciferase activity but not increased Thrombospondin-1 expression. Inhibition of Gremlin-1 overexpression cells with antibodies to TGF-β1 but not isotype controls led to reduced collagen and various TGF-β pathway chemical inhibitors also led to reduced collagen levels. In SSc cells siRNA mediated reduction of Gremlin-1 reduced collagen expression and CTGF gene and protein levels in these cells. IL-13 did not lead to elevated Gremlin-1 expression nor did IL-11. Gremlin-1 was elevated in an animal model of fibrosis compared to NaCl-treated mice. Conclusion Gremlin-1 is a key regulator of myofibroblast transition leading to enhanced ECM deposition. Strategies that block Gremlin-1 maybe a possible therapeutic target in fibrotic diseases such as SSc.
Collapse
Affiliation(s)
- Laura Duffy
- Faculty of Health and Life Science, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - John Henderson
- Faculty of Health and Life Science, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Max Brown
- Biosciences Department, Durham University, Durham, United Kingdom
| | | | - Nicola Fullard
- Biosciences Department, Durham University, Durham, United Kingdom
| | - Lena Summa
- Department of Internal Medicine 3 Friedrich-Alexander-University, Erlangen-Nurnberg, Germany
| | - Jorg H W Distler
- Department of Internal Medicine 3 Friedrich-Alexander-University, Erlangen-Nurnberg, Germany
| | - Richard Stratton
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Steven O'Reilly
- Biosciences Department, Durham University, Durham, United Kingdom
| |
Collapse
|
17
|
Wilson SE. TGF beta -1, -2 and -3 in the modulation of fibrosis in the cornea and other organs. Exp Eye Res 2021; 207:108594. [PMID: 33894227 DOI: 10.1016/j.exer.2021.108594] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/10/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
The TGF beta-1, -2 and -3 isoforms are transcribed from different genes but bind to the same receptors and signal through the same canonical and non-canonical signal transduction pathways. There are numerous regulatory mechanisms controlling the action of each isoform that include the organ-specific cells producing latent TGF beta growth factors, multiple effectors that activate the isoforms, ECM-associated SLRPs and basement membrane components that modulate the activity and localization of the isoforms, other interactive cytokine-growth factor receptor systems, such as PDGF and CTGF, TGF beta receptor expression on target cells, including myofibroblast precursors, receptor binding competition, positive and negative signal transduction effectors, and transcription and translational regulatory mechanisms. While there has long been the view that TGF beta-1and TGF beta-2 are pro-fibrotic, while TGF beta-3 is anti-fibrotic, this review suggests that view is too simplistic, at least in adult tissues, since TGF beta-3 shares far more similarities in its modulation of fibrotic gene expression with TGF beta-1 and TGF beta-2, than it does differences, and often the differences are subtle. Rather, TGF beta-3 should be seen as a fibro-modulatory partner to the other two isoforms that modulates a nuanced and better controlled response to injury. The complex interplay between the three isoforms and numerous interactive proteins, in the context of the cellular milieu, controls regenerative non-fibrotic vs. fibrotic healing in a response to injury in a particular organ, as well as the resolution of fibrosis, when that occurs.
Collapse
Affiliation(s)
- Steven E Wilson
- The Cole Eye Institute, The Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
18
|
Wilson SE, Sampaio LP, Shiju TM, Carlos de Oliveira R. Fibroblastic and bone marrow-derived cellularity in the corneal stroma. Exp Eye Res 2020; 202:108303. [PMID: 33068626 DOI: 10.1016/j.exer.2020.108303] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 09/18/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022]
Abstract
The unwounded, normal corneal stroma is a relatively simple, avascular tissue populated with quiescent keratocytes, along with corneal nerves and a few resident dendritic and monocyte/macrophage cells. In the past, the resting keratocytes were thought of as a homogenous cellular population, but recent work has shown local variations in vimentin and nestin expression, and responsiveness to transforming growth factor (TGF)-β1. Studies have also supported there being "stromal stem cells" in localized areas. After corneal wounding, depending on the site and severity of injury, profound changes in stromal cellularity occur. Anterior or posterior injuries to the epithelium or endothelium, respectively, trigger apoptosis of adjacent keratocytes. Many contiguous keratocytes transition to keratocan-negative corneal fibroblasts that are proliferative and produce limited amounts of disorganized extracellular matrix components. Simultaneously, large numbers of bone marrow-derived cells, including monocytes, neutrophils, fibrocytes and lymphocytes, invade the stroma from the limbal blood vessels. Ongoing adequate levels of TGFβ1, TGFβ2 and platelet-derived growth factor (PDGF) from epithelium, tears, endothelium and aqueous humor that penetrate defective or absent epithelial barrier function (EBF) and epithelial basement membrane (EBM) and/or Descemet's basement membrane (DBM) drive corneal fibroblasts and fibrocytes to differentiate into alpha-smooth muscle actin (SMA)-positive myofibroblasts. If the EBF, EBM and/or DBM are repaired or replaced in a timely manner, typically measured in weeks, then corneal fibroblast and fibrocyte progeny, deprived of requisite levels of TGFβ1 and TGFβ2, undergo apoptosis or revert to their precursor cell-types. If the EBF, EBM and/or DBM are not repaired or replaced, stromal levels of TGFβ1 and TGFβ2 remain elevated, and mature myofibroblasts are generated from corneal fibroblasts and fibrocyte precursors that produce prodigious amounts of disordered extracellular matrix materials associated with scarring fibrosis. This fibrotic stromal matrix persists, at least until the EBF, EBM and/or DBM are regenerated or replaced, and keratocytes remove and reorganize the affected stromal matrix.
Collapse
Affiliation(s)
- Steven E Wilson
- Cole Eye Institute, I-32, Cleveland Clinic, 9500, Euclid Ave, Cleveland, OH, United States.
| | - Lycia Pedral Sampaio
- Cole Eye Institute, I-32, Cleveland Clinic, 9500, Euclid Ave, Cleveland, OH, United States
| | - Thomas Michael Shiju
- Cole Eye Institute, I-32, Cleveland Clinic, 9500, Euclid Ave, Cleveland, OH, United States
| | | |
Collapse
|
19
|
Worrell JC, Walsh SM, Fabre A, Kane R, Hinz B, Keane MP. CXCR3A promotes the secretion of the antifibrotic decoy receptor sIL-13Rα2 by pulmonary fibroblasts. Am J Physiol Cell Physiol 2020; 319:C1059-C1069. [PMID: 33026833 DOI: 10.1152/ajpcell.00076.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CXC chemokine receptor 3 (CXCR3) A and its IFN-inducible ligands CXCL9 and CXCL10 regulate vascular remodeling and fibroblast motility. IL-13 is a profibrotic cytokine implicated in the pathogenesis of inflammatory and fibroproliferative conditions. Previous work from our laboratory has shown that CXCR3A is negatively regulated by IL-13 and is necessary for the basal regulation of the IL-13 receptor subunit IL-13Rα2. This study investigates the regulation of fibroblast phenotype, function, and downstream IL-13 signaling by CXCR3A in vitro. CXCR3A was overexpressed via transient transfection. CXCR3A-/- lung fibroblasts were isolated for functional analysis. Additionally, the contribution of CXCR3A to tissue remodeling following acute lung injury was assessed in vivo with wild-type (WT) and CXCR3-/- mice challenged with IL-13. CXCR3 and IL-13Rα2 displayed a reciprocal relationship after stimulation with either IL-13 or CXCR3 ligands. CXCR3A reduced expression of fibroblast activation makers, soluble collagen production, and proliferation. CXCR3A enhanced the basal expression of pERK1/2 while inducing IL-13-mediated downregulation of NF-κB-p65. CXCR3A-/- pulmonary fibroblasts were increasingly proliferative and displayed reduced contractility and α-smooth muscle actin expression. IL-13 challenge regulated expression of the CXCR3 ligands and soluble IL-13Rα2 levels in lungs and bronchoalveolar lavage fluid (BALF) of WT mice; this response was absent in CXCR3-/- mice. Alveolar macrophage accumulation and expression of genes involved in lung remodeling was increased in CXCR3-/- mice. We conclude that CXCR3A is a central antifibrotic factor in pulmonary fibroblasts, limiting fibroblast activation and reducing extracellular matrix (ECM) production. Therefore, targeting of CXCR3A may be a novel approach to regulating fibroblast activity in lung fibrosis and remodeling.
Collapse
Affiliation(s)
- Julie C Worrell
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Sinead M Walsh
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Aurélie Fabre
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,UCD Conway Research Pathology Core Technology, University College Dublin, Dublin, Ireland
| | - Rosemary Kane
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada
| | - Michael P Keane
- St. Vincent's University Hospital and School of Medicine, University College Dublin and UCD Conway Institute of Biomolecular and Biomedical Research, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
20
|
Wilson SE. Corneal myofibroblasts and fibrosis. Exp Eye Res 2020; 201:108272. [PMID: 33010289 DOI: 10.1016/j.exer.2020.108272] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 12/16/2022]
Abstract
Myofibroblasts are alpha-smooth muscle actin (SMA)+ cells that have a critical role in the corneal stromal response to infections, injuries, and surgeries, and which produce corneal scarring fibrosis when they develop in excess. These contractile and opaque cells-produce large amounts of disordered extracellular matrix (ECM)-and develop from keratocyte-derived corneal fibroblasts or bone marrow-derived fibrocytes, and possibly other cell types, in response to TGFβ1, TGFβ2 and PDGF from the epithelium, tears, endothelium, and other stromal cells. Recent proteomic analyses have revealed that the myofibroblasts that develop from different progenitors aren't interchangeable, but have major differences in protein expression and functions. Absence or defective regeneration of the epithelial basement membrane (EBM) and/or Descemet's basement membrane (DBM) results in development and persistence of myofibroblasts in the corneal stroma. The functions of myofibroblasts in the cornea include production of volume-additive ECM, tissue contraction, production of various growth factors, cytokines and chemokines that regulate stromal cells, including other myofibroblasts, production of collagenases and metalloproteinases involved in tissue remodeling, and the expression of toll-like receptors that likely have critical roles in the clearance of bacteria and viruses causing corneal infections.
Collapse
|
21
|
Feghali-Bostwick C. Pulmonary fibrosis: something old, something new…still waiting for a breakthrough. Am J Physiol Lung Cell Mol Physiol 2020; 319:L560-L561. [PMID: 32755317 PMCID: PMC7518060 DOI: 10.1152/ajplung.00366.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Carol Feghali-Bostwick
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
22
|
Herrera JA, Mallikarjun V, Rosini S, Montero MA, Lawless C, Warwood S, O’Cualain R, Knight D, Schwartz MA, Swift J. Laser capture microdissection coupled mass spectrometry (LCM-MS) for spatially resolved analysis of formalin-fixed and stained human lung tissues. Clin Proteomics 2020; 17:24. [PMID: 32565759 PMCID: PMC7302139 DOI: 10.1186/s12014-020-09287-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/11/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Haematoxylin and eosin (H&E)-which respectively stain nuclei blue and other cellular and stromal material pink-are routinely used for clinical diagnosis based on the identification of morphological features. A richer characterization can be achieved by laser capture microdissection coupled to mass spectrometry (LCM-MS), giving an unbiased assay of the proteins that make up the tissue. However, the process of fixing and H&E staining of tissues provides challenges with standard sample preparation methods for mass spectrometry, resulting in low protein yield. Here we describe a microproteomics technique to analyse H&E-stained, formalin-fixed paraffin-embedded (FFPE) tissues. METHODS Herein, we utilize heat extraction, physical disruption, and in column digestion for the analysis of H&E stained FFPE tissues. Micro-dissected morphologically normal human lung alveoli (0.082 mm3) and human lung blood vessels (0.094 mm3) from FFPE-fixed H&E-stained sections from Idiopathic Pulmonary Fibrosis (IPF) specimens (n = 3 IPF specimens) were then subject to a qualitative and then quantitative proteomics approach using BayesENproteomics. In addition, we tested the sensitivity of this method by processing and analysing a range of micro-dissected human lung blood vessel tissue volumes. RESULTS This approach yields 1252 uniquely expressed proteins (at a protein identification threshold of 3 unique peptides) with 892 differentially expressed proteins between these regions. In accord with prior knowledge, our methodology approach confirms that human lung blood vessels are enriched with smoothelin, CNN1, ITGA7, MYH11, TAGLN, and PTGIS; whereas morphologically normal human lung alveoli are enriched with cytokeratin-7, -8, -18, -19, 14, and -17. In addition, we identify a total of 137 extracellular matrix (ECM) proteins and immunohistologically validate that laminin subunit beta-1 localizes to morphologically normal human lung alveoli and tenascin localizes to human lung blood vessels. Lastly, we show that this micro-proteomics technique can be applied to tissue volumes as low as 0.0125 mm3. CONCLUSION Herein we show that our multistep sample preparation methodology of LCM-MS can identify distinct, characteristic proteomic compositions of anatomical features within complex fixed and stained tissues.
Collapse
Affiliation(s)
- Jeremy A. Herrera
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Venkatesh Mallikarjun
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Silvia Rosini
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Maria Angeles Montero
- Histopathology Department, Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT UK
| | - Craig Lawless
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Stacey Warwood
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Ronan O’Cualain
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - David Knight
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Martin A. Schwartz
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| | - Joe Swift
- The Wellcome Centre for Cell-Matrix Research, University of Manchester, Manchester, M13 9PT UK
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL UK
| |
Collapse
|
23
|
de Brito AA, da Silveira EC, Rigonato-Oliveira NC, Soares SS, Brandao-Rangel MAR, Soares CR, Santos TG, Alves CE, Herculano KZ, Vieira RP, Lino-Dos-Santos-Franco A, Albertini R, Aimbire F, de Oliveira AP. Low-level laser therapy attenuates lung inflammation and airway remodeling in a murine model of idiopathic pulmonary fibrosis: Relevance to cytokines secretion from lung structural cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 203:111731. [PMID: 31935633 DOI: 10.1016/j.jphotobiol.2019.111731] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 09/25/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and chronic inflammatory disease with a poor prognosis and very few available treatment options. Low-level laser therapy (LLLT) has been gaining prominence as a new and effective anti-inflammatory and immunomodulatory agent. Can lung inflammation and the airway remodeling be regulated by LLLT in an experimental model of IPF in C57Bl/6 mice? The present study investigated if laser attenuates cellular migration to the lungs, the airway remodeling as well as pro-fibrotic cytokines secretion from type II pneumocytes and fibroblasts. Mice were irradiated (780 nm and 30 mW) and then euthanized fifteen days after bleomycin-induced lung fibrosis. Lung inflammation and airway remodeling were evaluated through leukocyte counting in bronchoalveolar lavage fluid (BALF) and analysis of collagen in lung, respectively. Inflammatory cells in blood were also measured. For in vitro assays, bleomycin-activated fibroblasts and type II pneumocytes were irradiated with laser. The pro- and anti-inflammatory cytokines level in BALF as well as cells supernatant were measured by ELISA, and the TGFβ in lung was evaluated by flow cytometry. Lung histology was used to analyze collagen fibers around the airways. LLLT reduced both migration of inflammatory cells and deposition of collagen fibers in the lungs. In addition, LLLT downregulated pro-inflammatory cytokines and upregulated the IL-10 secretion from fibroblasts and pneumocytes. Laser therapy greatly reduced total lung TGFβ. Systemically, LLLT also reduced the inflammatory cells counted in blood. There is no statistical difference in inflammatory parameters studied between mice of the basal group and the laser-treated mice. Data obtained indicate that laser effectively attenuates the lung inflammation, and the airway remodeling in experimental pulmonary fibrosis is driven to restore the balance between the pro- and anti-inflammatory cytokines in lung and inhibit the pro-fibrotic cytokines secretion from fibroblasts.
Collapse
Affiliation(s)
- Auriléia Aparecida de Brito
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Elaine Cristina da Silveira
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | | | - Stephanie Souza Soares
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Maysa Alves Rodrigues Brandao-Rangel
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São José dos Campos, SP, Brazil; Post Graduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Clariana Rodrigues Soares
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Tawany Gonçalves Santos
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Cintia Estefano Alves
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Karine Zanella Herculano
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Rodolfo Paula Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São José dos Campos, SP, Brazil; Post Graduate Program in Rehabilitation Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil; Post-Graduation Program in Sciences of Human Moviment and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil
| | - Adriana Lino-Dos-Santos-Franco
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Regiane Albertini
- Post-Graduation Program in Sciences of Human Moviment and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP, Brazil; Institute of Science and Technology, Federal University of Sao Paulo (UNIFESP), Sao Jose dos Campos, SP, Brazil
| | - Flavio Aimbire
- Institute of Science and Technology, Federal University of Sao Paulo (UNIFESP), Sao Jose dos Campos, SP, Brazil.
| | - Ana Paula de Oliveira
- Post Graduate Program in Biphotonic Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| |
Collapse
|
24
|
Valenzi E, Bulik M, Tabib T, Morse C, Sembrat J, Trejo Bittar H, Rojas M, Lafyatis R. Single-cell analysis reveals fibroblast heterogeneity and myofibroblasts in systemic sclerosis-associated interstitial lung disease. Ann Rheum Dis 2019; 78:1379-1387. [PMID: 31405848 DOI: 10.1136/annrheumdis-2018-214865] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/28/2019] [Accepted: 07/12/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Myofibroblasts are key effector cells in the extracellular matrix remodelling of systemic sclerosis-associated interstitial lung disease (SSc-ILD); however, the diversity of fibroblast populations present in the healthy and SSc-ILD lung is unknown and has prevented the specific study of the myofibroblast transcriptome. We sought to identify and define the transcriptomes of myofibroblasts and other mesenchymal cell populations in human healthy and SSc-ILD lungs to understand how alterations in fibroblast phenotypes lead to SSc-ILD fibrosis. METHODS We performed droplet-based, single-cell RNA-sequencing with integrated canonical correlation analysis of 13 explanted lung tissue specimens (56 196 cells) from four healthy control and four patients with SSc-ILD, with findings confirmed by cellular indexing of transcriptomes and epitopes by sequencing in additional samples. RESULTS Examination of gene expression in mesenchymal cells identified two major, SPINT2hi and MFAP5hi, and one minor, WIF1hi, fibroblast populations in the healthy control lung. Combined analysis of control and SSc-ILD mesenchymal cells identified SPINT2hi, MFAP5hi, few WIF1hi fibroblasts and a new large myofibroblast population with evidence of actively proliferating myofibroblasts. We compared differential gene expression between all SSc-ILD and control mesenchymal cell populations, as well as among the fibroblast subpopulations, showing that myofibroblasts undergo the greatest phenotypic changes in SSc-ILD and strongly upregulate expression of collagens and other profibrotic genes. CONCLUSIONS Our results demonstrate previously unrecognised fibroblast heterogeneity in SSc-ILD and healthy lungs, and define multimodal transcriptome-phenotypes associated with these populations. Our data indicate that myofibroblast differentiation and proliferation are key pathological mechanisms driving fibrosis in SSc-ILD.
Collapse
Affiliation(s)
- Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melissa Bulik
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christina Morse
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|