1
|
Colin Waldo MD, Quintero-Millán X, Negrete-García MC, Ruiz V, Sommer B, Romero-Rodríguez DP, Montes-Martínez E. Circulating MicroRNAs in Idiopathic Pulmonary Fibrosis: A Narrative Review. Curr Issues Mol Biol 2024; 46:13746-13766. [PMID: 39727949 DOI: 10.3390/cimb46120821] [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: 10/14/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, deathly disease with no recognized effective cure as yet. Furthermore, its diagnosis and differentiation from other diffuse interstitial diseases remain a challenge. Circulating miRNAs have been measured in IPF and have proven to be an adequate option as biomarkers for this disease. These miRNAs, released into the circulation outside the cell through exosomes and proteins, play a crucial role in the pathogenic pathways and mechanisms involved in IPF development. This review focuses on the serum/plasma miRNAs reported in IPF that have been validated by real-time PCR and the published evidence regarding the fibrotic process. First, we describe the mechanisms by which miRNAs travel through the circulation (contained in exosomes and bound to proteins), as well as the mechanism by which miRNAs perform their function within the cell. Subsequently, we summarize the evidence concerning miRNAs reported in serum/plasma, where we find contradictory functions in some miRNAs (dual functions in IPF) when comparing the findings in vitro vs. in vivo. The most relevant finding, for instance, the levels of miRNAs let-7d and miR-21 reported in the serum/plasma in IPF, correspond to those found in studies in lung fibroblasts and the murine bleomycin model, reinforcing the usefulness of these miRNAs as future biomarkers in IPF.
Collapse
Affiliation(s)
- Marisa Denisse Colin Waldo
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Xochipilzihuitl Quintero-Millán
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Maria Cristina Negrete-García
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Víctor Ruiz
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Bettina Sommer
- Bronchial Hyperreactivity Research Department, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Dámaris P Romero-Rodríguez
- Conahcyt National Laboratory for Research and Diagnosis by Immunocytofluorometry (LANCIDI), National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Eduardo Montes-Martínez
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| |
Collapse
|
2
|
Wu W, Yu N, Chen W, Zhu Y. ANRIL upregulates TGFBR1 to promote idiopathic pulmonary fibrosis in TGF-β1-treated lung fibroblasts via sequestering let-7d-5p. Epigenetics 2024; 19:2435682. [PMID: 39612365 DOI: 10.1080/15592294.2024.2435682] [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: 04/01/2024] [Revised: 10/20/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and life-threatening respiratory disease characterized by worsening lung function due to excessive scarring. The objective of this study was to investigate the role of the long non-coding RNA ANRIL (antisense non-coding RNA in the INK4 locus) in the development of IPF. Our research revealed a significant increase in ANRIL expression in pulmonary fibrosis, consistent with prior studies indicating elevated ANRIL levels in fibrotic tissues. In vitro experiments demonstrated that elevated ANRIL expression promoted fibroblast activation, as evidenced by the upregulation of fibrosis-related markers. Mechanistically, we found that ANRIL interacts with let-7d-5p, a microRNA involved in gene regulation, acting as a sponge for let-7d-5p. Functional experiments confirmed a potential influence of let-7d-5p on fibroblast activation through direct interaction with ANRIL. Furthermore, our investigation identified TGFBR1 as a potential mediator of ANRIL's fibrogenic effects. Silence of TGFBR1 mitigated the fibrotic phenotype induced by ANRIL overexpression. Collectively, these results suggest that ANRIL promotes fibroblast activation and fibrosis development, possibly through the let-7d-5p/TGFBR1 axis, indicating that ANRIL could be a potential therapeutic target for pulmonary fibrosis.
Collapse
Affiliation(s)
- Weidong Wu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, Fujian, China
| | - Nanding Yu
- Department of Pulmonary and Critical Care Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Department of Geriatric Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Weiming Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, Fujian, China
| | - Yong Zhu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
- Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
3
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis. Part I: Thoracic organs. J Transl Med 2024; 22:609. [PMID: 38956586 PMCID: PMC11218337 DOI: 10.1186/s12967-024-05244-1] [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: 02/12/2024] [Accepted: 04/27/2024] [Indexed: 07/04/2024] Open
Abstract
Sustained injury from factors such as hypoxia, infection, or physical damage may provoke improper tissue repair and the anomalous deposition of connective tissue that causes fibrosis. This phenomenon may take place in any organ, ultimately leading to their dysfunction and eventual failure. Tissue fibrosis has also been found to be central in both the process of carcinogenesis and cancer progression. Thus, its prompt diagnosis and regular monitoring is necessary for implementing effective disease-modifying interventions aiming to reduce mortality and improve overall quality of life. While significant research has been conducted on these subjects, a comprehensive understanding of how their relationship manifests through modern imaging techniques remains to be established. This work intends to provide a comprehensive overview of imaging technologies relevant to the detection of fibrosis affecting thoracic organs as well as to explore potential future advancements in this field.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
4
|
He W, Cui B, Chu Z, Chen X, Liu J, Pang X, Huang X, Yin H, Lin H, Peng L. Radiomics based on HRCT can predict RP-ILD and mortality in anti-MDA5 + dermatomyositis patients: a multi-center retrospective study. Respir Res 2024; 25:252. [PMID: 38902680 PMCID: PMC11191144 DOI: 10.1186/s12931-024-02843-w] [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: 01/09/2024] [Accepted: 05/08/2024] [Indexed: 06/22/2024] Open
Abstract
OBJECTIVES To assess the effectiveness of HRCT-based radiomics in predicting rapidly progressive interstitial lung disease (RP-ILD) and mortality in anti-MDA5 positive dermatomyositis-related interstitial lung disease (anti-MDA5 + DM-ILD). METHODS From August 2014 to March 2022, 160 patients from Institution 1 were retrospectively and consecutively enrolled and were randomly divided into the training dataset (n = 119) and internal validation dataset (n = 41), while 29 patients from Institution 2 were retrospectively and consecutively enrolled as external validation dataset. We generated four Risk-scores based on radiomics features extracted from four areas of HRCT. A nomogram was established by integrating the selected clinico-radiologic variables and the Risk-score of the most discriminative radiomics model. The RP-ILD prediction performance of the models was evaluated by using the area under the receiver operating characteristic curves, calibration curves, and decision curves. Survival analysis was conducted with Kaplan-Meier curves, Mantel-Haenszel test, and Cox regression. RESULTS Over a median follow-up time of 31.6 months (interquartile range: 12.9-49.1 months), 24 patients lost to follow-up and 46 patients lost their lives (27.9%, 46/165). The Risk-score based on bilateral lungs performed best, attaining AUCs of 0.869 and 0.905 in the internal and external validation datasets. The nomogram outperformed clinico-radiologic model and Risk-score with AUCs of 0.882 and 0.916 in the internal and external validation datasets. Patients were classified into low- and high-risk groups with 50:50 based on nomogram. High-risk group patients demonstrated a significantly higher risk of mortality than low-risk group patients in institution 1 (HR = 4.117) and institution 2 cohorts (HR = 7.515). CONCLUSION For anti-MDA5 + DM-ILD, the nomogram, mainly based on radiomics, can predict RP-ILD and is an independent predictor of mortality.
Collapse
Affiliation(s)
- Wenzhang He
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610000, China
- Department of Radiology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Beibei Cui
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610000, China
| | - Zhigang Chu
- Department of Radiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyi Chen
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610000, China
| | - Jing Liu
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610000, China
| | - Xueting Pang
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610000, China
| | - Xuan Huang
- Biomedical Big Data Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongkun Yin
- Institute of Advanced Research, Infervision Medical Technology, Beijing, China
| | - Hui Lin
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, Sichuan, 610000, China.
| | - Liqing Peng
- Department of Radiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610000, China.
| |
Collapse
|
5
|
Lee HM, Liu DY, Hsu HL, Yu TL, Yu TS, Shen TC, Tsai FJ. Risk of depression in patients with pneumoconiosis: A population-based retrospective cohort study. J Affect Disord 2024; 352:146-152. [PMID: 38369263 DOI: 10.1016/j.jad.2024.02.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Pneumoconiosis is an important occupational disease; the association between pneumoconiosis and depression was largely unknown. This study aimed to investigate the relationship between pneumoconiosis and the risk of subsequent depression. METHODS A retrospective cohort study was conducted using Taiwan's National Health Insurance database. The study included 16,795 patients diagnosed with pneumoconiosis between 2008 and 2018 and a comparison cohort of 67,180 individuals without pneumoconiosis, propensity score matched in a 1:4 ratio based on age, sex, monthly income, residential urbanization level, and date of pneumoconiosis diagnosis. The development of depression was monitored until the end of 2019. RESULTS The incidence of depression was 1.68 times higher in the pneumoconiosis cohort than that in the comparison cohort, with an incidence rate of 10.07 versus 5.99 per 1000 person-years (adjusted hazard ratio [aHR] = 1.84, 95 % confidence interval [CI] = 1.70-1.99). The risk of depression increased with an increased mean annual number of emergency department visits for pneumoconiosis, with aHRs of 1.34 (95 % CI = 1.13-1.59) and 2.31 (95 % CI = 1.94-2.76) for 1 ≤ n < 2, and n ≥ 2 compared to n < 1, respectively. LIMITATION The database lacked detailed socioeconomic history, family history, and clinical variables. CONCLUSION This study found that patients with pneumoconiosis have a significantly higher risk of depression than those without pneumoconiosis. Furthermore, the risk of depression increases with the frequency of emergency department visits for pneumoconiosis. Healthcare professionals should pay close attention to the mental health of patients with pneumoconiosis.
Collapse
Affiliation(s)
- Hsiu-Ming Lee
- Department of Education, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - De-Yi Liu
- Department of Education, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiang-Li Hsu
- Department of Education, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Tsai-Ling Yu
- Department of Education, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Teng-Shun Yu
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Te-Chun Shen
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan; Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; Division of Critical Care Medicine, Chu Shang Show Chwan Hospital, Nantou, Taiwan.
| | - Fuu-Jen Tsai
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
6
|
Sun C, Liu H, Chi B, Han J, Koga Y, Afshar K, Liu X. Improvement of idiopathic pulmonary fibrosis through a combination of Astragalus radix and Angelica sinensis radix via mammalian target of rapamycin signaling pathway-induced autophagy in rat. J Thorac Dis 2024; 16:1397-1411. [PMID: 38505077 PMCID: PMC10944740 DOI: 10.21037/jtd-24-28] [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: 01/05/2024] [Accepted: 02/18/2024] [Indexed: 03/21/2024]
Abstract
Background There is a major need for effective, well-tolerated treatments for idiopathic pulmonary fibrosis (IPF) in clinic. Astragalus radix (AR; Huangqi) and Angelica sinensis radix (AS; Danggui) have been frequently used in the treatment of IPF. This study aimed to reveal the pharmacological effects and the mechanisms of the action of an AR-AS combination in treating IPF. Methods Sprague-Dawley rats were randomly divided into six groups (n=5): control, bleomycin (BLM) model, AR, AS, AR + AS, and prednisone (PDN) groups. A transforming growth factor-β1 (TGF-β1)-induced MRC-5 cell model were also used. Pulmonary fibrosis, inflammation, oxidative stress, and autophagy were evaluated by performing hematoxylin and eosin (H&E) staining, Masson staining, immunohistochemical staining, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, enzyme-linked immunosorbent assay (ELISA), immunofluorescence, and hydroxyproline assay following the treatment of AR, AS, and the AR-AS herb pair. Results Rats administered the AR-AS herb pair had lower α-smooth muscle actin (α-SMA), collagen I, fibronectin, and vimentin levels in lung tissues, and lower inflammatory cytokine levels in rat serum. In addition, the AR-AS herb pair induced mammalian target of rapamycin (mTOR)-mediated autophagy and reduced oxidative stress in BLM-induced rats. The effects of the AR and AS combination were confirmed in MRC-5 cells treated with TGF-β1. Specifically, the combination of AR and AS attenuated MRC-5 cell fibrosis, inflammation, and oxidative stress while inducing autophagy. Conclusions The combination of AR and AS protects against IPF by inducing autophagy via inhibiting the mTOR signaling pathway. The synergistic action of AR and AS is superior to that of either AR or AS alone.
Collapse
Affiliation(s)
- Chao Sun
- Department of Disease Prevention, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huaman Liu
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Baihong Chi
- Department of Pulmonary and Critical Medicine, People’s Hospital Rizhao, Rizhao, China
| | - Jia Han
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yasuhiko Koga
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Kamyar Afshar
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California San Diego, La Jolla, CA, USA
| | - Xue Liu
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
7
|
Pitre T, Lupas D, Ebeido I, Colak A, Modi M, Kachkovski GV, Montesi SB, Khor YH, Kawano-Dourado L, Jenkins G, Fisher JH, Shapera S, Rochwerg B, Couban R, Zeraatkar D. Prognostic factors associated with mortality in acute exacerbations of idiopathic pulmonary fibrosis: A systematic review and meta-analysis. Respir Med 2024; 222:107515. [PMID: 38154738 DOI: 10.1016/j.rmed.2023.107515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Acute exacerbations of idiopathic pulmonary fibrosis (AE-IPF) increases mortality risk, but which factors increase mortality is unknown. We aimed to perform a prognostic review of factors associated with mortality in patients with IPF. STUDY DESIGN and methods: We searched MEDLINE, EMBASE, and CINAHL for studies that reported on the association between any prognostic factor and AE-IPF. We assessed risk of bias using the QUIPS tool. We conduced pairwise meta-analyses using REML heterogeneity estimator, and GRADE approach to assess the certainty of the evidence. RESULTS We included 35 studies in our analysis. We found that long-term supplemental oxygen at baseline (aHR 2.52 [95 % CI 1.68 to 3.80]; moderate certainty) and a diagnosis of IPF compared to non-IPF ILD (aHR 2.19 [95 % CI 1.22 to 3.92]; moderate certainty) is associated with a higher risk of death in patients with AE-IPF. A diffuse pattern on high resolution computed tomography (HRCT) compared to a non-diffuse pattern (aHR 2.61 [95 % CI 1.32 to 2.90]; moderate certainty) is associated with a higher risk of death in patients with AE-IPF. We found that using corticosteroids prior to hospital admission (aHR 2.19 [95 % CI 1.26 to 3.82]; moderate certainty) and those with increased neutrophils (by % increase) in bronchoalveolar lavage (BAL) during the exacerbation is associated with a higher risk of death (aHR 1.02 [1.01 to 1.04]; moderate certainty). INTERPRETATION Our results have implications for healthcare providers in making treatment decisions and prognosticating the clinical trajectory of patients, for researchers to design future interventions to improve patient trajectory, and for guideline developers in making decisions about resource allocation.
Collapse
Affiliation(s)
- Tyler Pitre
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Daniel Lupas
- Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Ibrahim Ebeido
- Faculty of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexander Colak
- Faculty of Medicine, University of British Columbia, BC, Canada
| | - Mihir Modi
- Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - George V Kachkovski
- Faculty of Health Sciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yet H Khor
- Respiratory Research Alfred, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Respiratory and Sleep Medicine, Austin Health, Heidelberg, Victoria, Australia; Institute for Breathing and Sleep, Heidelberg, Victoria, Australia; Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Leticia Kawano-Dourado
- Hcor Research Institute, Hospital do Coracao, Sao Paulo, Brazil; Pulmonary Division, University of Sao Paulo, Sao Paulo, Brazil; MAGIC Evidence Ecosystem, Oslo, Norway
| | - Gisli Jenkins
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Heart and Lung Institute, Imperial College London, UK
| | - Jolene H Fisher
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shane Shapera
- Division of Respirology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Bram Rochwerg
- Department of Medicine, McMaster University, Hamilton, ON, Canada; Health Research Methods Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Rachel Couban
- Department of Anesthesiology, McMaster University, Hamilton, ON, Canada
| | - Dena Zeraatkar
- Department of Medicine, McMaster University, Hamilton, ON, Canada; Department of Anesthesiology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
8
|
Suzuki T, Karayama M, Aoshima Y, Mori K, Yoshizawa N, Ichikawa S, Kato S, Yokomura K, Kono M, Hashimoto D, Inoue Y, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Goshima S, Inui N, Suda T. Association of the lung immune prognostic index with the survival of patients with idiopathic interstitial pneumonias. Respirology 2024; 29:136-145. [PMID: 37921012 DOI: 10.1111/resp.14621] [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: 03/31/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND AND OBJECTIVE The lung immune prognostic index (LIPI), a simple index calculated from the blood lactate dehydrogenase level and derived neutrophil-to-lymphocyte ratio, is thought to be associated with host immune status. However, the utility of LIPI in patients with idiopathic interstitial pneumonias (IIPs) is unknown. METHODS In this multicentre, retrospective, observational study, an association between LIPI and the survival of patients with IIPs was evaluated. RESULTS Exploratory and validation cohorts consisting of 460 and 414 patients with IIPs, respectively, were included (159 and 159 patients had idiopathic pulmonary fibrosis [IPF], and 301 and 255 had non-IPF, respectively). In the exploratory cohort, patients with IPF and a low LIPI had significantly better survival than those with a high LIPI (median of 5.6 years vs. 3.9 years, p = 0.016). The predictive ability of LIPI for the survival of patients with IPF was validated in the validation cohort (median of 8.5 years vs. 4.4 years, p = 0.003). In a multivariate Cox proportional hazard analysis, LIPI was selected as an independent predictive factor for the survival of IPF patients. There was no significant association between LIPI and survival of non-IPF patients in the exploratory and validation cohorts. CONCLUSION The LIPI was a predictive factor for the survival of patients with IPF and could aid the management of IPF.
Collapse
Affiliation(s)
- Takahito Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yoichiro Aoshima
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazutaka Mori
- Department of Respiratory Medicine, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| | - Nobuko Yoshizawa
- Department of Radiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shintaro Ichikawa
- Department of Radiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Shinpei Kato
- Department of Respiratory Medicine, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Koshi Yokomura
- Department of Respiratory Medicine, Seirei Mikatahara General Hospital, Hamamatsu, Japan
| | - Masato Kono
- Department of Respiratory Medicine, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Dai Hashimoto
- Department of Respiratory Medicine, Seirei Hamamatsu General Hospital, Hamamatsu, Japan
| | - Yusuke Inoue
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideki Yasui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Goshima
- Department of Radiology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
- Department of Clinical Pharmacology and Therapeutics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
9
|
Reghelin CK, Bastos MS, de Souza Basso B, Costa BP, Lima KG, de Sousa AC, Haute GV, Diz FM, Dias HB, Luft C, Rodrigues KF, Garcia MCR, Matzenbacher LS, Adami BS, Xavier LL, Donadio MVF, de Oliveira JR, da Silva Melo DA. Bezafibrate reduces the damage, activation and mechanical properties of lung fibroblast cells induced by hydrogen peroxide. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3857-3866. [PMID: 37358795 DOI: 10.1007/s00210-023-02595-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
In pulmonary fibrosis, the proliferation of fibroblasts and their differentiation into myofibroblasts is often caused by tissue damage, such as oxidative damage caused by reactive oxygen species, which leads to progressive rupture and thus destruction of the alveolar architecture, resulting in cell proliferation and tissue remodeling. Bezafibrate (BZF) is an important member of the peroxisome proliferator-activated receptor (PPARs) family agonists, used in clinical practice as antihyperlipidemic. However, the antifibrotic effects of BZF are still poorly studied. The objective of this study was to evaluate the effects of BZF on pulmonary oxidative damage in lung fibroblast cells. MRC-5 cells were treated with hydrogen peroxide (H2O2) to induce oxidative stress activation and BZF treatment was administered at the same moment as H2O2 induction. The outcomes evaluated were cell proliferation and cell viability; oxidative stress markers such as reactive oxygen species (ROS), catalase (CAT) levels and thiobarbituric acid reactive substances (TBARS); col-1 and α-SMA mRNA expression and cellular elasticity through Young's modulus analysis evaluated by atomic force microscopy (AFM). The H2O2-induced oxidative damage decreased the cell viability and increased ROS levels and decreased CAT activity in MRC-5 cells. The expression of α-SMA and the cell stiffness increased in response to H2O2 treatment. Treatment with BZF decreased the MRC-5 cell proliferation, ROS levels, reestablished CAT levels, decreased the mRNA expression of type I collagen protein (col-1) and α-smooth muscle actin (α-SMA), and cellular elasticity even with H2O2 induction. Our results suggest that BZF has a potential protective effect on H2O2-induced oxidative stress. These results are based on an in vitro experiment, derived from a fetal lung cell line and may emerge as a possible new therapy for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Camille Kirinus Reghelin
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Matheus Scherer Bastos
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil.
- Laboratório de Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), 6681 Ipiranga Ave., Porto Alegre, RS, Zip Code: 90619-900, Brazil.
| | - Bruno de Souza Basso
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Bruna Pasqualotto Costa
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Kelly Goulart Lima
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Arieli Cruz de Sousa
- Departamento de Bioquímica, ICBS, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo I, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Gabriela Viegas Haute
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Fernando Mendonça Diz
- Programa de Pós-Graduação Em Engenharia E Tecnologia de Materiais, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Henrique Bregolin Dias
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Carolina Luft
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Kétlin Fernanda Rodrigues
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Maria Cláudia Rosa Garcia
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Lucas Strassburger Matzenbacher
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Bruno Silveira Adami
- Laboratório Central de Microscopia E Microanálise (LabCEMM), Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Léder Leal Xavier
- Laboratório Central de Microscopia E Microanálise (LabCEMM), Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Márcio Vinícius Fagundes Donadio
- Laboratório de Atividade Física Pediátrica, Centro Infantil, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Jarbas Rodrigues de Oliveira
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Denizar Alberto da Silva Melo
- Laboratório de Pesquisa Em Biofísica Celular E Inflamação, Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), Porto Alegre, RS, Brazil
| |
Collapse
|
10
|
Wu Y, Zhang J, Wang X, Xu Y, Zheng J. Saikosaponin-d regulates angiogenesis in idiopathic pulmonary fibrosis through angiopoietin/Tie-2 pathway. Acta Histochem 2023; 125:152100. [PMID: 37837833 DOI: 10.1016/j.acthis.2023.152100] [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: 04/12/2023] [Revised: 09/06/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is considered as a chronic interstitial lung disease with underlying mechanism of IPF remaining unclear, while there are no definitive treatment options. In recent years, scientists have gradually paid attention to the influence of angiogenesis on IPF. Because IPF is a progressive with microvascular remodeling disorder, scientists have postulated that angiogenesis may also be one of the initiating and contributing factors of the disease. Bupleurum is a common natural Chinese herbal medicine with antibacterial, anti-inflammatory, anti-tumor and other pharmacological effects. As the most important active monomer of Bupleurum, Saikosaponin-d (SSd) is a new discovery with anti-pulmonary fibrosis (PF) activity. This study attempts to investigate the role of SSd in the interference of PF through regulation of angiogenesis in IPF through Angiopoietin (Angpt) /Tie receptor 2 (Tie2) pathway. METHODS Randomly, we allocated C57BL/6 mice into four groups (n = 20 in each group). Afterwards, establishment of IPF model was accomplished via intratracheal administration of bleomycin (BLM, 5 mg/kg), while corresponding drug intervention was given accordingly. On 3rd, 7th, 14th and 28th days after modeling, we performed histopathological examination through staining. Meanwhile, immunohistochemistry (IHC) of PF and the expression of related factors were observed, while Ang/Tie2 pathway was assessed by ELISA with the effect of SSd on angiogenesis related proteins in IPF being explored with IHC and Western Blot technique. RESULTS Our results showed that SSd could reduce inflammation and PF levels in lung tissue of experimental mice, while levels of angiogenesis-related factors, namely Tie-2, Ang-1 and ANGPT2 (Ang-2), fibrosis- associated factors like Alpha-smooth muscle actin (α-SMA), collagen-I and hydroxyproline in SSd and dexamethasone (DXM) mice were significantly reduced at each time point compared to BLM (p < 0.01). Additionally, we discovered substantial decreased expressions of Ang-1, Ang-2, Tie-2, α-SMA and collagen-I at protein level in SSd and DXM mice at each time point compared to BLM (p < 0.05). Besides, insignificant differences were observed between SSd and DXM groups (p > 0.05). CONCLUSION This study has demonstrated that SSd could down-regulate the expression of ANG-1, Ang-2 and Tie2 in the Ang/Tie2 pathway, and may reduce lung inflammation and PF in BLM-induced mice via inhibition of angiogenesis.
Collapse
Affiliation(s)
- Yan Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Binhu District, Wuxi City, Jiangsu 214122, China
| | - Jun Zhang
- Department of Respiratory and Critical Care Medicine, Aoyang Hospital Affiliated to Jiangsu University, 279 Jingang Dadao, Zhangjiagang City, Jiangsu 215631, China
| | - Xintian Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China
| | - Yuncong Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China
| | - Jinxu Zheng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Jingkou District, Zhenjiang City, Jiangsu 212000, China.
| |
Collapse
|
11
|
Feng X, Duan Y, Lv X, Li Q, Liang B, Ou X. The Impact of Lung Cancer in Patients with Combined Pulmonary Fibrosis and Emphysema (CPFE). J Clin Med 2023; 12:jcm12031100. [PMID: 36769748 PMCID: PMC9917551 DOI: 10.3390/jcm12031100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 02/02/2023] Open
Abstract
Given the high risk of lung cancer (LC) in patients with combined pulmonary fibrosis and emphysema (CPFE), and the difficulty of early diagnosis, it is important to understand the impact of LC in these patients. The effect of LC on the development of acute exacerbation (AE) as a natural course of CPFE is still unknown. We retrospectively reviewed medical records of patients at the West China Hospital and enrolled 59 patients with CPFE combined with LC and 68 CPFE patients without LC for initial diagnosis matched in the same period. We compared the clinical characteristics and imaging features of CPFE patients with LC and without LC, and analyzed the associated factors for the prevalence of LC using binary logistic regression. Cox proportional hazards regression analysis was performed to explore risk factors of AE as a natural course of CPFE. Patients with CPFE combined with LC were more common among elderly male smokers. The most common pathological type of tumor was adenocarcinoma (24/59, 40.7%) and squamous cell carcinoma (18/59, 30.5%). Compared with those in the without LC group, the proportions of men, and ex- or current smokers, and the levels of smoking pack-years, serum CRP, IL-6, fibrinogen, complement C3 and C4 in patients with LC were significantly higher (p < 0.05). There was no significant difference in the proportion of natural-course-related AE (10.2% vs. 16.2%, p > 0.05) between the two groups. Logistic regression analysis demonstrated that pack-years ≥ 20 (OR: 3.672, 95% CI: 1.165-11.579), family history of cancer (OR: 8.353, 95% CI: 2.368-10.417), the level of fibrinogen > 4.81 g/L (OR: 3.628, 95% CI: 1.403-9.385) and serum C3 > 1.00 g/L (OR: 5.299, 95% CI: 1.727-16.263) were independently associated with LC in patients with CPFE. Compared to those without AE, CPFE patients with AE had significantly higher levels of PLR and serum CRP, with obviously lower DLCO and VC. The obviously increased PLR (HR: 3.731, 95% CI: 1.288-10.813), and decreased DLCO%pred (HR: 0.919, 95% CI: 0.863-0.979) and VC%pred (HR: 0.577, 95% CI: 0.137-0.918) rather than the presence of LC independently contributed to the development of natural-course-related AE in patients with CPFE. Pack-years, family history of cancer, the levels of fibrinogen and serum C3 were independently associated with LC in patients with CPFE. The presence of LC did not significantly increase the risk of AE as a natural course of CPFE. Clinicians should give high priority to CPFE patients, especially those with more severe fibrosis and systemic inflammation, in order to be alert for the occurrence of AE.
Collapse
Affiliation(s)
- Xiaoyi Feng
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yishan Duan
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiafei Lv
- Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qinxue Li
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Binmiao Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xuemei Ou
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, China
- Correspondence:
| |
Collapse
|
12
|
Pernia Marin M, Salvatore M. Fibrosis. J Transl Med 2023; 21:59. [PMID: 36717924 PMCID: PMC9887912 DOI: 10.1186/s12967-022-03789-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
13
|
Rodriguez K, Ashby CL, Varela VR, Sharma A. High-Resolution Computed Tomography of Fibrotic Interstitial Lung Disease. Semin Respir Crit Care Med 2022; 43:764-779. [PMID: 36307108 DOI: 10.1055/s-0042-1755563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
While radiography is the first-line imaging technique for evaluation of pulmonary disease, high-resolution computed tomography (HRCT) provides detailed assessment of the lung parenchyma and interstitium, allowing normal anatomy to be differentiated from superimposed abnormal findings. The fibrotic interstitial lung diseases have HRCT features that include reticulation, traction bronchiectasis and bronchiolectasis, honeycombing, architectural distortion, and volume loss. The characterization and distribution of these features result in distinctive CT patterns. The CT pattern and its progression over time can be combined with clinical, serologic, and pathologic data during multidisciplinary discussion to establish a clinical diagnosis. Serial examinations identify progression, treatment response, complications, and can assist in determining prognosis. This article will describe the technique used to perform HRCT, the normal and abnormal appearance of the lung on HRCT, and the CT patterns identified in common fibrotic lung diseases.
Collapse
Affiliation(s)
- Karen Rodriguez
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Christian L Ashby
- School of Medicine, Universidad Central del Caribe School of Medicine, Bayamón, Puerto Rico
| | - Valeria R Varela
- School of Medicine, Universidad Central del Caribe School of Medicine, Bayamón, Puerto Rico
| | - Amita Sharma
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
14
|
Fikry H, Saleh LA, Gawad SA. Therapeutic effect of adipose-derived mesenchymal stem cells (AD-MSCs) compared to pirfenidone on corticosteroid resistance in a mouse model of acute exacerbation of idiopathic pulmonary fibrosis. Histol Histopathol 2022; 37:1065-1083. [PMID: 35816024 DOI: 10.14670/hh-18-493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute exacerbation-idiopathic pulmonary fibrosis (AE-IPF) is a life-threatening condition. In the treatment of AE-IPF, corticosteroid medication is commonly utilized. However, there is insufficient evidence to justify its usage. Pirfenidone (PFD) has recently been discovered to be effective in the treatment of AE-IPF patients. However, regenerative therapy, such as stem cell therapy or tissue engineering, is necessary due to ineffective and limited therapies. Combining MSC transplantation with pharmacological therapy may also give additional benefits; nevertheless, its use must be proven experimentally. As a result, the goal of this study was to assess the therapeutic effects of adipose-derived mesenchymal stem cells (AD-MSCs) on corticosteroid resistance in an animal model of AE-IPF caused by bleomycin compared to PFD. MATERIALS AND METHODS Seventy C57BL/6J male mice were randomly divided into seven groups, control, BLM, methylprednisolone (MP), PFD, AD-MSCs, PFD +MP, and AD-MSCs +MP. RESULTS In terms of survival, collagen deposition, the acute lung injury score (ALI), and the Ashcroft score, AD-MSCs exceeded PFD. AD-MSCs + MP provided protection and preserved the lung's architecture in BLM-induced AE. In addition, AD-MSCs successfully decreased chemokine (CC motif) ligand-2 (CCL2) positive cells and lower pro-fibrotic and pro-inflammatory cytokines. CONCLUSIONS AD-MSCs enhanced histological structure, Ashcroft and ALI scores, lung collagen deposition, survival, and cytokines in an animal model of AE-IPF. As a result, we believe that AD-MSCs may be more therapeutically helpful for AE-IPF than presently available therapies, either alone or in conjunction with MP.
Collapse
Affiliation(s)
- Heba Fikry
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Lobna A Saleh
- Department of Clinical Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara Abdel Gawad
- Department of Histology and Cell Biology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
15
|
Liu M, Xue M, Zhang T, Lin R, Guo B, Chen Y, Cheng ZJ, Sun B. Detection of interstitial pneumonia with autoimmune features and idiopathic pulmonary fibrosis are enhanced by involvement of matrix metalloproteinases levels and clinical diagnosis. J Clin Lab Anal 2022; 36:e24734. [PMID: 36250225 DOI: 10.1002/jcla.24734] [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: 07/21/2022] [Revised: 09/06/2022] [Accepted: 10/05/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Higher detection of interstitial pneumonia with autoimmune features (IPAF), and idiopathic pulmonary fibrosis (IPF), has significant diagnostic and therapeutic implications. Some matrix metalloproteinases (MMPs) have become reliable diagnostic biomarkers in IPAF and IPF in previous studies, yet relevant reliability remains to be recognized. MATERIALS AND METHODS In this study, 36 ILDs patients, including 31 IPAF patients (Mean ± SD, 50.20 ± 5.10 years; 16 [51.6%] females) and five IPF patients (Mean ± SD, 61.20 ± 6.73 years; one [20.0%] females) were retrospectively enrolled. Serial serum samples were collected from patients with IPAF and IPF between January 2019 and December 2020. Notably, Serum MMPs levels were measured by U-PLEX Biomarker Group 1(Human) Multiplex Assays (MSD, USA). RESULTS A combination of MMPs and combinatorial biomarkers was strongly associated with clinical subjects in this study (AUC, 0.597 for Stability vs. Improvement and 0.756 for Stability vs. Exacerbation). Importantly, the AUC of MMP-12 reaches 0.730 (p < 0.05, Stability AUC vs. Improvement AUC) while MMP-13 reaches 0.741 (p < 0.05, Stability AUC vs. Exacerbation AUC) showed better performance than other MMPs in two comparisons. CONCLUSIONS Clinical risk factors and MMPs are strongly associated with either stratification of the disease of progression of IPAF or in two IPAF and IPF independent cohorts. To our knowledge, this is the first to illustrate that MMP-12 and MMP-13 may be expected to become typical promising biomarkers in Improvement - IPAF and Exacerbation - IPAF, respectively.
Collapse
Affiliation(s)
- Mingtao Liu
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingshan Xue
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Teng Zhang
- Faculty of Health Sciences, University of Macau, Taipa, China
| | - Runpei Lin
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baojun Guo
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - Zhangkai J Cheng
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
Zhou L, Tian H, Wang Q, Xiong W, Zhou X, Yan J. Effect of Qingfei Huaxian Decoction combined with prednisone acetate on serum inflammatory factors and pulmonary function of patients with idiopathic pulmonary fibrosis. Am J Transl Res 2022; 14:5905-5914. [PMID: 36105016 PMCID: PMC9452306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To determine the effect of Qingfei Huaxian Decoction combined with prednisone acetate on serum inflammatory factors and pulmonary function in patients with idiopathic pulmonary fibrosis (IPF). METHODS The clinical data of 118 patients with IPF treated in Wuhan Hospital of Traditional Chinese Medicine from June 2019 to August 2021 were retrospectively analyzed. Among the patients, 56 patients treated with prednisone acetate were assigned to the control group, and the remaining 62 patients treated with Qingfei Huaxian Decoction combined with prednisone acetate were assigned to the observation group. The efficacy and incidence of adverse reactions were compared between the two groups, and forced expiratory volume in 1 second (FEV1), forced vital capacity (FVC), FEV1/FVC, interleukin-6 (IL-6), interleukin-12 (IL-12), interleukin-18 (IL-18), hyaluronic acid (HA) and laminin (LN) in the two groups were evaluated before and after therapy. Logistic regression was conducted to analyze the risk factors impacting the treatment efficacy in patients. RESULTS After therapy, the observation group showed significantly higher efficacy than the control group. Compared with the control group, the observation group showed significantly higher levels of FEV1, FVC and FEV1/FVC, significantly lower levels of HA and LN, and a significantly higher IL-12 level (all P < 0.05). Therapeutic regimen, IL-6, IL-12, IL-18 and HA were independent risk factors impacting the efficacy of treatment in patients (P < 0.05). CONCLUSION Qingfei Huaxian Decoction combined with prednisone acetate has greater treatmetn efficacy in patients with IPF by improving the serum inflammatory factors and pulmonary function.
Collapse
Affiliation(s)
- Lei Zhou
- Nephrology Department, Wuhan Hospital of Traditional Chinese MedicineNo. 303 Sixin Avenue, Hanyang District, Wuhan 430050, Hubei, China
| | - Hui Tian
- Department of Pulmonary Diseases, Wuhan Hospital of Traditional Chinese MedicineWuhan 430050, Hubei, China
| | - Qiong Wang
- Endocrine Department, Wuhan Hospital of Traditional Chinese MedicineWuhan 430050, Hubei, China
| | - Wuzhong Xiong
- Endocrine Department, Wuhan Hospital of Traditional Chinese MedicineWuhan 430050, Hubei, China
| | - Xiang Zhou
- Clinical Laboratory, Wuhan Hospital of Traditional Chinese MedicineWuhan 430050, Hubei, China
| | - Jingjing Yan
- Department of Pulmonary Diseases, Wuhan Hospital of Traditional Chinese MedicineWuhan 430050, Hubei, China
| |
Collapse
|
17
|
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: 0.7] [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
|
18
|
Liu J, Gu L, Li W. The Prognostic Value of Integrated Analysis of Inflammation and Hypoxia-Related Genes in Idiopathic Pulmonary Fibrosis. Front Immunol 2022; 13:730186. [PMID: 35309336 PMCID: PMC8929415 DOI: 10.3389/fimmu.2022.730186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
Currently, the aetiology and pathogenesis of idiopathic pulmonary fibrosis (IPF) are still largely unclear. Moreover, patients with IPF exhibit a considerable difference in clinical presentation, treatment, and prognosis. Optimal biomarkers or models for IPF prognosis are lacking. Therefore, this study quantified the levels of various hallmarks using a single-sample gene set enrichment analysis algorithm. The hazard ration was calculated using Univariate Cox regression analysis based on the transcriptomic profile of bronchoalveolar lavage cells and clinical survival information. Afterwards, weighted Gene Co-expression Network Analysis was performed to construct a network between gene expression, inflammation response, and hypoxia. Subsequently, univariate Cox, random forest, and multivariate Cox regressions were applied to develop a robust inflammation and hypoxia-related gene signature for predicting clinical outcomes in patients with IPF. Furthermore, a nomogram was constructed to calculate risk assessment. The inflammation response and hypoxia were identified as latent risk factors for patients with IPF. Five genes, including HS3ST1, WFDC2, SPP1, TFPI, and CDC42EP2, were identified that formed the inflammation-hypoxia-related gene signature. Kaplan-Meier plotter showed that the patients with high-risk scores had a worse prognosis than those with low-risk scores in training and validation cohorts. The time-dependent concordance index and the receiver operating characteristic analysis revealed that the risk model could accurately predict the clinical outcome of patients with IPF. Therefore, this study contributes to elucidating the role of inflammation and hypoxia in IPF, which can aid in assessing individual prognosis and personalised treatment decisions.
Collapse
Affiliation(s)
- Jun Liu
- Reproductive Medicine Center, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Medical Research Center, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Wenli Li
- Reproductive Medicine Center, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, China
| |
Collapse
|
19
|
Liu L, Yu N, Leng W, Lu Y, Xia X, Yuan H. 6-Gingerol, a functional polyphenol of ginger, reduces pulmonary fibrosis by activating Sirtuin1. Allergol Immunopathol (Madr) 2022; 50:104-114. [PMID: 35257553 DOI: 10.15586/aei.v50i2.533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/07/2021] [Indexed: 11/18/2022]
Abstract
Pulmonary fibrosis in general is the final common outcome of various interstitial lung diseases. In recent years, the incidence of pulmonary fibrosis has been rising with poor prognosis. 6-gingerol is deemed as a functional polyphenol of ginger. The aim of the present study was to investigate the effect of 6-gingerol, on pulmonary fibrosis. Mice were randomly divided into four groups: control, bleomycin, bleomycin + 6-gingerol 100 mg/kg, bleomycin + 6-gingerol 250 mg/kg, and the survival rates of the groups were recorded. Pathological and fibrotic changes in the lungs were identified by H&E and Masson staining, respectively. The levels of hydroxyproline and protein deposited in lung tissues were then, respectively, determined by colorimetry and western blotting. Subsequently, the proportion of cells and inflammatory factors in the alveolar lavage fluid were estimated. Following the identification of the possibility of Sirtuin1 (SIRT1) in the pharmacological mechanism through molecular docking and western blotting, human embryonic lung fibroblasts MRC-5 were treated with TGF-β1 and SIRT1 inhibitor to study the role of SIRT1 in the regulatory effect of 6-gingerol. From the results, 6-gingerol was found to increase the survival rate of mice and reduce lung pathology and fibrosis in mice. And, it significantly reduced the levels of hydroxyproline and the proteins deposited in lung tissues. Moreover, the number of neutrophils, basophils, monocytes, and the levels of inflammatory factors in the alveolar lavage fluid were also reduced. SIRT1 inhibitor blocked the function of 6-gingerol to inhibit fibrosis. To sum up, 6-gingerol relieves pulmonary fibrosis via activating SIRT1. This finding expands the pharmacological effect of 6-gingerol, and it is expected to advance the development of treatments for pulmonary fibrosis.
Collapse
Affiliation(s)
- Li Liu
- The Affiliation Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nan Yu
- The Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Wei Leng
- The Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Yun Lu
- The Affiliation Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinxin Xia
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Yuan
- The Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xianyang, China;
| |
Collapse
|
20
|
Li XZ, Wang XL, Wang YJ, Liang QK, Li Y, Chen YW, Ming HX. Total flavonoids of Oxytropis falcata Bunge have a positive effect on idiopathic pulmonary fibrosis by inhibiting the TGF-β1/Smad signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 285:114858. [PMID: 34826543 DOI: 10.1016/j.jep.2021.114858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease with unknown etiology. Oxytropis falcata Bunge (O. falcata) is a 1-35 cm high perennial clustered herb, also known as edaxia, has viscosity and a special smell, and is mainly distributed in the western areas of China. The root of O. falcata has a diameter of 6 mm, is straight and deep, dark red and its stems are shortened, woody and multibranched. O. falcata has heat-clearing, detoxification, analgesic, anti-inflammatory, antibacterial, hemostatic and antitumor activities. Furthermore, O. falcata has excellent anti-inflammatory and analgesic effects, and it is one of the three major anti-inflammatory drugs in Tibetan medicine, known as "the king of herbs". Total flavonoids of Oxytropis falcata Bunge (FOFB) were previously extracted, and their pharmacological activities are consistent with those of the whole herb. In this study, FOFB was extracted from O. falcata by ethanol extraction, and the mechanism of FOFB on IPF was verified by in vivo and in vitro experiments. AIM OF THE STUDY In this study, we aimed to observe the effects of FOFB on idiopathic pulmonary fibrosis. MATERIALS AND METHODS In in vivo experiments, an IPF rat model was established by bleomycin induction. The rats were treated with FOFB (100, 200, 400 mg kg-1·d-1) for 4 weeks. Masson staining and the expression of TGF-β, p-Smad2, p-Smad3 and Smad7 in the lung tissue of rats were detected. In in vitro experiments, we perfused normal rats with FOFB (100, 200, 400 mg kg-1·d-1) and obtained the corresponding drug-containing serum. The HFL-1 cell model induced by TGF-β1 was used to detect the corresponding indices through intervention with drug-containing serum. The best intervention time for drug-containing serum was detected by the CCK-8 method. Changes in apoptosis, cytoskeleton and rough endoplasmic reticulum structure were detected. Finally, the expression of TGF-β, p-Smad2, p-Smad3 and Smad7 in cells was examined. RESULTS In vivo, Masson staining indicated that the degree of pulmonary fibrosis increased significantly, the expression of TGF-β, p-smad2 and p-Smad3 increased significantly, and the expression of Smad7 decreased in the model group. We found that the degree of pulmonary fibrosis gradually decreased and that the inhibition of the TGF-β/Smad signaling pathway became more obvious with increasing FOFB dose. FOFB (400 mg kg-1·d-1) significantly improved the degree of pulmonary fibrosis in rats. In in vitro experiments, the CCK-8 results showed that 120 h was the best intervention time for drug-containing serum. In the model group, there was no obvious apoptosis or changes in microfilaments and microtubules, the number of rough endoplasmic reticulum increased, and the expression of TGF-β, p-Smad2 and p-Smad3 increased significantly, while the expression of Smad7 decreased significantly. We found that with the increase in drug-containing serum concentration, the apoptosis, cytoskeleton and degree of destruction of the rough endoplasmic reticulum in the HFL-1 cell model also increased, and the inhibition of the TGF-β/Smad signaling pathway became more pronounced; the effect of the drug-containing serum administered with FOFB (400 mg kg-1·d-1) was the most significant. CONCLUSIONS The results suggest that FOFB can improve the occurrence and development of IPF. The effect of FOFB on IPF may be mediated by inhibition of the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xin-Ze Li
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Basic Subjects of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu, Lanzhou, 730000, China; Institute of Integrative Medicine with Gansu University of Traditional Chinese Medicine, Gansu, Lanzhou, 730000, China
| | - Xue-Lin Wang
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Shanxi, Xianyang, 712000, China
| | - Yan-Jun Wang
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Basic Subjects of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu, Lanzhou, 730000, China; Institute of Integrative Medicine with Gansu University of Traditional Chinese Medicine, Gansu, Lanzhou, 730000, China
| | - Qian-Kun Liang
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China
| | - Yang Li
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China
| | - Yan-Wen Chen
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China
| | - Hai-Xia Ming
- Basic Medical College, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Basic Subjects of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Gansu, Lanzhou, 730000, China; Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities, Gansu, Lanzhou, 730000, China.
| |
Collapse
|
21
|
He J, Li X. Identification and Validation of Aging-Related Genes in Idiopathic Pulmonary Fibrosis. Front Genet 2022; 13:780010. [PMID: 35211155 PMCID: PMC8863089 DOI: 10.3389/fgene.2022.780010] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Aging plays a significant role in the occurrence and development of idiopathic pulmonary fibrosis (IPF). In this study, we aimed to identify and verify potential aging-associated genes involved in IPF using bioinformatic analysis. The mRNA expression profile dataset GSE150910 available in the Gene Expression Omnibus (GEO) database and R software were used to identify the differentially expressed aging-related genes involved in IPF. Hub gene expression was validated by other GEO datasets. Gene ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were performed on differentially expressed aging-related genes. Subsequently, aging-related genes were further screened using three techniques (least absolute shrinkage and selection operator (LASSO) regression, support vector machine, and random forest), and the receiver operating characteristic curves were plotted based on screening results. Finally, real-time quantitative polymerase chain reaction (qRT-PCR) was performed to verify the RNA expression of the six differentially expressed aging-related genes using the blood samples of patients with IPF and healthy individuals. Sixteen differentially expressed aging-related genes were detected, of which the expression of 12 were upregulated and four were downregulated. GO and KEGG enrichment analyses indicated the presence of several enriched terms related to senescence and apoptotic mitochondrial changes. Further screening by LASSO regression, support vector machine, and random forest identified six genes (IGF1, RET, IGFBP2, CDKN2A, JUN, and TFAP2A) that could serve as potential diagnostic biomarkers for IPF. Furthermore, qRT-PCR analysis indicated that among the above-mentioned six aging-related genes, only the expression levels of IGF1, RET, and IGFBP2 in patients with IPF and healthy individuals were consistent with the results of bioinformatic analysis. In conclusion, bioinformatics analysis identified 16 potential aging-related genes associated with IPF, and clinical sample validation suggested that among these, IGF1, RET, and IGFBP2 might play a role in the incidence and prognosis of IPF. Our findings may help understand the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jie He
- Clinical Medical College of Chengdu Medical College, Chengdu, China.,Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoyan Li
- Clinical Medical College of Chengdu Medical College, Chengdu, China.,Department of Endocrinology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
22
|
The Extent of Honeycombing on Computed Tomography Cannot Predict the Treatment Outcome of Patients with Acute Exacerbations of Interstitial Lung Disease. Can Respir J 2021; 2021:7456315. [PMID: 34824651 PMCID: PMC8610694 DOI: 10.1155/2021/7456315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/23/2021] [Accepted: 10/23/2021] [Indexed: 11/17/2022] Open
Abstract
Background The purpose of this retrospective study was to clarify whether the presence of honeycombing on computed tomography (CT) can affect the prognosis of patients with acute exacerbations (AEs) of interstitial lung diseases (ILDs). Methods Clinical parameters including age, sex, Charlson Comorbidity Index Score (CCIS), blood biomarkers, and 3-month mortality were retrospectively compared between the CT honeycombing present and absent groups at the diagnosis of AEs of ILDs. Results Ninety-five patients who were on corticosteroid pulse therapy were assessed. Though log-rank tests showed that Kaplan–Meier survival curves of the high and low ground-glass opacity (GGO) score groups differed significantly in 3-month mortality in patients with AEs of idiopathic ILDs (P = 0.007) and overall patients (P = 0.045), there was no significant difference between the CT honeycombing present and absent groups in patients with AEs of idiopathic ILDs (P = 0.472) and AEs of secondary ILDs (P = 0.905), as well as of overall patients (P = 0.600). In addition, whereas CCIS (OR, 1.436; 95% CI, 1.156–1.842; P < 0.001) was a significant predictor of 3-month mortality in the CT honeycombing absent group, serum LDH (OR, 1.005; 95% CI, 1.002–1.007; P = 0.001) was a significant predictor in the CT honeycombing present group. Conclusions The clinical features of patients with or without honeycombing may differ due to the difference in prognostic factors, but these groups were found to have similar prognoses 3 months after AE onset, and clinicopathological examinations according to these groups are essential.
Collapse
|
23
|
Abstract
Purpose of Review With the projected increase in the geriatric patient population, it is of the utmost importance to understand and optimize conditions in the perioperative period to ensure the best surgical outcome. Age-associated changes in respiratory physiology affect the surgical management of geriatric patients. This review focuses on perioperative pulmonary management of elderly individuals. Recent Findings The physiological changes associated with aging include both physical and biochemical alterations that are detrimental to pulmonary function. There is an increased prevalence of chronic lung disease such as COPD and interstitial lung disease which can predispose patients to postoperative pulmonary complications. Additionally, elderly patients, especially those with chronic lung disease, are at risk for frailty. Screening tools have been developed to evaluate risk and aid in the judicious selection of patients for surgical procedures. The concept of "prehabilitation" has been developed to best prepare patients for surgery and may be more influential in the reduction of postoperative pulmonary complications than postoperative rehabilitation. Understanding the age-associated changes in metabolism of drugs has led to dose adjustments in the intraoperative and postoperative periods, reducing respiratory depression and lung protective ventilation and minimally invasive procedures have yielded reductions in postoperative pulmonary complications. Summary The perioperative management of the geriatric population can be divided into three key areas: preoperative risk mitigation, intraoperative considerations, and postoperative management. Preoperative considerations include patient selection and thorough history and physical, along with smoking cessation and prehabilitation in a subset of patients. Operative aspects include careful selection of anesthetic agents, lung protective ventilation, and choice of surgical procedure. Postoperative management should focus on selective use of agents that may contribute to respiratory depression and encouragement of rehabilitation.
Collapse
Affiliation(s)
- Catherine Entriken
- Section of General Surgery, Department of Surgery, University of Cincinnati, Cincinnati, OH USA
| | - Timothy A. Pritts
- Section of General Surgery, Department of Surgery, University of Cincinnati, Cincinnati, OH USA
| |
Collapse
|
24
|
Galioto F, Palmucci S, Astuti GM, Vancheri A, Distefano G, Tiralongo F, Libra A, Cusumano G, Basile A, Vancheri C. Complications in Idiopathic Pulmonary Fibrosis: Focus on Their Clinical and Radiological Features. Diagnostics (Basel) 2020; 10:diagnostics10070450. [PMID: 32635390 PMCID: PMC7399856 DOI: 10.3390/diagnostics10070450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/09/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrotic lung disease with uncertain origins and pathogenesis; it represents the most common interstitial lung disease (ILD), associated with a pathological pattern of usual interstitial pneumonitis (UIP). This disease has a poor prognosis, having the most lethal prognosis among ILDs. In fact, the progressive fibrosis related to IPF could lead to the development of complications, such as acute exacerbation, lung cancer, infections, pneumothorax and pulmonary hypertension. Pneumologists, radiologists and pathologists play a key role in the identification of IPF disease, and in the characterization of its complications-which unfortunately increase disease mortality and reduce overall survival. The early identification of these complications is very important, and requires an integrated approach among specialists, in order to plane the correct treatment. In some cases, the degree of severity of patients having IPF complications may require a personalized approach, based on palliative care services. Therefore, in this paper, we have focused on clinical and radiological features of the complications that occurred in our IPF patients, providing a comprehensive and accurate pictorial essay for clinicians, radiologists and surgeons involved in their management.
Collapse
Affiliation(s)
- Federica Galioto
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
| | - Stefano Palmucci
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
- Correspondence: ; Tel.: +39-347-833-0775
| | - Giovanna M. Astuti
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
| | - Ada Vancheri
- Regional Centre for Interstitial and Rare Lung Disease, Department of Clinical and Molecular Biomedicine, University of Catania, 95123 Catania, Italy; (A.V.); (A.L.); (G.C.); (C.V.)
| | - Giulio Distefano
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
| | - Francesco Tiralongo
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
| | - Alessandro Libra
- Regional Centre for Interstitial and Rare Lung Disease, Department of Clinical and Molecular Biomedicine, University of Catania, 95123 Catania, Italy; (A.V.); (A.L.); (G.C.); (C.V.)
| | - Giacomo Cusumano
- Regional Centre for Interstitial and Rare Lung Disease, Department of Clinical and Molecular Biomedicine, University of Catania, 95123 Catania, Italy; (A.V.); (A.L.); (G.C.); (C.V.)
| | - Antonio Basile
- Radiology Unit 1, Department of Medical Surgical Sciences and Advanced Technologies—University Hospital “Policlinico-Vittorio Emanuele”, University of Catania, Via Santa Sofia n. 78, 95123 Catania, Italy; (F.G.); (G.M.A.); (G.D.); (F.T.); (A.B.)
| | - Carlo Vancheri
- Regional Centre for Interstitial and Rare Lung Disease, Department of Clinical and Molecular Biomedicine, University of Catania, 95123 Catania, Italy; (A.V.); (A.L.); (G.C.); (C.V.)
| |
Collapse
|