1
|
Xu HP, Niu H, Wang H, Lin J, Yao JJ. Knockdown of RTEL1 Alleviates Chronic Obstructive Pulmonary Disease by Modulating M1, M2 Macrophage Polarization and Inflammation. COPD 2024; 21:2316607. [PMID: 38420994 DOI: 10.1080/15412555.2024.2316607] [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: 10/29/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic disease characterized by airflow obstruction, which seriously threatens people's health. The COPD mouse model was established with cigarette smoke induction. Hematoxylin-eosin staining and Masson staining were carried out to observe the pathological changes of lung tissues in COPD mice. RTEL1 was silenced in COPD mice, and immunohistochemistry was used to detect RTEL1, ki67 and Caspase-3 expression. The role of RTEL1 in inflammation were evaluated by ELISA, and the impacts of RTEL1 on M1 and M2 macrophage markers (iNOS and CD206) were evaluated by qPCR and western blotting. In COPD model, there was an increase in the number of inflammatory cells, with slightly disorganized cell arrangement, unclear hierarchy, condensed and solidified nuclei, while knockdown of RTEL1 improved the inflammatory infiltration. Moreover, knockdown of RTEL1 reduced ki67-positive cells and increased Caspase-3 positive cells in COPD group. The increased inflammatory factors (IL-1β, MMP-9, TNF-α, IL-4, IL-6, and IL-23) in COPD were suppressed by knockdown of RTEL1, while iNOS was raised and CD206 was inhibited. In conclusion, knockdown of RTEL1 promoted M1 and inhibited M2 macrophage polarization and inflammation to alleviate COPD.
Collapse
Affiliation(s)
- He-Ping Xu
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Huan Niu
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Hong Wang
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jie Lin
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jin-Jian Yao
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
2
|
Mao Y, Suryawanshi A, Patial S, Saini Y. Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Front Pharmacol 2024; 15:1460692. [PMID: 39386033 PMCID: PMC11461968 DOI: 10.3389/fphar.2024.1460692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Exosomes, membrane-bound extracellular vesicles, ranging from approximately 30-200 nm in diameter, are released by almost all cell types and play critical roles in intercellular communication. In response to the prevailing stress, the exosome-bound protein signatures vary in abundance and composition. To identify the bronchoalveolar lavage fluid (BALF) exosome-bound proteins associated with mucoinflammatory lung disease and to gain insights into their functional implications, we compared BALF exosomes-derived proteins from adult Scnn1b transgenic (Scnn1b-Tg+) and wild type (WT) mice. A total of 3,144 and 3,119 proteins were identified in BALF exosomes from Scnn1b-Tg+ and WT mice, respectively. Using cutoff criteria (Log2 fold-change > 1 and adjusted p-value < 0.05), the comparison of identified proteins revealed 127 and 30 proteins that were significantly upregulated and downregulated, respectively, in Scnn1b-Tg+ versus WT mice. In addition, 52 and 27 proteins were exclusively enriched in Scnn1b-Tg+ and WT mice, respectively. The identified exosome-bound proteins from the homeostatic airspaces of WT mice were mostly relevant to the normal physiological processes. The protein signatures enriched in the BALF exosomes of Scnn1b-Tg+ mice were relevant to macrophage activation and mucoinflammatory processes. Ingenuity pathway analyses revealed that the enriched proteins in Scnn1b-Tg+ mice contributed to the inflammatory responses and antimicrobial defense pathways. Selective proteins including, RETNLA/FIZZ1, LGALS3/Galectin-3, S100A8/MRP8, and CHIL3/YM1 were immunolocalized to specific cell types. The comparative analysis between enriched BALF exosome proteins and previously identified differentially upregulated genes in Scnn1b-Tg+ versus WT mice suggested that the compartment-/cell-specific upregulation in gene expression dictates the enrichment of their respective proteins in the lung airspaces. Taken together, this study demonstrates that the BALF exosome-bound protein signatures reflect disease-relevant disturbances. Our findings suggest that the exosomes carry disease-relevant protein signatures that can be used as a diagnostic as well as predictive biomarkers for mucoinflammatory lung disease.
Collapse
Affiliation(s)
- Yun Mao
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sonika Patial
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
3
|
Baltazar-García EA, Vargas-Guerrero B, Gasca-Lozano LE, Gurrola-Díaz CM. Molecular changes underlying pulmonary emphysema and chronic bronchitis in Chronic Obstructive Pulmonary Disease: An updated review. Histol Histopathol 2024; 39:805-816. [PMID: 38226432 DOI: 10.14670/hh-18-699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The aim of this review is to update and synthesize the molecular mechanisms that lead to the heterogeneous effect on tissue remodeling observed in the two most important clinical phenotypes of chronic obstructive pulmonary disease (COPD), pulmonary emphysema (PE) and chronic bronchitis (CB). Clinical and experimental evidence suggests that this heterogeneous response to promote PE, CB, or both, is related to differentiated genetic, epigenetic, and molecular conditions. Specifically, a tendency toward PE could be related to a variant in the DSP gene, SIRT1 downregulation, macrophage polarization to M1, as well as the involvement of the noncanonical Wnt5A signaling pathway, among other alterations. Additionally, in advanced stages of COPD, PE development is potentiated by dysregulations in autophagy, which promotes senescence and subsequently cell apoptosis, through exacerbated inflammasome activation and release of caspases. On the other hand, CB or the pro-fibrotic phenotype could be potentiated by the downregulated activity of HDAC2, the activation of the TGF-β/Smad or Wnt/β-catenin signaling pathways, macrophage polarization to M2, upregulation of TIMP-1, and/or the presence of the epithelial-mesenchymal transition (EMT) mechanism. Interestingly, the upregulated activity of MMPs, especially MMP-9, is widely involved in the development of both phenotypes. Furthermore, MMP-9 and MMP-12 enhance the severity, perpetuation, and exacerbation of COPD, as well as the development of autoimmunity in this disease.
Collapse
Affiliation(s)
- Elia A Baltazar-García
- Transdisciplinary Institute for Research and Innovation in Health Sciences/Institute for Research in Chronic-Degenerative Diseases, Department of Molecular Biology and Genomics, University Campus for Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Belinda Vargas-Guerrero
- Transdisciplinary Institute for Research and Innovation in Health Sciences/Institute for Research in Chronic-Degenerative Diseases, Department of Molecular Biology and Genomics, University Campus for Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Luz E Gasca-Lozano
- Transdisciplinary Institute for Research and Innovation in Health Sciences/Institute for Research in Chronic-Degenerative Diseases, Department of Molecular Biology and Genomics, University Campus for Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Carmen M Gurrola-Díaz
- Transdisciplinary Institute for Research and Innovation in Health Sciences/Institute for Research in Chronic-Degenerative Diseases, Department of Molecular Biology and Genomics, University Campus for Health Sciences, University of Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
4
|
Baltazar-García EA, Vargas-Guerrero B, Lima A, Boavida Ferreira R, Mendoza-Magaña ML, Ramírez-Herrera MA, Baltazar-Díaz TA, Domínguez-Rosales JA, Salazar-Montes AM, Gurrola-Díaz CM. Deflamin Attenuated Lung Tissue Damage in an Ozone-Induced COPD Murine Model by Regulating MMP-9 Catalytic Activity. Int J Mol Sci 2024; 25:5063. [PMID: 38791100 PMCID: PMC11121448 DOI: 10.3390/ijms25105063] [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: 03/28/2024] [Revised: 04/26/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is comprised of histopathological alterations such as pulmonary emphysema and peribronchial fibrosis. Matrix metalloproteinase 9 (MMP-9) is one of the key enzymes involved in both types of tissue remodeling during the development of lung damage. In recent studies, it was demonstrated that deflamin, a protein component extracted from Lupinus albus, markedly inhibits the catalytic activity of MMP-9 in experimental models of colon adenocarcinoma and ulcerative colitis. Therefore, in the present study, we investigated for the first time the biological effect of deflamin in a murine COPD model induced by chronic exposure to ozone. Ozone exposure was carried out in C57BL/6 mice twice a week for six weeks for 3 h each time, and the treated group was orally administered deflamin (20 mg/kg body weight) after each ozone exposure. The histological results showed that deflamin attenuated pulmonary emphysema and peribronchial fibrosis, as evidenced by H&E and Masson's trichrome staining. Furthermore, deflamin administration significantly decreased MMP-9 activity, as assessed by fluorogenic substrate assay and gelatin zymography. Interestingly, bioinformatic analysis reveals a plausible interaction between deflamin and MMP-9. Collectively, our findings demonstrate the therapeutic potential of deflamin in a COPD murine model, and suggest that the attenuation of the development of lung tissue damage occurs by deflamin-regulated MMP-9 catalytic activity.
Collapse
Affiliation(s)
- Elia Ana Baltazar-García
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Belinda Vargas-Guerrero
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Ana Lima
- CECAV—Centro de Ciência Animal e Veterinária, Faculty of Veterinary Medicine, Lusófona University, Campo Grande, 376, 1749-024 Lisbon, Portugal;
| | - Ricardo Boavida Ferreira
- LEAF—Landscape Environment Agriculture and Food, Instituto Superior de Agronomia, Universidade de Lisboa, 1349-017 Lisbon, Portugal;
| | - María Luisa Mendoza-Magaña
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (M.L.M.-M.); (M.A.R.-H.)
| | - Mario Alberto Ramírez-Herrera
- Laboratorio de Neurofisiología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (M.L.M.-M.); (M.A.R.-H.)
| | - Tonatiuh Abimael Baltazar-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - José Alfredo Domínguez-Rosales
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Adriana María Salazar-Montes
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| | - Carmen Magdalena Gurrola-Díaz
- Instituto de Investigación en Enfermedades Crónico-Degenerativas, Instituto Transdisciplinar de Investigación e Innovación en Salud, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Sierra Mojada 950, Puerta peatonal 7, Col. Independencia, Guadalajara 44350, Jalisco, Mexico; (E.A.B.-G.); (B.V.-G.); (T.A.B.-D.); (J.A.D.-R.); (A.M.S.-M.)
| |
Collapse
|
5
|
Yoon EC, Koo SM, Park HY, Kim HC, Kim WJ, Kim KU, Jung KS, Yoo KH, Yoon HK, Yoon HY. Predictive Role of White Blood Cell Differential Count for the Development of Acute Exacerbation in Korean Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:17-31. [PMID: 38192972 PMCID: PMC10773455 DOI: 10.2147/copd.s435921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/15/2023] [Indexed: 01/10/2024] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a respiratory disease characterized by chronic inflammation. Acute exacerbation of COPD (AECOPD) manifests as acute worsening of respiratory symptoms and is associated with high morbidity and mortality. The aim of the present study was to evaluate the predictive value of white blood count (WBC) and its derived inflammatory biomarkers for AECOPD. Methods From the Korean COPD Subgroup Study cohort, a prospective and multicenter observational study, 826 patients who had baseline complete blood count (CBC) and 3-year AECOPD data were included. Follow-up CBC data at 1 (n = 385), 2 (n = 294), and 3 (n = 231) years were collected for available patients. The primary outcome was the occurrence of AECOPD at 3 years. The risk of AECOPD was evaluated using a binary logistic analysis. Results The cumulative incidences of 12-, 24-, and 36-month AECOPD were 47.6%, 60.5%, and 67.6%, respectively. Patients with AECOPD at 3 years had higher baseline WBC counts, neutrophil counts, neutrophil/lymphocyte ratio (NLR), and neutrophil/monocyte ratio than those without AECOPD. Higher WBC count, neutrophil count, and NLR were associated with the 3-year occurrence of AECOPD in the univariate analysis, but only the higher neutrophil count was a significant risk factor (odds ratio [OR] = 1.468; 95% confidence interval [CI]: 1.024-2.104) in the covariates-adjusted analysis. In the analysis of changes in inflammatory parameters, a decrease in the platelet count (OR = 0.502; 95% CI: 0.280-0.902) and NLR (OR = 0.535; 95% CI: 0.294-0.974) at 2 years and an increase in the eosinophil count (OR = 2.130; 95% CI: 1.027-4.416) at 3 years were significantly associated with AECOPD in the adjusted analysis. Conclusion Our data suggest that a high baseline WBC count, particularly neutrophil count, was associated with a higher incidence of long-term AECOPD.
Collapse
Affiliation(s)
- Eun Chong Yoon
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - So-My Koo
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hye Yun Park
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ho Cheol Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University School of Medicine, Changwon, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, Kangwon National University, Chuncheon, Republic of Korea
| | - Ki Uk Kim
- Department of Internal Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Ki-Suck Jung
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Kwang Ha Yoo
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hyoung Kyu Yoon
- Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee-Young Yoon
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
6
|
Papakonstantinou E, Christopoulou ME, Karakioulaki M, Grize L, Tamm M, Stolz D. Ηeparan sulphate in infectious and non-infectious exacerbations of COPD. Respirology 2023. [PMID: 37311657 DOI: 10.1111/resp.14531] [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: 11/16/2022] [Accepted: 05/24/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE Acute exacerbations of chronic obstructive pulmonary disease (AECOPD) are associated with worsening health outcomes and effective treatment of each episode is essential. In this study, we aimed to investigate if plasma levels of heparan sulphate (HS) are associated with the aetiology of AECOPD. METHODS COPD patients (N = 1189), GOLD grade II-IV, from a discovery cohort (N = 638) and from a validation cohort (N = 551), were included in the study. HS and heparanase (HSPE-1) were measured longitudinally in plasma at stable state, at AECOPD and at 4 weeks follow-up. RESULTS Plasma HS was higher in patients with COPD as compared with non-COPD controls and was significantly increased at AECOPD as compared to stable state (p < 0.001) in the discovery and in the validation cohorts. Four distinct exacerbation groups were classified based on aetiology (no-infection/bacterial-infection/viral-infection/bacterial and viral coinfection) in the validation cohort. The fold-increase of HS from stable state to AECOPD was associated with the aetiology of exacerbation and was higher in cases with bacterial and viral coinfections. HSPE-1 was also significantly increased at AECOPD, however, there was no association of HSPE-1 levels with the aetiology of these events. The probability of having an infection at AECOPD was raised as HS levels increased from stable state to AECOPD. This probability was higher for bacterial infections than viral infections. CONCLUSION The results of our study indicate that circulating levels of HS are increased at AECOPD and this increase may be associated with the aetiology of these events.
Collapse
Affiliation(s)
- Eleni Papakonstantinou
- Clinic of Respiratory Medicine and Pulmonary cell Research, University Hospital, Basel, Switzerland
- Department of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria-Elpida Christopoulou
- Department of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Meropi Karakioulaki
- Clinic of Respiratory Medicine and Pulmonary cell Research, University Hospital, Basel, Switzerland
| | - Leticia Grize
- Clinic of Respiratory Medicine and Pulmonary cell Research, University Hospital, Basel, Switzerland
| | - Michael Tamm
- Clinic of Respiratory Medicine and Pulmonary cell Research, University Hospital, Basel, Switzerland
| | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary cell Research, University Hospital, Basel, Switzerland
- Department of Pneumology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Stolz D, Matera MG, Rogliani P, van den Berge M, Papakonstantinou E, Gosens R, Singh D, Hanania N, Cazzola M, Maitland-van der Zee AH, Fregonese L, Mathioudakis AG, Vestbo J, Rukhadze M, Page CP. Current and future developments in the pharmacology of asthma and COPD: ERS seminar, Naples 2022. Breathe (Sheff) 2023; 19:220267. [PMID: 37377851 PMCID: PMC10292790 DOI: 10.1183/20734735.0267-2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/28/2023] [Indexed: 06/29/2023] Open
Abstract
Pharmacological management of airway obstructive diseases is a fast-evolving field. Several advances in unravelling disease mechanisms as well as intracellular and molecular pathways of drug action have been accomplished. While the clinical translation and implementation of in vitro results to the bedside remains challenging, advances in comprehending the mechanisms of respiratory medication are expected to assist clinicians and scientists in identifying meaningful read-outs and designing clinical studies. This European Respiratory Society Research Seminar, held in Naples, Italy, 5-6 May 2022, focused on current and future developments of the drugs used to treat asthma and COPD; on mechanisms of drug action, steroid resistance, comorbidities and drug interactions; on prognostic and therapeutic biomarkers; on developing novel drug targets based on tissue remodelling and regeneration; and on pharmacogenomics and emerging biosimilars. Related European Medicines Agency regulations are also discussed, as well as the seminar's position on the above aspects.
Collapse
Affiliation(s)
- Daiana Stolz
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, and Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine, Department of Internal Medicine, Medical Center University of Freiburg, Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital of Basel, Basel, Switzerland
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD, and Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, University of Manchester, Manchester, UK
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Nicola Hanania
- Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mario Cazzola
- Unit of Respiratory Medicine, Department Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | | | | | - Alexander G. Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | - Maia Rukhadze
- Center of Allergy and Immunology, Teaching University Geomedi LLC, Tbilisi, Georgia
| | - Clive P. Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| |
Collapse
|
8
|
Christopoulou ME, Papakonstantinou E, Stolz D. Matrix Metalloproteinases in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2023; 24:ijms24043786. [PMID: 36835197 PMCID: PMC9966421 DOI: 10.3390/ijms24043786] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/01/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes that degrade proteins of the extracellular matrix and the basement membrane. Thus, these enzymes regulate airway remodeling, which is a major pathological feature of chronic obstructive pulmonary disease (COPD). Furthermore, proteolytic destruction in the lungs may lead to loss of elastin and the development of emphysema, which is associated with poor lung function in COPD patients. In this literature review, we describe and appraise evidence from the recent literature regarding the role of different MMPs in COPD, as well as how their activity is regulated by specific tissue inhibitors. Considering the importance of MMPs in COPD pathogenesis, we also discuss MMPs as potential targets for therapeutic intervention in COPD and present evidence from recent clinical trials in this regard.
Collapse
Affiliation(s)
- Maria-Elpida Christopoulou
- Department of Pneumology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Eleni Papakonstantinou
- Department of Pneumology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital, 4031 Basel, Switzerland
| | - Daiana Stolz
- Department of Pneumology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital, 4031 Basel, Switzerland
- Correspondence: ; Tel.: +49-(0)-761-270-37050
| |
Collapse
|
9
|
Lu L, Peng J, Wu F, Yang H, Zheng Y, Deng Z, Zhao N, Dai C, Xiao S, Wen X, Xu J, Wu X, Zhou K, Ran P, Zhou Y. Clinical characteristics of airway impairment assessed by impulse oscillometry in patients with chronic obstructive pulmonary disease: findings from the ECOPD study in China. BMC Pulm Med 2023; 23:52. [PMID: 36737731 PMCID: PMC9896683 DOI: 10.1186/s12890-023-02311-z] [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: 09/07/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The role of airway impairment assessed by impulse oscillometry (IOS) in patients with chronic obstructive pulmonary disease (COPD) remains unclear. Therefore, this study aimed to analyze the proportion and clinical characteristics of airway impairment assessed by IOS across COPD severities, and explore whether airway impairment is a subtype of COPD. METHODS This study was based on cross-sectional data from the ECOPD cohort in Guangdong, China. Subjects were consecutively recruited from July 2019 to August 2021. They filled out questionnaires and underwent lung function tests, IOS and computed tomography (CT). COPD was defined as post-bronchodilator forced expiratory volume in one second/forced vital capacity < lower limit of normal (LLN). Meanwhile, airway impairment was defined as IOS parameters > upper limit of normal or < LLN. On the one hand, Poisson regression was employed to analyze the associations between acute exacerbations of COPD (AECOPD) in the previous year and airway impairment. On the other hand, logistic regression was used to assess differences in CT imaging between patients with IOS parameters' abnormalities and patients with normal IOS parameters. RESULTS 768 COPD subjects were finally enrolled in the study. The proportion of airway impairment assessed by R5, R20, R5-R20, X5, AX, and Fres was 59.8%, 29.7%, 62.5%, 52.9%, 60.9% and 67.3%, respectively. Airway impairment assessed by IOS parameters (R5, R5-R20, X5, AX, and Fres) in patients with COPD was present across all severities of COPD, particularly in GOLD 3-4 patients. Compared with patients with normal IOS parameters, patients with IOS parameters' abnormalities had more respiratory symptoms, more severe airway obstruction and imaging structural abnormalities. Patients with IOS parameters' abnormalities assessed by R5 [risk ratio (RR): 1.58, 95% confidential interval (CI): 1.13-2.19, P = 0.007], R5-R20 [RR: 1.73, 95%CI: 1.22-2.45, P = 0.002], X5 [RR: 2.11, 95%CI: 1.51-2.95, P < 0.001], AX [RR: 2.20, 95%CI: 1.53-3.16, P < 0.001], and Fres [RR: 2.13, 95%CI: 1.44-3.15, P < 0.001] had a higher risk of AECOPD in the previous year than patients with normal IOS parameters. CONCLUSIONS Airway impairment assessed by IOS may be a subtype of COPD. Future studies are warranted to identify the underlying mechanisms and longitudinal progression of airway impairment.
Collapse
Affiliation(s)
- Lifei Lu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Jieqi Peng
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China ,Guangzhou Laboratory, Guangzhou, China
| | - Fan Wu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China ,Guangzhou Laboratory, Guangzhou, China
| | - Huajing Yang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Youlan Zheng
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Zhishan Deng
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Ningning Zhao
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Cuiqiong Dai
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Shan Xiao
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Xiang Wen
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Jianwu Xu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Xiaohui Wu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Kunning Zhou
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China
| | - Pixin Ran
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China ,Guangzhou Laboratory, Guangzhou, China
| | - Yumin Zhou
- grid.470124.4State Key Laboratory of Respiratory Disease, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, China ,Guangzhou Laboratory, Guangzhou, China
| |
Collapse
|
10
|
Usmani OS, Dhand R, Lavorini F, Price D. Why We Should Target Small Airways Disease in Our Management of Chronic Obstructive Pulmonary Disease. Mayo Clin Proc 2021; 96:2448-2463. [PMID: 34183115 DOI: 10.1016/j.mayocp.2021.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/12/2021] [Accepted: 03/16/2021] [Indexed: 12/23/2022]
Abstract
For more than 50 years, small airways disease has been considered a key feature of chronic obstructive pulmonary disease (COPD) and a major cause of airway obstruction. Both preventable and treatable, small airways disease has important clinical consequences if left unchecked. Small airways disease is associated with poor spirometry results, increased lung hyperinflation, and poor health status, making the small airways an important treatment target in COPD. The early detection of small airways disease remains the key barrier; if detected early, treatments designed to target small airways may help reduce symptoms and allow patients to maintain their activities. Studies are needed to evaluate the possible role of new drugs and novel drug formulations, inhalers, and inhalation devices for treating small airways disease. These developments will help to improve our management of small airways disease in patients with COPD.
Collapse
Affiliation(s)
- Omar S Usmani
- National Heart and Lung Institute, Imperial College London, and Royal Brompton Hospital, Airways Disease Section, London, UK.
| | - Rajiv Dhand
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville
| | - Federico Lavorini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - David Price
- Observational and Pragmatic Research Institute, Singapore; Optimum Patient Care, Cambridge, UK; Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
11
|
Increased Serum Levels of Matrix-metalloproteinase-9, Cyclo-oxygenase-2 and Prostaglandin E-2 in Patients with Chronic Obstructive Pulmonary Disease (COPD). Indian J Clin Biochem 2021; 37:169-177. [PMID: 35463116 PMCID: PMC8994009 DOI: 10.1007/s12291-021-00973-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD), a heterogeneous lung disorder that is characterized by airflow obstruction and the third leading cause of death, globally. COPD is influenced by environmental and genetic factors. Here, we measured the serum level of matrix metalloproteinase-9 (MMP-9), cyclooxygenase-2 (COX-2) and prostaglandin E-2 (PGE-2) and reveal the correlation between their levels in COPD subjects. In this study, we included a total of 79 COPD and 79 healthy controls. We assessed demographic profile, risk factors, respiratory symptoms, clinical history, COPD Assessment Test (CAT) score and spirometry. Further, we determined the serum levels of MMP-9, COX-2 and PGE-2 by enzyme-linked immunosorbent assay (ELISA). The correlation between their serum levels was also determined. Among the studied population age, gender, body mass index and socioeconomic status were comparable. Serum levels of MMP-9, COX-2 and PGE-2 were significantly increased in the COPD group than in healthy controls (P < 0.0001). Moreover, MMP-9, COX-2 and PGE-2 levels were increased with the GOLD grades and CAT score (> 10). Serum levels of MMP-9, COX-2 and PGE-2 was enhanced in patients with larger clinical history (> 20 years) than those with lower clinical history (< 10 years). Serum levels of MMP-9 and COX-2; MMP-9 and PGE-2; COX-2 and PGE-2 showed a positive correlation (P < 0.0001) with the COPD group. Our data demonstrate that serum levels of MMP-9, COX-2 and PGE-2 were correlated with the GOLD grade, CAT score and clinical history of the COPD group, pointing that they can be used as a indicators to understand the disease progression. Supplementary Information The online version contains supplementary material available at 10.1007/s12291-021-00973-2.
Collapse
|
12
|
Ni Y, Yu Y, Dai R, Shi G. Diffusing capacity in chronic obstructive pulmonary disease assessment: A meta-analysis. Chron Respir Dis 2021; 18:14799731211056340. [PMID: 34855516 PMCID: PMC8649441 DOI: 10.1177/14799731211056340] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 09/25/2021] [Indexed: 12/14/2022] Open
Abstract
To achieve a multidimensional evaluation of chronic obstructive pulmonary disease (COPD) patients, the spirometry measures are supplemented by assessment of symptoms, risk of exacerbations, and CT imaging. However, the measurement of diffusing capacity of the lung for carbon monoxide (DLCO) is not included in most common used models of COPD assessment. Here, we conducted a meta-analysis to evaluate the role of DLCO in COPD assessment.The studies were identified by searching the terms "diffusing capacity" OR "diffusing capacity for carbon monoxide" or "DLCO" AND "COPD" AND "assessment" in Pubmed, Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, Scopus, and Web of Science databases. The mean difference of DLCO % predict was assessed in COPD patient with different severity (according to GOLD stage and GOLD group), between COPD patients with or without with frequent exacerbation, between survivors and non-survivors, between emphysema dominant and non-emphysema dominant COPD patients, and between COPD patients with or without pulmonary hypertension.43 studies were included in the meta-analysis. DLCO % predicted was significantly lower in COPD patients with more severe airflow limitation (stage II/IV), more symptoms (group B/D), and high exacerbation risk (group C/D). Lower DLCO % predicted was also found in exacerbation patients and non-survivors. Low DLCO % predicted was related to emphysema dominant phenotype, and COPD patients with PH.The current meta-analysis suggested that DLCO % predicted might be an important measurement for COPD patients in terms of severity, exacerbation risk, mortality, emphysema domination, and presence of pulmonary hypertension. As diffusion capacity reflects pulmonary ventilation and perfusion at the same time, the predictive value of DLCO or DLCO combined with other criteria worth further exploration.
Collapse
Affiliation(s)
- Yingmeng Ni
- Department of Respiratory and Critical Care
Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Youchao Yu
- Department of Respiratory and Critical Care
Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Ranran Dai
- Department of Respiratory and Critical Care
Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Guochao Shi
- Department of Respiratory and Critical Care
Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| |
Collapse
|
13
|
Fang L, Roth M, S'ng CT, Tamm M, Han B, Hoang BX. Zinc salicylate reduces airway smooth muscle cells remodelling by blocking mTOR and activating p21 (Waf1/Cip1). J Nutr Biochem 2020; 89:108563. [PMID: 33326841 DOI: 10.1016/j.jnutbio.2020.108563] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/22/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Asthma is characterized by chronic inflammation and tissue remodeling of the airways. Remodeling is resistant to pharmaceutical therapies. This study investigated the effect of zinc salicylate-methylsulfonylmethane (Zn-Sal-MSM) compared to zinc salicylate (Zn-Sal), or sodium salicylate (Na-Sal), or zinc chloride (ZnCl2) on remodeling parameters of human airway smooth muscle cells (ASMC). Human ASMC obtained from asthma patients (n=7) and non-asthma controls (n=7) were treated with one of the reagents. Cell proliferation and viability was determined by direct cell counts and MTT assay. The expression of and phosphorylation proteins was determined by Western-blotting, ELISA, immunofluorescence, and mass spectrometry. Extracellular matrix deposition by ELISA. Zn-Sal-MSM, Zn-Sal and Na-Sal (0.1-100 µg/mL) significantly reduced PDGF-BB-induced proliferation in a concentration dependent manner, while ZnCl2 was toxic. The reduced proliferation correlated with increased expression of the cell cycle inhibitor p21(Waf1/Cip1), and reduced activity of Akt, p70S6K, and Erk1/2. Zn-Sal-MSM, Zn-Sal, but not Na-Sal reduced the deposition of fibronectin and collagen type-I. Furthermore, Zn-Sal-MSM reduced the mitochondria specific COX4 expression. Mass spectrometry indicated that Zn-Sal-MSM modified the expression of several signaling proteins and zinc-dependent enzymes. In conclusion, Zn-Sal-MSM and Zn-Sal potentially prevent airway wall remodeling in asthma by inhibition of both the Erk1/2 and mTOR signaling pathways.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research/Pneumology, Department of Biomedicine/Internal Medicine, University and University Hospital Basel, Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research/Pneumology, Department of Biomedicine/Internal Medicine, University and University Hospital Basel, Basel, Switzerland.
| | | | - Michael Tamm
- Pulmonary Cell Research/Pneumology, Department of Biomedicine/Internal Medicine, University and University Hospital Basel, Basel, Switzerland
| | - Bo Han
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, California
| | - Ba Xuan Hoang
- Cordoba-Nimni Tissue Engineering and Drug Discovery Lab, Department of Surgery, University of Southern California, Los Angeles, California
| |
Collapse
|
14
|
Karakioulaki M, Papakonstantinou E, Stolz D. Extracellular matrix remodelling in COPD. Eur Respir Rev 2020; 29:29/158/190124. [PMID: 33208482 DOI: 10.1183/16000617.0124-2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/16/2020] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the lung plays several important roles in lung function, as it offers a low resistant pathway that allows the exchange of gases, provides compressive strength and elasticity that supports the fragile alveolar-capillary intersection, controls the binding of cells with growth factors and cell surface receptors and acts as a buffer against retention of water.COPD is a chronic inflammatory respiratory condition, characterised by various conditions that result in progressive airflow limitation. At any stage in the course of the disease, acute exacerbations of COPD may occur and lead to accelerated deterioration of pulmonary function. A key factor of COPD is airway remodelling, which refers to the serious alterations of the ECM affecting airway wall thickness, resistance and elasticity. Various studies have shown that serum biomarkers of ECM turnover are significantly associated with disease severity in patients with COPD and may serve as potential targets to control airway inflammation and remodelling in COPD. Unravelling the complete molecular composition of the ECM in the diseased lungs will help to identify novel biomarkers for disease progression and therapy.
Collapse
Affiliation(s)
- Meropi Karakioulaki
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland.,Dept of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daiana Stolz
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| |
Collapse
|
15
|
Zheng XY, Li ZL, Li C, Guan WJ, Li LX, Xu YJ. Effects of cigarette smoking and biomass fuel on lung function and respiratory symptoms in middle-aged adults and the elderly in Guangdong province, China: A cross-sectional study. INDOOR AIR 2020; 30:860-871. [PMID: 32249960 DOI: 10.1111/ina.12671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 06/11/2023]
Abstract
We sought to investigate the association between active cigarette smoking (ex- and current smokers) with or without exposure to biomass fuels and respiratory symptoms/lung function in middle-aged adults and the elderly. In the chronic obstructive pulmonary disease surveillance conducted in six cities of Guangdong province, China, we surveyed 1986 residents aged 40-93 years. We recorded respiratory symptoms, smoking status, use of biomass fuel, and other covariates by using a structured questionnaire. All models were adjusted for second-hand smoking. Active smoking with or without exposure to biomass fuels was significantly associated with wheezing, chronic cough, and phlegm (all P < .05). Active smoking alone with or without exposure to biomass fuels was associated with 2.5% and 0.6% reduction in the mean forced vital capacity predicted, 6.8% and 4.2% reduction in the mean forced expiratory volume in one second predicted, and 9.0% and 4.7% reduction in the mean maximal mid-expiratory flow predicted compared with the exposure to neither smoking nor biomass fuels, respectively. However, exposure to biomass fuel alone was associated with neither greater lung function impairment nor respiratory symptoms except for chronic cough. Efforts should be made to protect home owners and their family from adverse effects of indoor air pollution.
Collapse
Affiliation(s)
- Xue-Yan Zheng
- Institute of Non-Communicable Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, China
| | - Zhang-Long Li
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chuan Li
- Institute of Non-Communicable Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Li-Xia Li
- Department of Epidemiology and Biostatistics, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yan-Jun Xu
- Institute of Non-Communicable Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangdong, China
| |
Collapse
|
16
|
Toumpanakis D, Mizi E, Vassilakopoulou V, Dettoraki M, Chatzianastasiou A, Perlikos F, Giatra G, Moscholaki M, Theocharis S, Vassilakopoulos T. Spontaneous Breathing Through Increased Airway Resistance Augments Elastase-Induced Pulmonary Emphysema. Int J Chron Obstruct Pulmon Dis 2020; 15:1679-1688. [PMID: 32764913 PMCID: PMC7367735 DOI: 10.2147/copd.s256750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/02/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Resistive breathing (RB), the pathophysiologic hallmark of chronic obstructive pulmonary disease (COPD), especially during exacerbations, is associated with significant inflammation and mechanical stress on the lung. Mechanical forces are implicated in the progression of emphysema that is a major pathologic feature of COPD. We hypothesized that resistive breathing exacerbates emphysema. Methods C57BL/6 mice were exposed to 0.75 units of pancreatic porcine elastase intratracheally to develop emphysema. Resistive breathing was applied by suturing a nylon band around the trachea to reduce surface area to half for the last 24 or 72 hours of a 21-day time period after elastase treatment in total. Following RB (24 or 72 hours), lung mechanics were measured and bronchoalveolar lavage (BAL) was performed. Emphysema was quantified by the mean linear intercept (Lm) and the destructive index (DI) in lung tissue sections. Results Following 21 days of intratracheal elastase exposure, Lm and DI increased in lung tissue sections [Lm (μm), control 39.09±0.76, elastase 62.05±2.19, p=0.003 and DI, ctr 30.95±2.75, elastase 73.12±1.75, p<0.001]. RB for 72 hours further increased Lm by 64% and DI by 19%, compared to elastase alone (p<0.001 and p=0.02, respectively). RB induced BAL neutrophilia in elastase-treated mice. Static compliance (Cst) increased in elastase-treated mice [Cst (mL/cmH2O), control 0.067±0.001, elastase 0.109±0.006, p<0.001], but superimposed RB decreased Cst, compared to elastase alone [Cst (mL/cmH2O), elastase+RB24h 0.090±0.004, p=0.006 to elastase, elastase+RB72h 0.090±0.005, p=0.006 to elastase]. Conclusion Resistive breathing augments pulmonary inflammation and emphysema in an elastase-induced emphysema mouse model.
Collapse
Affiliation(s)
- Dimitrios Toumpanakis
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece.,3rd Department of Critical Care Medicine, Evgenideio Hospital, Medical School, University of Athens, Athens, Greece
| | - Eleftheria Mizi
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Vyronia Vassilakopoulou
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Maria Dettoraki
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Athanasia Chatzianastasiou
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Fotis Perlikos
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | - Georgia Giatra
- 3rd Department of Critical Care Medicine, Evgenideio Hospital, Medical School, University of Athens, Athens, Greece
| | - Marina Moscholaki
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece
| | | | - Theodoros Vassilakopoulos
- "Marianthi Simou" Applied Biomedical Research and Training Center, Medical School, University of Athens, Evangelismos Hospital, Athens, Greece.,3rd Department of Critical Care Medicine, Evgenideio Hospital, Medical School, University of Athens, Athens, Greece
| |
Collapse
|
17
|
Hu L, Liu F, Li L, Zhang L, Yan C, Li Q, Qiu J, Dong J, Sun J, Zhang H. Effects of icariin on cell injury and glucocorticoid resistance in BEAS-2B cells exposed to cigarette smoke extract. Exp Ther Med 2020; 20:283-292. [PMID: 32550884 PMCID: PMC7296294 DOI: 10.3892/etm.2020.8702] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/11/2020] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) exert a therapeutic effect in numerous chronic inflammatory diseases. However, chronic obstructive pulmonary disease (COPD) tends to be GC-resistant. Icariin, a major component of flavonoids isolated from Epimedium brevicornum Maxim (Berberidaceae), significantly relieves symptoms in patients with COPD. However, the mechanism of action remains unclear and further investigation is required to establish whether it may serve as an alternative or complementary therapy for COPD. The aim of the present study was to determine the effects of icariin in human bronchial epithelial cells exposed to cigarette smoke extract (CSE) and to determine whether icariin reverses GC resistance. The results revealed that icariin significantly increased the proliferation of CSE-exposed cells. Furthermore, icariin significantly increased protein expression of the anti-inflammatory factor interleukin (IL)-10 and significantly decreased protein expression of the pro-inflammatory factors IL-8 and tumor necrosis factor α. Icariin also attenuated the expression of the cellular matrix remodelling biomarkers matrix metallopeptidase 9 and tissue inhibitor of metalloproteinase 1, and decreased the production of reactive oxygen species (ROS). In addition, icariin regulated the expression of GC resistance-related factors, such as GC receptors, histone deacetylase 2, nuclear factor erythroid-2-related factor 2 and nuclear factor κ B. The results obtained in the present study suggested that icariin may decrease CSE-induced inflammation, airway remodelling and ROS production by mitigating GC resistance. In conclusion, icariin may potentially be used in combination with GCs to increase therapeutic efficacy and reduce GC resistance in COPD.
Collapse
Affiliation(s)
- Lingli Hu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Feng Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Lulu Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Li Zhang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Chen Yan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Qiuping Li
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Jian Qiu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| | - Hongying Zhang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China.,Institute of Integrated Traditional Chinese and Western Medicine, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
18
|
Mamber SW, Gurel V, Lins J, Ferri F, Beseme S, McMichael J. Effects of cannabis oil extract on immune response gene expression in human small airway epithelial cells (HSAEpC): implications for chronic obstructive pulmonary disease (COPD). J Cannabis Res 2020; 2:5. [PMID: 33526116 PMCID: PMC7819312 DOI: 10.1186/s42238-019-0014-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 12/29/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is commonly associated with both a pro-inflammatory and a T-helper 1 (Th1) immune response. It was hypothesized that cannabis oil extract can alleviate COPD symptoms by eliciting an anti-inflammatory Th2 immune response. Accordingly, the effects of cannabis oil extract on the expression of 84 Th2 and related immune response genes in human small airways epithelial cells (HSAEpC) were investigated. METHODS HSAEpC from a single donor were treated with three dilutions of a standardized cannabis oil extract (1:400, 1:800 and 1:1600) along with a solvent control (0.25% [2.5 ul/ml] ethanol) for 24 h. There were four replicates per treatment dilution, and six for the control. RNA isolated from cells were employed in pathway-focused quantitative polymerase chain reaction (qPCR) microarray assays. RESULTS The extract induced significant (P < 0.05) changes in expression of 37 tested genes. Six genes (CSF2, IL1RL1, IL4, IL13RA2, IL17A and PPARG) were up-regulated at all three dilutions. Another two (CCL22 and TSLP) were up-regulated while six (CLCA1, CMA1, EPX, LTB4R, MAF and PMCH) were down-regulated at the 1:400 and 1:800 dilutions. The relationship of differentially-expressed genes of interest to biologic pathways was explored using the Database for Annotation, Visualization and Integrated Discovery (DAVID). CONCLUSIONS This exploratory investigation indicates that cannabis oil extract may affect expression of specific airway epithelial cell genes that could modulate pro-inflammatory or Th1 processes in COPD. These results provide a basis for further investigations and have prompted in vivo studies of the effects of cannabis oil extract on pulmonary function. TRIAL REGISTRATION NONE (all in vitro experiments).
Collapse
Affiliation(s)
- Stephen W Mamber
- Beech Tree Labs Inc., 1 Virginia Ave, Suite 103, Providence, RI, 02905, USA
- The Institute for Therapeutic Discovery, Delanson, NY, 12053, USA
| | - Volkan Gurel
- Beech Tree Labs Inc., 1 Virginia Ave, Suite 103, Providence, RI, 02905, USA
| | - Jeremy Lins
- Beech Tree Labs Inc., 1 Virginia Ave, Suite 103, Providence, RI, 02905, USA
| | - Fred Ferri
- NCM Biotechnology, Newport, RI, 02840, USA
| | - Sarah Beseme
- Beech Tree Labs Inc., 1 Virginia Ave, Suite 103, Providence, RI, 02905, USA.
| | - John McMichael
- Beech Tree Labs Inc., 1 Virginia Ave, Suite 103, Providence, RI, 02905, USA
- The Institute for Therapeutic Discovery, Delanson, NY, 12053, USA
| |
Collapse
|
19
|
Du X, Yuan L, Wu M, Men M, He R, Wang L, Wu S, Xiang Y, Qu X, Liu H, Qin X, Hu C, Qin L, Liu C. Variable DNA methylation of aging-related genes is associated with male COPD. Respir Res 2019; 20:243. [PMID: 31684967 PMCID: PMC6829949 DOI: 10.1186/s12931-019-1215-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a chronic lung inflammatory disease which has a close relationship with aging. Genome-wide analysis reveals that DNA methylation markers vary obviously with age. DNA methylation variations in peripheral blood have the potential to be biomarkers for COPD. However, the specific DNA methylation of aging-related genes in the peripheral blood of COPD patients remains largely unknown. Methods Firstly, 9 aging-related differentially expressed genes (DEGs) in COPD patients were screened out from the 25 aging-related genes profile through a comprehensive screening strategy. Secondly, qPCR and multiple targeted bisulfite enrichment sequencing (MethTarget) were used to detect the mRNA level and DNA methylation level of the 9 differentially expressed genes in the peripheral blood of 60 control subjects and 45 COPD patients. The candidate functional CpG sites were selected on the basis of the regulation ability of the target gene expression. Thirdly, the correlation was evaluated between the DNA methylation level of the key CpG sites and the clinical parameters of COPD patients, including forced expiratory volume in one second (FEV1), forced expiratory volume in one second as percentage of predicted volume (FEV1%), forced expiratory volume/ forced vital capacity (FEV/FVC), modified British medical research council (mMRC) score, acute exacerbation frequency and the situation of frequent of acute aggravation (CAT) score. Lastly, differentially methylated CpG sites unrelated to smoking were also determined in COPD patients. Results Of the 9 differentially expressed aging-related genes, the mRNA expression of 8 genes were detected to be significantly down-regulated in COPD group, compared with control group. Meanwhile, the methylated level of all aging-related genes was changed in COPD group containing 219 COPD-related CpG sites in total. Notably, 27 CpG sites of FOXO3 gene showed a lower False Discovery Rate (FDR) and higher methylation difference values. Also, some variable DNA methylation is associated with the severity of COPD. Additionally, of the 219 COPD-related CpG sites, 147 CpG sites were not related to smoking. Conclusion These results identified that the mRNA expression and DNA methylation level of aging-related genes were changed in male COPD patients, which provides a molecular link between aging and COPD. The identified CpG markers are associated with the severity of COPD and provide new insights into the prediction and identification of COPD.
Collapse
Affiliation(s)
- Xizi Du
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.,Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Mengping Wu
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Meichao Men
- Health Management Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruoxi He
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leyuan Wang
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Shuangyan Wu
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Yang Xiang
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiangping Qu
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Huijun Liu
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Chengping Hu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chi Liu
- Department of Physiology; China-Africa Infection Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
20
|
Dreschers S, Platen C, Ludwig A, Gille C, Köstlin N, Orlikowsky TW. Metalloproteinases TACE and MMP-9 Differentially Regulate Death Factors on Adult and Neonatal Monocytes After Infection with Escherichia coli. Int J Mol Sci 2019; 20:ijms20061399. [PMID: 30897723 PMCID: PMC6471605 DOI: 10.3390/ijms20061399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/22/2019] [Accepted: 02/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Cleaving ligands and receptors of the tumor necrosis factor (TNF) superfamily can critically regulate the induction of apoptosis. Matrix metalloproteinases (MMPs) such as MMP-9 and tumor necrosis factor-α-converting enzyme (TACE) have been shown to cleave CD95-Ligand (CD95L) and TNF/(TNF receptor-1) TNFR1 which induce phagocytosis induced cell death (PICD) in adult monocytes. This process is reduced in neonatal monocytes. Methods: Here we tested in vitro, whether Escherichia coli infection mounts for activation of MMP-9 and TACE in monocytes and whether this process regulates PICD. Results: The surface expression of TACE was most prominent on infected adult monocytes. In contrast, surface presentation of MMP-9 was highest on infected neonatal monocytes. Selective blocking of MMP-9 decreased CD95L secretion, while inhibition of TACE left CD95L secretion unaltered. Blocking of MMP-9 increased surface CD95L (memCD95L) expression on infected neonatal monocytes to levels comparable to infected adult monocytes. Moreover, MMP-9 inhibition raised PICD of infected neonatal monocytes to levels observed for infected adult monocytes. In contrast, TACE inhibition decreased PICD in infected monocytes. Addition of extracellular TNF effectively induced memCD95L presentation and PICD of adult monocytes and less of neonatal monocytes. Conclusion: MMP-9 activity is crucial for downregulating cell-contact dependent PICD in E. coli infected neonatal monocytes. By this mechanism, MMP-9 could contribute to reducing sustained inflammation in neonates.
Collapse
Affiliation(s)
- Stephan Dreschers
- Department of Neonatology, University Children's Hospital, Aachen 52074, Germany.
| | - Christopher Platen
- Department of Neonatology, University Children's Hospital, Aachen 52074, Germany.
| | - Andreas Ludwig
- Department of Pharmacology and Toxicology, University Hospital, Aachen 52074, Germany.
| | - Christian Gille
- Department of Neonatology, University Children's Hospital, Tuebingen 72074, Germany.
| | - Natascha Köstlin
- Department of Pharmacology and Toxicology, University Hospital, Aachen 52074, Germany.
| | | |
Collapse
|
21
|
Higham A, Quinn AM, Cançado JED, Singh D. The pathology of small airways disease in COPD: historical aspects and future directions. Respir Res 2019; 20:49. [PMID: 30832670 PMCID: PMC6399904 DOI: 10.1186/s12931-019-1017-y] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/25/2019] [Indexed: 12/16/2022] Open
Abstract
Small airways disease (SAD) is a cardinal feature of chronic obstructive pulmonary disease (COPD) first recognized in the nineteenth century. The diverse histopathological features associated with SAD underpin the heterogeneous nature of COPD. Our understanding of the key molecular mechanisms which drive the pathological changes are not complete. In this article we will provide a historical overview of key histopathological studies which have helped shape our understanding of SAD and discuss the hallmark features of airway remodelling, mucous plugging and inflammation. We focus on the relationship between SAD and emphysema, SAD in the early stages of COPD, and the mechanisms which cause SAD progression, including bacterial colonization and exacerbations. We discuss the need to specifically target SAD to attenuate the progression of COPD.
Collapse
Affiliation(s)
- Andrew Higham
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Anne Marie Quinn
- Department of Histopathology, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Dave Singh
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Medicines Evaluation Unit, The Langley Building, Southmoor Road, Manchester, UK
| |
Collapse
|
22
|
Wells JM, Parker MM, Oster RA, Bowler RP, Dransfield MT, Bhatt SP, Cho MH, Kim V, Curtis JL, Martinez FJ, Paine R, O'Neal W, Labaki WW, Kaner RJ, Barjaktarevic I, Han MK, Silverman EK, Crapo JD, Barr RG, Woodruff P, Castaldi PJ, Gaggar A. Elevated circulating MMP-9 is linked to increased COPD exacerbation risk in SPIROMICS and COPDGene. JCI Insight 2018; 3:123614. [PMID: 30429371 DOI: 10.1172/jci.insight.123614] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/04/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Matrix metalloprotease 9 (MMP-9) is associated with inflammation and lung remodeling in chronic obstructive pulmonary disease (COPD). We hypothesized that elevated circulating MMP-9 represents a potentially novel biomarker that identifies a subset of individuals with COPD with an inflammatory phenotype who are at increased risk for acute exacerbation (AECOPD). METHODS We analyzed Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS) and Genetic Epidemiology of COPD (COPDGene) cohorts for which baseline and prospective data were available. Elevated MMP-9 was defined based on >95th percentile plasma values from control (non-COPD) sample in SPIROMICS. COPD subjects were classified as having elevated or nonelevated MMP-9. Logistic, Poisson, and Kaplan-Meier analyses were used to identify associations with prospective AECOPD in both cohorts. RESULTS Elevated MMP-9 was present in 95/1,053 (9%) of SPIROMICS and 41/140 (29%) of COPDGene participants with COPD. COPD subjects with elevated MMP-9 had a 13%-16% increased absolute risk for AECOPD and a higher median (interquartile range; IQR) annual AECOPD rate (0.33 [0-0.74] versus 0 [0-0.80] events/year and 0.9 [0.5-2] versus 0.5 [0-1.4] events/year for SPIROMICS and COPDGene, respectively). In adjusted models within each cohort, elevated MMP-9 was associated with increased odds (odds ratio [OR], 1.71; 95%CI, 1.00-2.90; and OR, 3.03; 95%CI, 1.02-9.01), frequency (incidence rate ratio [IRR], 1.45; 95%CI, 1.23-1.7; and IRR, 1.24; 95%CI, 1.03-1.49), and shorter time-to-first AECOPD (21.7 versus 31.7 months and 14 versus 21 months) in SPIROMICS and COPDGene, respectively. CONCLUSIONS Elevated MMP-9 was independently associated with AECOPD risk in 2 well-characterized COPD cohorts. These findings provide evidence for MMP-9 as a prognostic biomarker and potential therapeutic target in COPD. TRIAL REGISTRATION ClinicalTrials.gov: NCT01969344 (SPIROMICS) and NCT00608764 (COPDGene). FUNDING This work was funded by K08 HL123940 to JMW; R01HL124233 to PJC; Merit Review I01 CX000911 to JLC; R01 (R01HL102371, R01HL126596) and VA Merit (I01BX001756) to AG. SPIROMICS (Subpopulations and Intermediate Outcomes in COPD Study) is funded by contracts from the NHLBI (HHSN268200900013C, HHSN268200900014C,HHSN268200900015C HHSN268200900016C, HHSN268200900017C, HHSN268200900018C, HHSN268200900019C, and HHSN268200900020C) and a grant from the NIH/NHLBI (U01 HL137880), and supplemented by contributions made through the Foundation for the NIH and the COPD Foundation from AstraZeneca/MedImmune; Bayer; Bellerophon Therapeutics; Boehringer-Ingelheim Pharmaceuticals Inc.; Chiesi Farmaceutici; Forest Research Institute Inc.; GlaxoSmithKline; Grifols Therapeutics Inc.; Ikaria Inc.; Novartis Pharmaceuticals Corporation; Nycomed GmbH; ProterixBio; Regeneron Pharmaceuticals Inc.; Sanofi; Sunovion; Takeda Pharmaceutical Company; and Theravance Biopharma and Mylan. COPDGene is funded by the NHLBI (R01 HL089897 and R01 HL089856) and by the COPD Foundation through contributions made to an Industry Advisory Board composed of AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Novartis, Pfizer, Siemens, and Sunovion.
Collapse
Affiliation(s)
- J Michael Wells
- Division of Pulmonary and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Margaret M Parker
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Robert A Oster
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Russ P Bowler
- Division of Pulmonary, Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - Mark T Dransfield
- Division of Pulmonary and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Surya P Bhatt
- Division of Pulmonary and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, Birmingham, Alabama, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victor Kim
- Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Fernando J Martinez
- Departments of Medicine and Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah, USA.,Medical Service, Salt Lake City VA Medical Center, Salt Lake City, Utah, USA
| | - Wanda O'Neal
- Marsico Lung Institute/Cystic Fibrosis Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert J Kaner
- Departments of Medicine and Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Igor Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, UCLA, Los Angeles, California, USA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - James D Crapo
- Division of Pulmonary, Critical Care Medicine, National Jewish Health, Denver, Colorado, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Prescott Woodruff
- Division of Pulmonary and Critical Care Medicine, UCSF, School of Medicine, San Francisco, California, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Amit Gaggar
- Division of Pulmonary and Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, Birmingham, Alabama, USA.,Birmingham VA Medical Center, Birmingham, Alabama, USA
| | -
- The SPIROMICS and COPDGene groups are detailed in the Supplemental Acknowledgments
| |
Collapse
|
23
|
Liu Z, Shi F, Liu J, Gao C, Pei M, Li J, Li P. Effect of the emphysema subtypes of patients with chronic obstructive pulmonary disease on airway inflammation and COTE index. Exp Ther Med 2018; 16:4745-4752. [DOI: 10.3892/etm.2018.6799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 07/13/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Zheng Liu
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Fang Shi
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Jun‑Xia Liu
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Chang‑Lan Gao
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Meng‑Miao Pei
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Jing Li
- Department of Respiratory Tract Medicine, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| | - Pei‑Xiu Li
- Department of Radiology, The Petroleum Clinical Medical College of Hebei Medical University, Langfang, Hebei 065000, P.R. China
| |
Collapse
|
24
|
Yu WC, Cong JP, Mi LY. Expressions of TOLL-like receptor 4 (TLR-4) and matrix metalloproteinase 9 (MMP-9)/Tissue inhibitor of metalloproteinase 1 (TIMP-1) in pulmonary blood vessels with chronic obstructive pulmonary diseases and their relationships with pulmonary vascular remodelling. ACTA ACUST UNITED AC 2018; 64:361-367. [PMID: 30133616 DOI: 10.1590/1806-9282.64.04.361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/16/2017] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aims at investigating the expressions of TOLL-like receptor 4 (TLR-4) and matrix metalloproteinase 9 (MMP-9)/ tissue inhibitor of metalloproteinase 1 (TIMP-1) in pulmonary blood vessels with chronic obstructive pulmonary disease (COPD) and their relationships with pulmonary vascular remodelling (PVR). METHODS 60 para-tumour tissues were divided into the COPD group and the control group (n=30); the inflammations, pulmonary artery wall area/total artery area (WA%), and wall thickness/vascular outer diameter (WT%) were compared. The expressions of TLR-4, MMP-9/TIMP-1, and PCNA in pulmonary vascular smooth muscle cells were detected, and their relationships with PVR were then analysed. RESULTS The inflammations (1.6±0.8), WA% (44.0±6.4), and WT% (27.3±3.3) in the COPD group were higher than in the control group (0.3±0.5, 26.1±2.8, 15.6±1.8), and the expressions of TLR-4 (31.4±147) and MMP-9/TIMP-1 (2.2±2.6) were increased compared to the control group (4.7±4.5, 1.9±12). Correlation analysis: TLR-4 and MMP-9/TIMP-1 were positively correlated with the inflammations (r=0.18, P<0.01), WA% (r=0.68, P<0.01), and WT% (r=0.73, P<0.01), as well as positively correlated with the expression of PCNA (r=0.44, P<0.01); the upregulation of TLR-4 was positively correlated with the expressions of MMP-9 and TIMP-1. CONCLUSIONS The upregulation of TLR-4 in the pulmonary arterial smooth muscle cells of COPD patients could promote the inflammations and the MMP-9 expression, thus causing abnormal degradation of extracellular matrix, so it played an important role in the process of PVR.
Collapse
Affiliation(s)
- Wen-Cheng Yu
- Department of Respiratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jin-Peng Cong
- Department of Respiratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Li-Yun Mi
- Department of Respiratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
25
|
Proteases and Their Inhibitors in Chronic Obstructive Pulmonary Disease. J Clin Med 2018; 7:jcm7090244. [PMID: 30154365 PMCID: PMC6162857 DOI: 10.3390/jcm7090244] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/21/2022] Open
Abstract
In the context of respiratory disease, chronic obstructive pulmonary disease (COPD) is the leading cause of mortality worldwide. Despite much development in the area of drug development, currently there are no effective medicines available for the treatment of this disease. An imbalance in the protease: Antiprotease ratio in the COPD lung remains an important aspect of COPD pathophysiology and several studies have shown the efficacy of antiprotease therapy in both in vitro and in vivo COPD models. However more in-depth studies will be required to validate the efficacy of lead drug molecules targeting these proteases. This review discusses the current status of protease-directed drugs used for treating COPD and explores the future prospects of utilizing the potential of antiprotease-based therapeutics as a treatment for this disease.
Collapse
|
26
|
Hou HH, Wang HC, Cheng SL, Chen YF, Lu KZ, Yu CJ. MMP-12 activates protease-activated receptor-1, upregulates placenta growth factor, and leads to pulmonary emphysema. Am J Physiol Lung Cell Mol Physiol 2018; 315:L432-L442. [PMID: 29722565 DOI: 10.1152/ajplung.00216.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Because of the expansion of aging and smoking populations, chronic obstructive pulmonary disease (COPD) is predicted to be the third leading cause of death worldwide in 2030. Therefore, it is pertinent to develop effective therapy to improve management for COPD. Cigarette smoke-mediated protease-antiprotease imbalance is a major pathogenic mechanism for COPD and results in massive pulmonary infiltration of neutrophils and macrophages, releasing excessive neutrophil elastase (NE) and matrix metalloproteinases (MMPs). Our previous studies indicated that placenta growth factor (PGF) and PGF-triggered downstream signaling molecules mediate NE-induced lung epithelial cell apoptosis, which is a major pathogenic mechanism for pulmonary emphysema. However, the relationship between MMP-directed COPD and PGF remains elusive. We hypothesize that MMPs may upregulate PGF expression and be involved in MMP-mediated pathogenesis of COPD. In this study, we demonstrate that only MMP-12 can increase the expression of PGF by increasing early-growth response protein 1 (Egr-1) level through the activation of protease-activated receptor 1 (PAR-1). The PGF-mediated downstream signaling molecules drive caspase-3 and caspase-9-dependent apoptosis in bronchial epithelial cells. Both the upregulation of PGF by MMP-12 and PGF downstream signaling molecules with pulmonary apoptosis and emphysema were also demonstrated in animals. Given these findings, we suggest that both human COPD-associated elastases, NE, and MMP-12, upregulate PGF expression and promote the progression of emphysema and COPD.
Collapse
Affiliation(s)
- Hsin-Han Hou
- Department of Internal Medicine, National Taiwan University Hospital , Taiwan.,Department of Internal Medicine, National Taiwan University, College of Medicine , Taiwan
| | - Hao-Chien Wang
- Department of Internal Medicine, National Taiwan University Hospital , Taiwan.,Department of Internal Medicine, National Taiwan University, College of Medicine , Taiwan
| | - Shih-Lung Cheng
- Department of Internal Medicine, Far Eastern Memorial Hospital , Taiwan.,Department of Chemical Engineering and Materials Science, Yuan-Ze University , Taiwan
| | - Yen-Fu Chen
- Department of Internal Medicine, National Taiwan University Hospital, Yunlin Branch , Taiwan
| | - Kai-Zen Lu
- Department of Internal Medicine, National Taiwan University Hospital , Taiwan.,Department of Internal Medicine, National Taiwan University, College of Medicine , Taiwan
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital , Taiwan.,Department of Internal Medicine, National Taiwan University, College of Medicine , Taiwan
| |
Collapse
|
27
|
Abstract
Despite induction immunosuppression and the use of aggressive maintenance immunosuppressive regimens, acute allograft rejection following lung transplantation is still a problem with important diagnostic and therapeutic challenges. As well as causing early graft loss and mortality, acute rejection also initiates the chronic alloimmune responses and airway-centred inflammation that predispose to bronchiolitis obliterans syndrome (BOS), also known as chronic lung allograft dysfunction (CLAD), which is a major source of morbidity and mortality after lung transplantation. Cellular responses to human leukocyte antigens (HLAs) on the allograft have traditionally been considered the main mechanism of acute rejection, but the influence of humoral immunity is increasingly recognised. As with other several other solid organ transplants, antibody-mediated rejection (AMR) is now a well-accepted and distinct clinical entity in lung transplantation. While acute cellular rejection (ACR) has defined histopathological criteria, transbronchial biopsy is less useful in AMR and its diagnosis is complicated by challenges in the measurement of antibodies directed against donor HLA, and a determination of their significance. Increasing awareness of the importance of non-HLA antigens further clouds this issue. Here, we review the pathophysiology, diagnosis, clinical presentation and treatment of ACR and AMR in lung transplantation, and discuss future potential biomarkers of both processes that may forward our understanding of these conditions.
Collapse
Affiliation(s)
- Mark Benzimra
- Heart and Lung Transplant Unit, St Vincent's Hospital, Sydney, Australia
| | - Greg L Calligaro
- Division of Pulmonology, Department of Medicine, Groote Schuur Hospital, Cape Town, South Africa
| | - Allan R Glanville
- Heart and Lung Transplant Unit, St Vincent's Hospital, Sydney, Australia
| |
Collapse
|
28
|
Ezegbunam W, Foronjy R. Posttranscriptional control of airway inflammation. WILEY INTERDISCIPLINARY REVIEWS-RNA 2017; 9. [PMID: 29071794 DOI: 10.1002/wrna.1455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/07/2017] [Accepted: 09/08/2017] [Indexed: 12/18/2022]
Abstract
Acute inflammation in the lungs is a vital protective response, efficiently and swiftly eliminating inciters of tissue injury. However, in respiratory diseases characterized by chronic inflammation, such as chronic obstructive pulmonary disease and asthma, enhanced expression of inflammatory mediators leads to tissue damage and impaired lung function. Although transcription is an essential first step in the induction of proinflammatory genes, tight regulation of inflammation requires more rapid, flexible responses. Increasing evidence shows that such responses are achieved by posttranscriptional mechanisms directly affecting mRNA stability and translation initiation. RNA-binding proteins, microRNAs, and long noncoding RNAs interact with messenger RNA and each other to impact the stability and/or translation of mRNAs implicated in lung inflammation. Recent research has shown that these biological processes play a central role in the pathogenesis of several important pulmonary conditions. This review will highlight several posttranscriptional control mechanisms that influence lung inflammation and the known associations of derangements in these mechanisms with common respiratory diseases. WIREs RNA 2018, 9:e1455. doi: 10.1002/wrna.1455 This article is categorized under: RNA in Disease and Development > RNA in Disease RNA Structure and Dynamics > Influence of RNA Structure in Biological Systems RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Turnover and Surveillance > Regulation of RNA Stability.
Collapse
Affiliation(s)
- Wendy Ezegbunam
- Division of Pulmonary and Critical Care Medicine, Department of Medicine and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Robert Foronjy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
29
|
Ssanghwa-Tang, a traditional herbal formula, suppresses cigarette smoke-induced airway inflammation via inhibition of MMP-9 and Erk signaling. Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0033-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
30
|
Ginsenoside Rg1 Attenuates Cigarette Smoke-Induced Pulmonary Epithelial-Mesenchymal Transition via Inhibition of the TGF- β1/Smad Pathway. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7171404. [PMID: 29104873 PMCID: PMC5572594 DOI: 10.1155/2017/7171404] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 05/24/2017] [Accepted: 06/27/2017] [Indexed: 11/17/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a process associated with airway remodeling in chronic obstructive pulmonary disease (COPD), which leads to progressive pulmonary destruction. Panax ginseng is a traditional herbal medicine that has been shown to improve pulmonary function and exercise capacity in patients with COPD. Ginsenoside Rg1 is one of the main active components and was shown to inhibit oxidative stress and inflammation. The present study investigated the hypothesis that ginsenoside Rg1 attenuates EMT in COPD rats induced by cigarette smoke (CS) and human bronchial epithelial (HBE) cells exposed to cigarette smoke extract (CSE). Our data showed that CS or CSE exposure increased expression of the mesenchymal marker α-smooth muscle actin (α-SMA) and decreased expression of the epithelial marker epithelial cadherin (E-cad) in both lung tissues and HBE cells, which was markedly suppressed by ginsenoside Rg1. Importantly, CS-induced upregulation of TGF-β1/Smad pathway components, including TGF-β1, TGF-βR1, phospho-Smad2, and phospho-Smad3, was also inhibited by ginsenoside Rg1. Additionally, ginsenoside Rg1 mimicked the effect of SB525334, a TGF-βR1-Smad2/3 inhibitor, on suppression of EMT in CSE-induced HBE cells. Collectively, we concluded that ginsenoside Rg1 alleviates CS-induced pulmonary EMT, in both COPD rats and HBE cells, via inhibition of the TGF-β1/Smad pathway.
Collapse
|
31
|
Dransfield MT, Kunisaki KM, Strand MJ, Anzueto A, Bhatt SP, Bowler RP, Criner GJ, Curtis JL, Hanania NA, Nath H, Putcha N, Roark SE, Wan ES, Washko GR, Wells JM, Wendt CH, Make BJ. Acute Exacerbations and Lung Function Loss in Smokers with and without Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 195:324-330. [PMID: 27556408 DOI: 10.1164/rccm.201605-1014oc] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Acute exacerbations of chronic obstructive pulmonary disease (COPD) increase the risk of death and drive healthcare costs, but whether they accelerate loss of lung function remains controversial. Whether exacerbations in subjects with mild COPD or similar acute respiratory events in smokers without airflow obstruction affect lung function decline is unknown. OBJECTIVES To determine the association between acute exacerbations of COPD (and acute respiratory events in smokers without COPD) and the change in lung function over 5 years of follow-up. METHODS We examined data on the first 2,000 subjects who returned for a second COPDGene visit 5 years after enrollment. Baseline data included demographics, smoking history, and computed tomography emphysema. We defined exacerbations (and acute respiratory events in those without established COPD) as acute respiratory symptoms requiring either antibiotics or systemic steroids, and severe events by the need for hospitalization. Throughout the 5-year follow-up period, we collected self-reported acute respiratory event data at 6-month intervals. We used linear mixed models to fit FEV1 decline based on reported exacerbations or acute respiratory events. MEASUREMENTS AND MAIN RESULTS In subjects with COPD, exacerbations were associated with excess FEV1 decline, with the greatest effect in Global Initiative for Chronic Obstructive Lung Disease stage 1, where each exacerbation was associated with an additional 23 ml/yr decline (95% confidence interval, 2-44; P = 0.03), and each severe exacerbation with an additional 87 ml/yr decline (95% confidence interval, 23-151; P = 0.008); statistically significant but smaller effects were observed in Global Initiative for Chronic Obstructive Lung Disease stage 2 and 3 subjects. In subjects without airflow obstruction, acute respiratory events were not associated with additional FEV1 decline. CONCLUSIONS Exacerbations are associated with accelerated lung function loss in subjects with established COPD, particularly those with mild disease. Trials are needed to test existing and novel therapies in subjects with early/mild COPD to potentially reduce the risk of progressing to more advanced lung disease. Clinical trial registered with www.clinicaltrials.gov (NCT 00608764).
Collapse
Affiliation(s)
- Mark T Dransfield
- 1 Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama.,2 Birmingham VA Medical Center, Birmingham, Alabama
| | - Ken M Kunisaki
- 3 Minneapolis VA Health Care System, Minneapolis, Minnesota.,4 University of Minnesota, Minneapolis, Minnesota
| | | | - Antonio Anzueto
- 6 Pulmonary/Critical Care, University of Texas Health Science Center, San Antonio, Texas.,7 South Texas Veterans Health Care System, San Antonio, Texas
| | - Surya P Bhatt
- 1 Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | - Jeffrey L Curtis
- 9 University of Michigan, Ann Arbor, Michigan.,10 VA Ann Arbor Health Care System, Ann Arbor, Michigan
| | | | - Hrudaya Nath
- 1 Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Nirupama Putcha
- 12 Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | | | - Emily S Wan
- 13 Brigham and Women's Hospital, Boston, Massachusetts
| | | | - J Michael Wells
- 1 Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama.,2 Birmingham VA Medical Center, Birmingham, Alabama
| | - Christine H Wendt
- 3 Minneapolis VA Health Care System, Minneapolis, Minnesota.,4 University of Minnesota, Minneapolis, Minnesota
| | | | | |
Collapse
|
32
|
Lee JW, Park JW, Kwon OK, Lee HJ, Jeong HG, Kim JH, Oh SR, Ahn KS. NPS2143 Inhibits MUC5AC and Proinflammatory Mediators in Cigarette Smoke Extract (CSE)-Stimulated Human Airway Epithelial Cells. Inflammation 2017; 40:184-194. [PMID: 27866297 DOI: 10.1007/s10753-016-0468-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mucus overproduction is a fundamental hallmark of COPD that is caused by exposure to cigarette smoke. MUC5AC is one of the main mucin genes expressed in the respiratory epithelium, and its transcriptional upregulation often correlates with increased mucus secretion. Calcium-sensing receptor (CaSR) antagonists have been reported to possess anti-inflammatory effects. The purpose of the present study was to investigate the protective role of NPS2143, a selective CaSR antagonist on cigarette smoke extract (CSE)-stimulated NCI-H292 mucoepidermoid human lung cells. Treatment of NPS2143 significantly inhibited the expression of MUC5AC in CSE-stimulated H292 cells. NPS2143 reduced the expression of MMP-9 in CSE-stimulated H292 cells. NPS2143 also decreased the release of proinflammatory cytokines such as IL-6 and TNF-α in CSE-stimulated H292 cells. Furthermore, NPS2143 attenuated the activation of MAPKs (JNK, p38, and ERK) and inhibited the nuclear translocation of NF-κB in CSE-stimulated H292 cells. These results indicate that NPS2143 had a therapeutic potential in COPD.
Collapse
Affiliation(s)
- Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
| | - Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Kangwon, 200-701, Republic of Korea
| | - Hye Gwang Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Jae-Hong Kim
- Department of Life Sciences and Biotechnology, Korea University, 5-1 Anam-dong, Sungbuk-gu, Seoul, 136-701, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea.
| | - Kyoung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea.
| |
Collapse
|
33
|
Immune Responses to Tissue-Restricted Nonmajor Histocompatibility Complex Antigens in Allograft Rejection. J Immunol Res 2017; 2017:6312514. [PMID: 28164137 PMCID: PMC5253484 DOI: 10.1155/2017/6312514] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/06/2016] [Indexed: 01/02/2023] Open
Abstract
Chronic diseases that result in end-stage organ damage cause inflammation, which can reveal sequestered self-antigens (SAgs) in that organ and trigger autoimmunity. The thymus gland deletes self-reactive T-cells against ubiquitously expressed SAgs, while regulatory mechanisms in the periphery control immune responses to tissue-restricted SAgs. It is now established that T-cells reactive to SAgs present in certain organs (e.g., lungs, pancreas, and intestine) are incompletely eliminated, and the dysregulation of peripheral immuneregulation can generate immune responses to SAgs. Therefore, chronic diseases can activate self-reactive lymphocytes, inducing tissue-restricted autoimmunity. During organ transplantation, donor lymphocytes are tested against recipient serum (i.e., cross-matching) to detect antibodies (Abs) against donor human leukocyte antigens, which has been shown to reduce Ab-mediated hyperacute rejection. However, primary allograft dysfunction and rejection still occur frequently. Because donor lymphocytes do not express tissue-restricted SAgs, preexisting Abs against SAgs are undetectable during conventional cross-matching. Preexisting and de novo immune responses to tissue-restricted SAgs (i.e., autoimmunity) play a major role in rejection. In this review, we discuss the evidence that supports autoimmunity as a contributor to rejection. Testing for preexisting and de novo immune responses to tissue-restricted SAgs and treatment based on immune responses after organ transplantation may improve short- and long-term outcomes after transplantation.
Collapse
|
34
|
Stolz D, Leeming DJ, Kristensen JHE, Karsdal MA, Boersma W, Louis R, Milenkovic B, Kostikas K, Blasi F, Aerts J, Sand JM, Wouters EF, Rohde G, Prat C, Torres A, Welte T, Roth M, Papakonstantinou E, Tamm M. Systemic Biomarkers of Collagen and Elastin Turnover Are Associated With Clinically Relevant Outcomes in COPD. Chest 2017; 151:47-59. [DOI: 10.1016/j.chest.2016.08.1440] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/16/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022] Open
|
35
|
Effects of transient receptor potential canonical 1 (TRPC1) on the mechanical stretch-induced expression of airway remodeling-associated factors in human bronchial epithelioid cells. J Biomech 2016; 51:89-96. [PMID: 27986325 DOI: 10.1016/j.jbiomech.2016.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/15/2016] [Accepted: 12/04/2016] [Indexed: 02/06/2023]
Abstract
Research has shown that mechanical stress stimulation can cause airway remodeling. We investigate the effects of mechanical stretch on the expression of the airway remodeling-associated factors interleukin-13 (IL-13) and matrix metalloprotein-9 (MMP-9) and signaling pathways in human bronchial epithelioid (16HBE) cells under mechanical stretch. A Flexcell FX-4000 Tension System with a flexible substrate was applied to stretch 16HBE cells at a 15% elongation amplitude and 1Hz frequency, with stretching for 0.5h, 1h, 1.5h and 2h. The experimental group with higher IL-13, MMP-9, and TRPC1 expression and higher Ca2+ levels was selected for performing intervention experiment. These cells were pretreated with the transient receptor potential canonical 1 (TRPC1) channel antagonist SKF96365 and TRPC1-specific siRNA, and then mechanical stretch was applied. Our results provided evidences that mechanical pressure significantly increased IL-13, MMP-9, and TRPC1 protein and mRNA expression levels and intracellular Ca2+ fluorescence intensity at 4 time points compared with the control group. The peak IL-13, MMP-9, and TRPC1 expression levels were observed at 0.5h after exposure to mechanical pressure. IL-13 and MMP-9 expression levels and Ca2+ fluorescence intensity in the stretch+SKF96365 group and in the stretch+TRPC1 siRNA group were significantly lower than those were in the mechanical stretch group. By incubating the cells with the intracellular calcium chelator BAPTA-AM, the expression of IL-13 and MMP9 was significantly decreased, and the expression level of TRPC1 remained unchanged. These observations suggest that mechanical stretch may induce an influx of Ca2+ and up-regulation of IL-13 and MMP-9 expression in 16HBE cells via activation of TRPC1.
Collapse
|
36
|
Murray LA, Grainge C, Wark PA, Knight DA. Use of biologics to treat acute exacerbations and manage disease in asthma, COPD and IPF. Pharmacol Ther 2016; 169:1-12. [PMID: 27889330 DOI: 10.1016/j.pharmthera.2016.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A common feature of chronic respiratory disease is the progressive decline in lung function. The decline can be indolent, or it can be accelerated by acute exacerbations, whereby the patient experiences a pronounced worsening of disease symptoms. Moreover, acute exacerbations may also be a marker of insufficient disease management. The underlying cause of an acute exacerbation can be due to insults such as pathogens or environmental pollutants, or the cause can be unknown. For each acute exacerbation, the patient may require medical intervention such as rescue medication, or in more severe cases, hospitalization and ventilation and have an increased risk of death. Biologics, such as monoclonal antibodies, are being developed for chronic respiratory diseases including asthma, COPD and IPF. This therapeutic approach is particularly well suited for chronic use based on the route and frequency of delivery and importantly, the potential for disease modification. In recent clinical trials, the frequency of acute exacerbation has often been included as an endpoint, to help determine whether the investigational agent is impacting disease. Therefore the significance of acute exacerbations in driving disease, and their potential as a marker of disease activity and progression, has recently received much attention. There is also now a need to standardize the definition of an acute exacerbation in specific disease settings, particularly as this endpoint is increasingly used in clinical trials to also assess therapeutic efficacy. Moreover, specifically targeting exacerbations may offer a new therapeutic approach for several chronic respiratory diseases.
Collapse
Affiliation(s)
| | - Chris Grainge
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, Australia; School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, University Drive, Callaghan, NSW 2308, Australia
| |
Collapse
|
37
|
Chen L, Yang H, Xiao Y, Tang X, Li Y, Han Q, Fu J, Yang Y, Zhu Y. LncRNA GAS5 is a critical regulator of metastasis phenotype of melanoma cells and inhibits tumor growth in vivo. Onco Targets Ther 2016; 9:4075-87. [PMID: 27445498 PMCID: PMC4938146 DOI: 10.2147/ott.s98203] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The present study intended to demonstrate the effects of long noncoding RNA growth arrest-specific transcript 5 (GAS5) on the migration and invasion of melanoma cells. We first detected the expression of GAS5 among four kinds of melanoma cell lines, followed by constructing GAS5-knocked down and overexpressed stable cells. Next, we evaluated the effects of GAS5 on cell migration and invasion using wound healing and gelatin zymography assays. Finally, melanoma cells with different GAS5 expression were injected into nude mice, and the tumor volumes were recorded and tumor tissues were analyzed after sacrificing the mice. This study systematically examined the function of GAS5 in mediating melanoma metastasis and revealed that GAS5 plays an anticancer role in melanoma via regulating gelatinase A and B, both in vitro and in vivo.
Collapse
Affiliation(s)
- Long Chen
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China; PET/CT Center, Yunan Tumor Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Huixin Yang
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yanbin Xiao
- Department of Orthopaedics, Yunan Tumor Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Xiaoxia Tang
- Department of Pharmacy, the Second Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yuqian Li
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Qiaoqiao Han
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Junping Fu
- Department of Orthopaedics, Yunan Tumor Hospital, the Third Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yuye Yang
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, Kunming Medical University, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
38
|
Tan WSD, Peh HY, Liao W, Pang CH, Chan TK, Lau SH, Chow VT, Wong WSF. Cigarette Smoke-Induced Lung Disease Predisposes to More Severe Infection with Nontypeable Haemophilus influenzae: Protective Effects of Andrographolide. JOURNAL OF NATURAL PRODUCTS 2016; 79:1308-15. [PMID: 27104764 DOI: 10.1021/acs.jnatprod.5b01006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Cigarette smoke (CS) is associated with many maladies, one of which is chronic obstructive pulmonary disease (COPD). As the disease progresses, patients are more prone to develop COPD exacerbation episodes by bacterial infection, particularly to nontypeable Haemophilus influenza (NTHi) infection. The present study aimed to develop a CS-exposed mouse model that increases inflammation induced by NTHi challenge and investigate the protective effects of andrographolide, a bioactive molecule with anti-inflammatory and antioxidant properties isolated from the plant Andrographis paniculata. Female BALB/c mice exposed to 2 weeks of CS followed by a single intratracheal instillation of NTHi developed increased macrophage and neutrophil pulmonary infiltration, augmented cytokine levels, and heightened oxidative damage. Andrographolide effectively reduced lung cellular infiltrates and decreased lung levels of TNF-α, IL-1β, CXCL1/KC, 8-OHdG, matrix metalloproteinase-8 (MMP-8), and MMP-9. The protective actions of andrographolide on CS-predisposed NTHi inflammation might be attributable to increased nuclear factor erythroid-2-related factor 2 (Nrf2) activation and decreased Kelch-like ECH-associated protein 1 (Keap1) repressor function, resulting in enhanced gene expression of antioxidant enzymes including heme oxygenase-1 (HO-1), glutathione reductase (GR), glutathione peroxidase-2 (GPx-2), glutamate-cysteine ligase modifier (GCLM), and NAD(P)H quinone oxidoreductase 1 (NQO1). Taken together, these findings strongly support a therapeutic potential for andrographolide in preventing lung inflammation caused by NTHi in cigarette smokers.
Collapse
Affiliation(s)
- W S Daniel Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
| | - Hong Yong Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
| | - Chu Hui Pang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
| | - Tze Khee Chan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology (SMART) , 1 CREATE Way, Singapore 138602
| | - Suk Hiang Lau
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System , 5 Science Drive 2, Singapore 117545
| | - Vincent T Chow
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology (SMART) , 1 CREATE Way, Singapore 138602
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System , 5 Science Drive 2, Singapore 117545
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System , 16 Medical Drive, Singapore 117600
- Immunology Program, Life Science Institute, National University of Singapore , 28 Medical Drive, Singapore 117456
| |
Collapse
|
39
|
Chiu S, Fernandez R, Subramanian V, Sun H, DeCamp MM, Kreisel D, Perlman H, Budinger GRS, Mohanakumar T, Bharat A. Lung Injury Combined with Loss of Regulatory T Cells Leads to De Novo Lung-Restricted Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2016; 197:51-7. [PMID: 27194786 DOI: 10.4049/jimmunol.1502539] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/21/2016] [Indexed: 01/02/2023]
Abstract
More than one third of patients with chronic lung disease undergoing lung transplantation have pre-existing Abs against lung-restricted self-Ags, collagen type V (ColV), and k-α1 tubulin (KAT). These Abs can also develop de novo after lung transplantation and mediate allograft rejection. However, the mechanisms leading to lung-restricted autoimmunity remain unknown. Because these self-Ags are normally sequestered, tissue injury is required to expose them to the immune system. We previously showed that respiratory viruses can induce apoptosis in CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs), the key mediators of self-tolerance. Therefore, we hypothesized that lung-tissue injury can lead to lung-restricted immunity if it occurs in a setting when Tregs are impaired. We found that human lung recipients who suffer respiratory viral infections experienced a decrease in peripheral Tregs. Pre-existing lung allograft injury from donor-directed Abs or gastroesophageal reflux led to new ColV and KAT Abs post respiratory viral infection. Similarly, murine parainfluenza (Sendai) respiratory viral infection caused a decrease in Tregs. Intratracheal instillation of anti-MHC class I Abs, but not isotype control, followed by murine Sendai virus infection led to development of Abs against ColV and KAT, but not collagen type II (ColII), a cartilaginous protein. This was associated with expansion of IFN-γ-producing CD4(+) T cells specific to ColV and KAT, but not ColII. Intratracheal anti-MHC class I Abs or hydrochloric acid in Foxp3-DTR mice induced ColV and KAT, but not ColII, immunity, only if Tregs were depleted using diphtheria toxin. We conclude that tissue injury combined with loss of Tregs can lead to lung-tissue-restricted immunity.
Collapse
Affiliation(s)
- Stephen Chiu
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Ramiro Fernandez
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | | | - Haiying Sun
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Malcolm M DeCamp
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - Daniel Kreisel
- Washington University School of Medicine, St. Louis, MO 63110
| | - Harris Perlman
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | - G R Scott Budinger
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| | | | - Ankit Bharat
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611; and
| |
Collapse
|
40
|
Grunig G, Baghdassarian A, Park SH, Pylawka S, Bleck B, Reibman J, Berman-Rosenzweig E, Durmus N. Challenges and Current Efforts in the Development of Biomarkers for Chronic Inflammatory and Remodeling Conditions of the Lungs. Biomark Insights 2016; 10:59-72. [PMID: 26917944 PMCID: PMC4756863 DOI: 10.4137/bmi.s29514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/14/2015] [Accepted: 10/18/2015] [Indexed: 02/06/2023] Open
Abstract
This review discusses biomarkers that are being researched for their usefulness to phenotype chronic inflammatory lung diseases that cause remodeling of the lung's architecture. The review focuses on asthma, chronic obstructive pulmonary disease (COPD), and pulmonary hypertension. Bio-markers of environmental exposure and specific classes of biomarkers (noncoding RNA, metabolism, vitamin, coagulation, and microbiome related) are also discussed. Examples of biomarkers that are in clinical use, biomarkers that are under development, and biomarkers that are still in the research phase are discussed. We chose to present examples of the research in biomarker development by diseases, because asthma, COPD, and pulmonary hypertension are distinct entities, although they clearly share processes of inflammation and remodeling.
Collapse
Affiliation(s)
- Gabriele Grunig
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.; Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Aram Baghdassarian
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Sung-Hyun Park
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| | - Serhiy Pylawka
- College of Dental Medicine, Columbia University, New York, NY, USA
| | - Bertram Bleck
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Joan Reibman
- Department of Medicine, New York University School of Medicine, New York, NY, USA
| | | | - Nedim Durmus
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
41
|
Nurwidya F, Damayanti T, Yunus F. The Role of Innate and Adaptive Immune Cells in the Immunopathogenesis of Chronic Obstructive Pulmonary Disease. Tuberc Respir Dis (Seoul) 2016; 79:5-13. [PMID: 26770229 PMCID: PMC4701795 DOI: 10.4046/trd.2016.79.1.5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/01/2015] [Accepted: 10/12/2015] [Indexed: 01/19/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic and progressive inflammatory disease of the airways and lungs that results in limitations of continuous airflow and is caused by exposure to noxious gasses and particles. A major cause of morbidity and mortality in adults, COPD is a complex disease pathologically mediated by many inflammatory pathways. Macrophages, neutrophils, dendritic cells, and CD8+ T-lymphocytes are the key inflammatory cells involved in COPD. Recently, the non-coding small RNA, micro-RNA, have also been intensively investigated and evidence suggest that it plays a role in the pathogenesis of COPD. Here, we discuss the accumulated evidence that has since revealed the role of each inflammatory cell and their involvement in the immunopathogenesis of COPD. Mechanisms of steroid resistance in COPD will also be briefly discussed.
Collapse
Affiliation(s)
- Fariz Nurwidya
- Department of Respiratory Medicine, Persahabatan General Hospital, University of Indonesia Faculty of Medicine, Jakarta, Indonesia
| | - Triya Damayanti
- Department of Respiratory Medicine, Persahabatan General Hospital, University of Indonesia Faculty of Medicine, Jakarta, Indonesia
| | - Faisal Yunus
- Department of Respiratory Medicine, Persahabatan General Hospital, University of Indonesia Faculty of Medicine, Jakarta, Indonesia
| |
Collapse
|