1
|
Sakurai K, Nakayama S, Chubachi S, Otake S, Shimada T, Irie H, Tsutsumi A, Kameyama N, Hegab AE, Shimoda M, Hamamoto J, Terai H, Yasuda H, Kanai Y, Fukunaga K. Roflumilast reduces the number of lung adenocarcinomas, inflammation, and emphysema in a smoking-induced mouse model. BMC Pulm Med 2025; 25:262. [PMID: 40420271 PMCID: PMC12105171 DOI: 10.1186/s12890-025-03730-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/15/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND The prognosis of lung cancer complicated by chronic obstructive pulmonary disease is poor, and effective prophylactic agents have not been established. Given that inflammation is a shared pathogenic mechanism of both diseases, we aimed to evaluate the efficacy of roflumilast, a novel anti-inflammatory drug, in preventing emphysema and lung cancer using a smoking-induced lung cancer mouse model. METHODS Male A/J mice were exposed to 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, a potent carcinogen, and intermittent mainstream cigarette smoke for 20 weeks. Roflumilast or vehicle was administered via intragastric gavage once daily. Lung tissues were assessed for tumor nodules and emphysema, and bronchoalveolar lavage fluid was collected for cell counting. Emphysema severity and concentrations of inflammatory cytokines (IL-6, IL-1β, and TNF-α) were assessed. RAW 264.7 macrophage cells were used to assess cellular responses to cigarette smoke extract. RESULTS Roflumilast attenuated the increase in total cells and macrophages in bronchoalveolar lavage fluid induced by intermittent smoking exposure and significantly suppressed smoking-induced expressions of IL-6, IL-1β, and TNF-α. Roflumilast also reduced emphysematous changes and the number of lung tumors. In vitro, roflumilast attenuated cigarette smoke extract-induced expression of IL-6, IL-1β, and TNF-α in RAW 264.7 cells. CONCLUSIONS This study highlights the potential use of roflumilast as a chemopreventive agent for patients with chronic obstructive pulmonary disease who are at risk of lung cancer and underscores its relevance for future clinical application and research on phosphodiesterase-4 inhibitors.
Collapse
Affiliation(s)
- Kaori Sakurai
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shingo Nakayama
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Shiro Otake
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takashi Shimada
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidehiro Irie
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Akihiro Tsutsumi
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naofumi Kameyama
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ahmed E Hegab
- Medical Education Center, School of Medicine, International University of Health and Welfare, 4-3 Kozunomori, Narita, Chiba, 286-8686, Japan
| | - Masayuki Shimoda
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
2
|
Wang T, Zhou D, Hong Z. Sarcopenia and cachexia: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2025; 6:e70030. [PMID: 39764565 PMCID: PMC11702502 DOI: 10.1002/mco2.70030] [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: 07/16/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 03/17/2025] Open
Abstract
Sarcopenia is defined as a muscle-wasting syndrome that occurs with accelerated aging, while cachexia is a severe wasting syndrome associated with conditions such as cancer and immunodeficiency disorders, which cannot be fully addressed through conventional nutritional supplementation. Sarcopenia can be considered a component of cachexia, with the bidirectional interplay between adipose tissue and skeletal muscle potentially serving as a molecular mechanism for both conditions. However, the underlying mechanisms differ. Recognizing the interplay and distinctions between these disorders is essential for advancing both basic and translational research in this area, enhancing diagnostic accuracy and ultimately achieving effective therapeutic solutions for affected patients. This review discusses the muscle microenvironment's changes contributing to these conditions, recent therapeutic approaches like lifestyle modifications, small molecules, and nutritional interventions, and emerging strategies such as gene editing, stem cell therapy, and gut microbiome modulation. We also address the challenges and opportunities of multimodal interventions, aiming to provide insights into the pathogenesis and molecular mechanisms of sarcopenia and cachexia, ultimately aiding in innovative strategy development and improved treatments.
Collapse
Affiliation(s)
- Tiantian Wang
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Dong Zhou
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| | - Zhen Hong
- Department of NeurologyWest China Hospital of Sichuan UniversityChengduSichuanChina
- Institute of Brain Science and Brain‐Inspired Technology of West China HospitalSichuan UniversityChengduSichuanChina
- Department of NeurologyChengdu Shangjin Nanfu HospitalChengduSichuanChina
| |
Collapse
|
3
|
Abbasi A, Wang D, Stringer WW, Casaburi R, Rossiter HB. Immune system benefits of pulmonary rehabilitation in chronic obstructive pulmonary disease. Exp Physiol 2024. [PMID: 39456127 DOI: 10.1113/ep091678] [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: 06/03/2024] [Accepted: 09/04/2024] [Indexed: 10/28/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease characterized by pulmonary and systemic inflammation. Inflammatory mediators show relationships with shortness of breath, exercise intolerance and health related quality of life. Pulmonary rehabilitation (PR), a comprehensive education and exercise training programme, is the most effective therapy for COPD and is associated with reduced exacerbation and hospitalization rates and increased survival. Exercise training, the primary physiological intervention within PR, is known to exert a beneficial anti-inflammatory effect in health and chronic diseases. The question of this review article is whether exercise training can also make such a beneficial anti-inflammatory effect in COPD. Experimental studies using smoke exposure mice models suggest that the response of the immune system to exercise training is favourably anti-inflammatory. However, the evidence about the response of most known inflammatory mediators (C-reactive protein, tumour necrosis factor α, interleukin 6, interleukin 10) to exercise training in COPD patients is inconsistent, making it difficult to conclude whether regular exercise training has an anti-inflammatory effect in COPD. It is also unclear whether COPD patients with more persistent inflammation are a subgroup that would benefit more from hypothesized immunomodulatory effects of exercise training (i.e., personalized treatment). Nevertheless, it seems that PR combined with maintenance exercise training (i.e., lifestyle change) might be more beneficial in controlling inflammation and slowing disease progress in COPD patients, specifically in those with early stages of disease.
Collapse
Affiliation(s)
- Asghar Abbasi
- Institute of Respiratory Medicine and Exercise Physiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - David Wang
- Institute of Respiratory Medicine and Exercise Physiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - William W Stringer
- Institute of Respiratory Medicine and Exercise Physiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Richard Casaburi
- Institute of Respiratory Medicine and Exercise Physiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Harry B Rossiter
- Institute of Respiratory Medicine and Exercise Physiology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| |
Collapse
|
4
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [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: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
5
|
Oh K, Lee GW, Kim HB, Park JH, Shin EY, Kim EG. Regorafenib prevents the development of emphysema in a murine elastase model. BMB Rep 2023; 56:439-444. [PMID: 37357536 PMCID: PMC10471461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/02/2023] [Accepted: 06/26/2023] [Indexed: 06/27/2023] Open
Abstract
Emphysema is a chronic obstructive lung disease characterized by inflammation and enlargement of the air spaces. Regorafenib, a potential senomorphic drug, exhibited a therapeutic effect in porcine pancreatic elastase (PPE)-induced emphysema in mice. In the current study we examined the preventive role of regorafenib in development of emphysema. Lung function tests and morphometry showed that oral administration of regorafenib (5 mg/kg/day) for seven days after instillation of PPE resulted in attenuation of emphysema. Mechanistically, regorafenib reduced the recruitment of inflammatory cells, particularly macrophages and neutrophils, in bronchoalveolar lavage fluid. In agreement with these findings, measurements using a cytokine array and ELISA showed that expression of inflammatory mediators including interleukin (IL)-1β, IL-6, and CXCL1/KC, and tissue inhibitor of matrix metalloprotease-1 (TIMP-1), was downregulated. The results of immunohistochemical analysis confirmed that expression of IL-6, CXCL1/KC, and TIMP-1 was reduced in the lung parenchyma. Collectively, the results support the preventive role of regorafenib in development of emphysema in mice and provide mechanistic insights into prevention strategies. [BMB Reports 2023; 56(8): 439-444].
Collapse
Affiliation(s)
- Kwangseok Oh
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Gun-Wu Lee
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Han-Byeol Kim
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Jin-Hee Park
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Eun-Young Shin
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| | - Eung-Gook Kim
- Department of Biochemistry, College of Medicine, and Medical Research Center, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
6
|
Rodriguez-Herrera AJ, de Souza ABF, Castro TDF, Machado-Junior PA, Marcano-Gomez EC, Menezes TP, Castro MLDC, Talvani A, Costa DC, Cangussú SD, Bezerra FS. Long-term e-cigarette aerosol exposure causes pulmonary emphysema in adult female and male mice. Regul Toxicol Pharmacol 2023; 142:105412. [PMID: 37247649 DOI: 10.1016/j.yrtph.2023.105412] [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: 12/27/2022] [Revised: 05/14/2023] [Accepted: 05/21/2023] [Indexed: 05/31/2023]
Abstract
This study aimed to evaluate long-term exposure to conventional cigarette smoke (CC) and electronic cigarette (EC) aerosol in adult male and female C57BL/6 mice. Forty-eight C57BL/6 mice were used, male (n = 24) and female (n = 24), both were divided into three groups: control, CC and EC. The CC and EC groups were exposed to cigarette smoke or electronic cigarette aerosol, respectively, 3 times a day for 60 consecutive days. Afterwards, they were maintained for 60 days without exposure to cigarettes or electronic cigarette aerosol. Both cigarettes promoted an influx of inflammatory cells to the lung in males and females. All animals exposed to CC and EC showed an increase in lipid peroxidation and protein oxidation. There was an increase of IL-6 in males and females exposed to EC. The IL-13 levels were higher in the females exposed to EC and CC. Both sexes exposed to EC and CC presented tissue damage characterized by septal destruction and increased alveolar spaces compared to control. Our results demonstrated that exposure to CC and EC induced pulmonary emphysema in both sexes, and females seem to be more susceptible to EC.
Collapse
Affiliation(s)
- Andrea Jazel Rodriguez-Herrera
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Pedro Alves Machado-Junior
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Elena Cecilia Marcano-Gomez
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Tatiana Prata Menezes
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Maria Laura da Cruz Castro
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Daniela Caldeira Costa
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology, Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto (UFOP), 35400-000, Ouro Preto, MG, Brazil.
| |
Collapse
|
7
|
Cross-talk between IL-6 trans-signaling and AIM2 inflammasome/IL-1β axes bridge innate immunity and epithelial apoptosis to promote emphysema. Proc Natl Acad Sci U S A 2022; 119:e2201494119. [PMID: 36037355 PMCID: PMC9457334 DOI: 10.1073/pnas.2201494119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pulmonary emphysema is associated with dysregulated innate immune responses that promote chronic pulmonary inflammation and alveolar apoptosis, culminating in lung destruction. However, the molecular regulators of innate immunity that promote emphysema are ill-defined. Here, we investigated whether innate immune inflammasome complexes, comprising the adaptor ASC, Caspase-1 and specific pattern recognition receptors (PRRs), promote the pathogenesis of emphysema. In the lungs of emphysematous patients, as well as spontaneous gp130F/F and cigarette smoke (CS)-induced mouse models of emphysema, the expression (messenger RNA and protein) and activation of ASC, Caspase-1, and the inflammasome-associated PRR and DNA sensor AIM2 were up-regulated. AIM2 up-regulation in emphysema coincided with the biased production of the mature downstream inflammasome effector cytokine IL-1β but not IL-18. These observations were supported by the genetic blockade of ASC, AIM2, and the IL-1 receptor and therapy with AIM2 antagonistic suppressor oligonucleotides, which ameliorated emphysema in gp130F/F mice by preventing elevated alveolar cell apoptosis. The functional requirement for AIM2 in driving apoptosis in the lung epithelium was independent of its expression in hematopoietic-derived immune cells and the recruitment of infiltrating immune cells in the lung. Genetic and inhibitor-based blockade of AIM2 also protected CS-exposed mice from pulmonary alveolar cell apoptosis. Intriguingly, IL-6 trans-signaling via the soluble IL-6 receptor, facilitated by elevated levels of IL-6, acted upstream of the AIM2 inflammasome to augment AIM2 expression in emphysema. Collectively, we reveal cross-talk between the AIM2 inflammasome/IL-1β and IL-6 trans-signaling axes for potential exploitation as a therapeutic strategy for emphysema.
Collapse
|
8
|
Li Y, Gao H, Zhao L, Wang J. Osteoporosis in COPD patients: Risk factors and pulmonary rehabilitation. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:487-496. [PMID: 35688435 PMCID: PMC9329018 DOI: 10.1111/crj.13514] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/15/2022]
Abstract
Objectives To present a review on the pathogenesis, risk factor and treatment of chronic obstructive pulmonary disease complicated with osteoporosis and provide new ideas for the diagnosis and treatment. Data source A systematic search is carried out using keywords as chronic obstructive pulmonary disease, osteoporosis, risk factors, and pulmonary rehabilitation. Results Patients with chronic obstructive pulmonary disease have a high prevalence of osteoporosis and a high risk of fracture. The mechanisms of osteoporosis in COPD patients are associated with general risk factors, such as smoking, reduced physical activity, low weight, and disease‐specific risk factors, such as systemic inflammatory, Vitamin D deficiency, use of glucocorticoid, anemia, hypoxemia, and hypercapnia. The treatment of osteoporosis in COPD emphasizes comprehensive intervention, which mainly include basic treatment and anti‐osteoporosis drugs. Noticeably, pulmonary rehabilitation program is an important part of treatment. Conclusions This work summarizes the pathogenesis, risk factor, prevention, and treatment of chronic obstructive pulmonary disease complicated with osteoporosis, and the latest progress of studies on chronic obstructive pulmonary disease and osteoporosis is discussed.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Hongchang Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Lei Zhao
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Jinrui Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| |
Collapse
|
9
|
Peng J, Cai Z, Wang Q, Zhou J, Xu J, Pan D, Chen T, Zhang G, Tao L, Chen Y, Shen X. Carboxymethyl Chitosan Modified Oxymatrine Liposomes for the Alleviation of Emphysema in Mice via Pulmonary Administration. Molecules 2022; 27:3610. [PMID: 35684546 PMCID: PMC9182538 DOI: 10.3390/molecules27113610] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023] Open
Abstract
Pulmonary emphysema is a fatal lung disease caused by the progressive thinning, enlargement and destruction of alveoli that is closely related to inflammation and oxidative stress. Oxymatrine (OMT), as a bioactive constituent of traditional Chinese herbal Sophora flavescens, has great potential to alleviate pulmonary emphysema via its anti-inflammatory and antioxidative activities. Pulmonary administration is the most preferable way for the treatment of lung diseases. To improve the in vivo stability and pulmonary retention of OMT, OMT-loaded liposome with carboxymethyl chitosan (CMCS) modification was developed. The CMCS was modified on the surface of OMT liposomes via electrostatic attraction and covalent conjugation to obtain Lipo/OMT@CMCS and CMCS-Lipo/OMT, respectively. A porcine pancreatic elastase (PPE)-induced emphysema mice model was established to evaluate the alleviation effects of OMT on alveolar expansion and destruction. CMCS-modified liposomal OMT exhibited superior ameliorative effects on emphysema regardless of the preparation methods, and higher sedimentation and longer retention in the lung were observed in the CMCS-Lipo group. The mechanisms of OMT on emphysema were related to the downregulation of inflammatory cytokines and the rebalancing of antioxidant/oxidation via the Nrf2/HO-1 and NF-κB/IκB-α signaling pathways, leading to reduced cell apoptosis. Moreover, the OMT liposomal preparations further enhanced its anti-inflammatory and antioxidative effects. In conclusion, pulmonary administration of OMT is a potential strategy for the treatment of emphysema and the therapeutic effects can be further improved by CMCS-modified liposomes.
Collapse
Affiliation(s)
- Jianqing Peng
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Zimin Cai
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Qin Wang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Jia Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Jinzhuan Xu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Di Pan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Tingting Chen
- Guiyang Maternal and Child Health Care Hospital, Guiyang 550003, China;
| | - Guangqiong Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Ling Tao
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Yi Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China; (J.P.); (Z.C.); (Q.W.); (J.Z.); (J.X.); (D.P.); (G.Z.); (L.T.)
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| |
Collapse
|
10
|
Chitosan-Coated Solid Lipid Nano-Encapsulation Improves the Therapeutic Antiairway Inflammation Effect of Berberine against COPD in Cigarette Smoke-Exposed Rats. Can Respir J 2022; 2022:8509396. [PMID: 35465190 PMCID: PMC9033382 DOI: 10.1155/2022/8509396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Berberine (Ber) is an isoquinoline alkaloid that has shown therapeutic potential in mice with chronic obstructive pulmonary disease (COPD). However, the therapeutic efficiency of Ber is restricted by its low aqueous solubility and bioavailability. Chitosan and solid lipid nanoparticles (SLNs) have demonstrated great abilities as delivery systems in enhancing the bioavailability of therapeutic compounds. The present study aimed to get together the biological features of SLNs with the advantages of chitosan to formulate an efficient nano-carrier platform for the oral delivery of Ber and evaluate the therapeutic effect of the prepared Ber-encapsulated nanoparticles on airway inflammation in cigarette smoke (CS)-induced COPD rats. The Ber-encapsulated SLE-chitosan formulation was manufactured using a modified solvent-injection method followed by a homogenization process. Physicochemical properties, encapsulation efficiency, in vitro stability and Ber release, and pharmacokinetics of the manufactured formulation were evaluated. The COPD rat model was developed by exposing animals to CS. To study the therapeutic efficiency of Ber-encapsulated SLE-chitosan nanoparticles and pure berberine, the histopathological changes of the lung tissues, levels of inflammatory cells and cytokines, and activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) enzymes were evaluated in bronchoalveolar lavage fluid (BALF). Ber-encapsulated SLE-chitosan showed the particle size in nano-range with high stability and controlled slow-release profile in vitro in simulated gastric (pH 1.5) and intestinal (pH 6.8) fluids. Administration of Ber-loaded SLE-chitosan nanoparticles could significantly ameliorate inflammation scores in lung tissues and reduce levels of inflammatory cells (neutrophils and macrophages) and inflammatory cytokines (IL-1β, Il-6, Il-17, and TNFα) in BALF when compared with the pure Ber. SLE-chitosan-based nanoparticles can strongly improve the therapeutic anti-inflammatory impact of Ber against CS-induced airway inflammation in COPD rats, suggesting the promising application of Ber-encapsulated SLN-chitosan nanoparticles for treating COPD and other inflammation-mediated diseases.
Collapse
|
11
|
Alhayyani S, McLeod L, West AC, Balic JJ, Hodges C, Yu L, Smith JA, Prodanovic Z, Bozinovski S, Kumar B, Ruwanpura SM, Saad MI, Jenkins BJ. Oncogenic dependency on STAT3 serine phosphorylation in KRAS mutant lung cancer. Oncogene 2022; 41:809-823. [PMID: 34857889 DOI: 10.1038/s41388-021-02134-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/15/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023]
Abstract
The oncogenic potential of the latent transcription factor signal transducer and activator of transcription (STAT)3 in many human cancers, including lung cancer, has been largely attributed to its nuclear activity as a tyrosine-phosphorylated (pY705 site) transcription factor. By contrast, an alternate mitochondrial pool of serine phosphorylated (pS727 site) STAT3 has been shown to promote tumourigenesis by regulating metabolic processes, although this has been reported in only a restricted number of mutant RAS-addicted neoplasms. Therefore, the involvement of STAT3 serine phosphorylation in the pathogenesis of most cancer types, including mutant KRAS lung adenocarcinoma (LAC), is unknown. Here, we demonstrate that LAC is suppressed in oncogenic KrasG12D-driven mouse models engineered for pS727-STAT3 deficiency. The proliferative potential of the transformed KrasG12D lung epithelium, and mutant KRAS human LAC cells, was significantly reduced upon pS727-STAT3 deficiency. Notably, we uncover the multifaceted capacity of constitutive pS727-STAT3 to metabolically reprogramme LAC cells towards a hyper-proliferative state by regulating nuclear and mitochondrial (mt) gene transcription, the latter via the mtDNA transcription factor, TFAM. Collectively, our findings reveal an obligate requirement for the transcriptional activity of pS727-STAT3 in mutant KRAS-driven LAC with potential to guide future therapeutic targeting approaches.
Collapse
Affiliation(s)
- Sultan Alhayyani
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
- Department of Chemistry, College of Sciences and Arts, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Alison C West
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Jesse J Balic
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Christopher Hodges
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Liang Yu
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Julian A Smith
- Department of Surgery, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, 3168, Australia
- Department of Cardiothoracic Surgery, Monash Health, Clayton, Victoria, 3168, Australia
| | | | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3082, Australia
| | - Beena Kumar
- Department of Anatomical Pathology, Monash Health, Clayton, Victoria, 3168, Australia
| | - Saleela M Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, 3168, Australia.
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria, 3168, Australia.
| |
Collapse
|
12
|
Ghio AJ, Pavlisko EN, Roggli VL, Todd NW, Sangani RG. Cigarette Smoke Particle-Induced Lung Injury and Iron Homeostasis. Int J Chron Obstruct Pulmon Dis 2022; 17:117-140. [PMID: 35046648 PMCID: PMC8763205 DOI: 10.2147/copd.s337354] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
It is proposed that the mechanistic basis for non-neoplastic lung injury with cigarette smoking is a disruption of iron homeostasis in cells after exposure to cigarette smoke particle (CSP). Following the complexation and sequestration of intracellular iron by CSP, the host response (eg, inflammation, mucus production, and fibrosis) attempts to reverse a functional metal deficiency. Clinical manifestations of this response can present as respiratory bronchiolitis, desquamative interstitial pneumonitis, pulmonary Langerhans’ cell histiocytosis, asthma, pulmonary hypertension, chronic bronchitis, and pulmonary fibrosis. If the response is unsuccessful, the functional deficiency of iron progresses to irreversible cell death evident in emphysema and bronchiectasis. The subsequent clinical and pathological presentation is a continuum of lung injuries, which overlap and coexist with one another. Designating these non-neoplastic lung injuries after smoking as distinct disease processes fails to recognize shared relationships to each other and ultimately to CSP, as well as the common mechanistic pathway (ie, disruption of iron homeostasis).
Collapse
Affiliation(s)
- Andrew J Ghio
- Human Studies Facility, US Environmental Protection Agency, Chapel Hill, NC, 27514, USA
- Correspondence: Andrew J Ghio Human Studies Facility, US Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC, USA Email
| | | | | | - Nevins W Todd
- Department of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Rahul G Sangani
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
13
|
Dobric A, De Luca SN, Spencer SJ, Bozinovski S, Saling MM, McDonald CF, Vlahos R. Novel pharmacological strategies to treat cognitive dysfunction in chronic obstructive pulmonary disease. Pharmacol Ther 2021; 233:108017. [PMID: 34626675 DOI: 10.1016/j.pharmthera.2021.108017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/19/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable global health burden and currently the 3rd largest cause of death in the world, with approximately 3.23 million deaths per year. Globally, the financial burden of COPD is approximately €82 billion per year and causes substantial morbidity and mortality. Importantly, much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and viral and bacterial-induced acute exacerbations (AECOPD). Recent clinical studies have shown that cognitive dysfunction is present in up to 60% of people with COPD, with impairments in executive function, memory, and attention, impacting on important outcomes such as quality of life, hospitalisation and survival. The high prevalence of cognitive dysfunction in COPD may also help explain the insufficient adherence to therapeutic plans and strategies, thus worsening disease progression in people with COPD. However, the mechanisms underlying the impaired neuropathology and cognition in COPD remain largely unknown. In this review, we propose that the observed pulmonary oxidative burden and inflammatory response of people with COPD 'spills over' into the systemic circulation, resulting in damage to the brain and leading to cognitive dysfunction. As such, drugs targeting the lungs and comorbidities concurrently represent an exciting and unique therapeutic opportunity to treat COPD and cognitive impairments, which may lead to the production of novel targets to prevent and reverse the debilitating and life-threatening effects of cognitive dysfunction in COPD.
Collapse
Affiliation(s)
- Aleksandar Dobric
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Simone N De Luca
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia; ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia
| | - Steven Bozinovski
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Michael M Saling
- Clinical Neuropsychology, The University of Melbourne and Austin Health, VIC, Australia
| | - Christine F McDonald
- Institute for Breathing and Sleep, Austin Health, Melbourne, VIC, Australia; Department of Respiratory & Sleep Medicine, The University of Melbourne and Austin Health, Melbourne, VIC, Australia
| | - Ross Vlahos
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Sin S, Choi HM, Lim J, Kim J, Bak SH, Choi SS, Park J, Lee JH, Oh YM, Lee MK, Hobbs BD, Cho MH, Silverman EK, Kim WJ. A genome-wide association study of quantitative computed tomographic emphysema in Korean populations. Sci Rep 2021; 11:16692. [PMID: 34404834 PMCID: PMC8371078 DOI: 10.1038/s41598-021-95887-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/28/2021] [Indexed: 11/18/2022] Open
Abstract
Emphysema is an important feature of chronic obstructive pulmonary disease (COPD). Genetic factors likely affect emphysema pathogenesis, but this question has predominantly been studied in those of European ancestry. In this study, we sought to determine genetic components of emphysema severity and characterize the potential function of the associated loci in Korean population. We performed a genome-wide association study (GWAS) on quantitative emphysema in subjects with or without COPD from two Korean COPD cohorts. We investigated the functional consequences of the loci using epigenetic annotation and gene expression data. We also compared our GWAS results with an epigenome-wide association study and previous differential gene expression analysis. In total, 548 subjects (476 [86.9%] male) including 514 COPD patients were evaluated. We identified one genome-wide significant SNP (P < 5.0 × 10-8), rs117084279, near PIBF1. We identified an additional 57 SNPs (P < 5.0 × 10-6) associated with emphysema in all subjects, and 106 SNPs (P < 5.0 × 10-6) in COPD patients. Of these candidate SNPs, 2 (rs12459249, rs11667314) near CYP2A6 were expression quantitative trait loci in lung tissue and a SNP (rs11214944) near NNMT was an expression quantitative trait locus in whole blood. Of note, rs11214944 was in linkage disequilibrium with variants in enhancer histone marks in lung tissue. Several genes near additional SNPs were identified in our previous EWAS study with nominal level of significance. We identified a novel SNP associated with quantitative emphysema on CT. Including the novel SNP, several candidate SNPs in our study may provide clues to the genetic etiology of emphysema in Asian populations. Further research and validation of the loci will help determine the genetic factors for the development of emphysema.
Collapse
Affiliation(s)
- Sooim Sin
- grid.412010.60000 0001 0707 9039Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Hye-Mi Choi
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jiwon Lim
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeeyoung Kim
- grid.412010.60000 0001 0707 9039Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - So Hyeon Bak
- grid.412010.60000 0001 0707 9039Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Sun Shim Choi
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jinkyeong Park
- grid.470090.a0000 0004 1792 3864Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Jin Hwa Lee
- grid.255649.90000 0001 2171 7754Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Yeon-Mok Oh
- grid.267370.70000 0004 0533 4667Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Kyeong Lee
- grid.280664.e0000 0001 2110 5790Epidemiology Branch, Division of Intramural Research, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC USA
| | - Brian D. Hobbs
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Michael H. Cho
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Edwin K. Silverman
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea.
| |
Collapse
|
15
|
Gui X, Yang Z, Li MD. Effect of Cigarette Smoke on Gut Microbiota: State of Knowledge. Front Physiol 2021; 12:673341. [PMID: 34220536 PMCID: PMC8245763 DOI: 10.3389/fphys.2021.673341] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoke is a representative source of toxic chemical exposures to humans, and the adverse consequences of cigarette smoking are mediated by its effect on both neuronal and immune-inflammatory systems. Cigarette smoking also is a major risk factor for intestinal disorders, such as Crohn's disease and peptic ulcer. On the other hand, cigarette smoking is protective against developing ulcerative colitis. The effects of cigarette smoking on intestinal disorders include changes in intestinal irrigation and microbiome, increases in permeability of the mucosa, and impaired mucosal immune responses. However, the underlying mechanism linking cigarette smoking with intestinal microbiota dysbiosis is largely unknown. In this communication, we first review the current knowledge about the mechanistic interaction between cigarette smoke and intestinal microbiota dysbiosis, which include the likely actions of nicotine, aldehydes, polycyclic aromatic hydrocarbons, heavy metals, volatile organic compounds and toxic gases, and then reveal the potential mechanisms of the lung-gut cross talk and skin-gut cross talk in regulating the balance of intestinal microbiota and the interrelation of intestinal microbiota dysbiosis and systemic disorders.
Collapse
Affiliation(s)
- Xiaohua Gui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming D. Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Dawson RE, Jenkins BJ, Saad MI. IL-6 family cytokines in respiratory health and disease. Cytokine 2021; 143:155520. [PMID: 33875334 DOI: 10.1016/j.cyto.2021.155520] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Chronic lung diseases including asthma, chronic obstructive pulmonary disease (COPD) and lung fibrosis represent a major burden on healthcare systems with limited effective therapeutic options. Developing effective treatments for these debilitating diseases requires an understanding of how alterations at the molecular level affect lung macroscopic architecture. A common theme among these lung disorders is the presence of an underlying dysregulated immune system which can lead to sustained chronic inflammation. In this respect, several inflammatory cytokines have been implicated in the pathogenesis of lung diseases, thus leading to the notion that cytokines are attractive therapeutic targets for these disorders. In this review, we discuss and highlight the recent breakthroughs that have enhanced our understanding of the role of the interleukin (IL)-6 family of cytokines in lung homeostasis and chronic diseases including asthma, COPD, lung fibrosis and lung cancer.
Collapse
Affiliation(s)
- Ruby E Dawson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
17
|
Kang HS, Bak SH, Oh HY, Lim MN, Cha YK, Yoon HJ, Kim WJ. Computed tomography-based visual assessment of chronic obstructive pulmonary disease: comparison with pulmonary function test and quantitative computed tomography. J Thorac Dis 2021; 13:1495-1506. [PMID: 33841942 PMCID: PMC8024830 DOI: 10.21037/jtd-20-3041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) has variable subtypes involving mixture of large airway inflammation, small airway disease, and emphysema. This study evaluated the relationship between visually assessed computed tomography (CT) subtypes and clinical/imaging characteristics. Methods In total, 452 participants were enrolled in this study between 2012 and 2017. Seven subtypes were defined by visual evaluation of CT images using Fleischner Society classification: normal, paraseptal emphysema (PSE), bronchial disease, and centrilobular emphysema (trace, mild, moderate and confluent/advanced destructive). The differences in several variables, including clinical, laboratory, spirometric, and quantitative CT features among CT-based visual subtypes, were compared using the chi-square tests and one-way analysis of variance. Results Subjects who had PSE had better forced expiratory volume in 1 second (FEV1) (P=0.03) percentage and higher lung density (P<0.05) than those with moderate to confluent/advanced destructive centrilobular emphysema. As the visual grade of centrilobular emphysema worsened, pulmonary function declined and modified Medical Research Council, COPD assessment test (CAT) score, and quantitative assessment (emphysema index and air trapping) increased. The bronchial subtype was associated with higher body mass index (BMI), better lung function and higher lung density. Participants with trace emphysema showed a rapid increase in functional small airway disease. Conclusions Classifying subtypes using visual CT imaging features can reflect heterogeneity and pathological processes of COPD.
Collapse
Affiliation(s)
- Han Sol Kang
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - So Hyeon Bak
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Ha Yeun Oh
- Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Myoung-Nam Lim
- Biomedical Research Institute, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Yoon Ki Cha
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Yoon
- Department of Radiology, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Woo Jin Kim
- Department of Internal Medicine and Environmental Health Center, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
18
|
Jundi B, Geraghty P. ADAM17: A Therapeutic Target for Patients with Emphysema? Am J Respir Cell Mol Biol 2021; 64:155-157. [PMID: 33522887 PMCID: PMC7874393 DOI: 10.1165/rcmb.2020-0467ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Bakr Jundi
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York
| |
Collapse
|
19
|
Saad MI, McLeod L, Hodges C, Vlahos R, Rose-John S, Ruwanpura S, Jenkins BJ. ADAM17 Deficiency Protects against Pulmonary Emphysema. Am J Respir Cell Mol Biol 2021; 64:183-195. [PMID: 33181031 DOI: 10.1165/rcmb.2020-0214oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022] Open
Abstract
Pulmonary emphysema is the major debilitating component of chronic obstructive pulmonary disease (COPD), which is a leading cause of morbidity and mortality worldwide. The ADAM17 (A disintegrin and metalloproteinase 17) protease mediates inflammation via ectodomain shedding of numerous proinflammatory cytokines, cytokine receptors, and adhesion molecules; however, its role in the pathogenesis of emphysema and COPD is poorly understood. This study aims to define the role of the protease ADAM17 in the pathogenesis of pulmonary emphysema. ADAM17 protein expression and activation was investigated in lung biopsies from patients with emphysema, as well as lungs of the emphysematous gp130F/F mouse model and an acute (4 d) cigarette smoke (CS)-induced lung pathology model. The Adam17ex/ex mice, which display significantly reduced global ADAM17 expression, were coupled with emphysema-prone gp130F/F mice to produce gp130F/F:Adam17ex/ex. Both Adam17ex/ex and wild-type mice were subjected to acute CS exposure. Histological, immunohistochemical, immunofluorescence, and molecular analyses as well as lung function tests were performed to assess pulmonary emphysema, inflammation, and alveolar cell apoptosis. ADAM17 was hyperphosphorylated in the lungs of patients with emphysema and also in emphysematous gp130F/F and CS-exposed mice. ADAM17 deficiency ameliorated the development of pulmonary emphysema in gp130F/F mice by suppressing elevated alveolar cell apoptosis. In addition, genetic blockade of ADAM17 protected mice from CS-induced pulmonary inflammation and alveolar cell apoptosis. Our study places the protease ADAM17 as a central molecular switch implicated in the development of pulmonary emphysema, which paves the way for using ADAM17 inhibitors as potential therapeutic agents to treat COPD and emphysema.
Collapse
Affiliation(s)
- Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christopher Hodges
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Saleela Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
20
|
Ng B, Cook SA, Schafer S. Interleukin-11 signaling underlies fibrosis, parenchymal dysfunction, and chronic inflammation of the airway. Exp Mol Med 2020; 52:1871-1878. [PMID: 33262481 PMCID: PMC7705429 DOI: 10.1038/s12276-020-00531-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 01/16/2023] Open
Abstract
Interleukin (IL)-11 evolved as part of the innate immune response. In the human lung, IL-11 upregulation has been associated with viral infections and a range of fibroinflammatory diseases, including idiopathic pulmonary fibrosis. Transforming growth factor-beta (TGFβ) and other disease factors can initiate an autocrine loop of IL-11 signaling in pulmonary fibroblasts, which, in a largely ERK-dependent manner, triggers the translation of profibrotic proteins. Lung epithelial cells also express the IL-11 receptor and transition into a mesenchymal-like state in response to IL-11 exposure. In mice, therapeutic targeting of IL-11 with antibodies can arrest and reverse bleomycin-induced pulmonary fibrosis and inflammation. Intriguingly, fibroblast-specific blockade of IL-11 signaling has anti-inflammatory effects, which suggests that lung inflammation is sustained, in part, through IL-11 activity in the stroma. Proinflammatory fibroblasts and their interaction with the damaged epithelium may represent an important but overlooked driver of lung disease. Initially thought of as a protective cytokine, IL-11 is now increasingly recognized as an important determinant of lung fibrosis, inflammation, and epithelial dysfunction.
Collapse
Affiliation(s)
- Benjamin Ng
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Stuart A Cook
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.,MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, London, United Kingdom.,National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Sebastian Schafer
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore, Singapore.
| |
Collapse
|
21
|
Zhang Z, Yao W, Yuan D, Huang F, Liu Y, Luo G, Hei Z. Effects of Connexin 32-Mediated Lung Inflammation Resolution During Liver Ischemia Reperfusion. Dig Dis Sci 2020; 65:2914-2924. [PMID: 31900713 DOI: 10.1007/s10620-019-06020-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatic ischemia reperfusion (HIR) leads to a lung inflammatory response and subsequent pulmonary barrier dysfunction. The gap junction communication protein connexin 32 (Cx32), which is widely expressed in the lungs, participates in intercellular signaling. This study determined whether the communication protein Cx32 could affect pulmonary inflammation caused by HIR. METHODS Mice were randomly allocated into four groups (n = 8/group): (i) Cx32+/+ sham group; (ii) Cx32+/+ HIR model group; (iii) Cx32-/- sham group; and (iv) Cx32-/- HIR model group. Twenty-four hours after surgery, lung tissues were collected for bright field microscopy, western blot (Cx32, JAK2, p-JAK2, STAT3, p-STAT3), and immunofluorescence (ZO-1, 8-OHDG) analyses. The collected bronchoalveolar fluid was tested for levels of interleukin-6 (IL-6), matrix metalloproteinase 12 (MMP-12), and antitrypsin (α1-AT). Lung mmu-miR-26a/b expression was detected using a PCR assay. RESULTS Increased expression of Cx32 mRNA and protein was noted in the lungs after HIR. Cx32 deletion significantly aggravated pulmonary function from acute lung injury induced by HIR. In addition, Cx32 deletion decreased the protein level of ZO-1 (pulmonary function) and increased the level of the oxidative stress marker 8-OHDG in the lungs. Moreover, in the Cx32-/- HIR model group, the levels of IL-6 and MMP-12 in bronchoalveolar lavage fluid were significantly increased leading to activation of the JAK2/STAT3 pathway, and decreased α1-AT levels. Furthermore, we found mmu-miR-26a/b was significantly downregulated in the Cx32-/- HIR model group. CONCLUSION HIR leads to acute lung inflammatory injury. Cx32 deletion aggravates hepatic-derived lung inflammation, partly through blocking the transferring of mmu-miR-26a/b and leading to IL-6-related JAK2/STAT3 pathway activation.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Weifeng Yao
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Dongdong Yuan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Fei Huang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yue Liu
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
22
|
Bon J, Nouraie SM, Smith KJ, Dransfield MT, McDonald ML, Hoffman EA, Newell JD, Comellas AP, Saha PK, Bowler RP, Regan EA. Lung-Specific Risk Factors Associated With Incident Hip Fracture in Current and Former Smokers. J Bone Miner Res 2020; 35:1952-1961. [PMID: 32754944 PMCID: PMC8773459 DOI: 10.1002/jbmr.4103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 05/14/2020] [Accepted: 06/01/2020] [Indexed: 11/06/2022]
Abstract
Hip fractures are associated with significant morbidity and mortality in smokers with lung disease, but whether lung-specific factors are associated with fracture risk is unknown. Our goal was to determine whether lung-specific factors associate with incident hip fracture and improve risk discrimination of traditional fracture risk models in smokers. The analysis consisted of a convenience sample of 9187 current and former smokers (58,477 participant follow-up years) participating in the Genetic Epidemiology of chronic obstructive pulmonary disease (COPD) longitudinal observational cohort study. Participants were enrolled between 2008 and 2011 with follow-up data collection through July 2018. Traditional risk factors associated with incident hip fracture (n = 361) included age, female sex, osteoporosis, prevalent spine and hip fracture, rheumatoid arthritis, and diabetes. Lung-specific risk factors included post-bronchodilator percent forced expiratory volume in 1 s (FEV1 %) predicted (OR, 0.95; 95% CI, 0.92-0.99 for each 10% increase), Global Initiative for Chronic Obstructive Lung Disease (GOLD) classification (OR, 1.09; 95% CI, 1.002-1.19 for each higher stage), presence of CT-determined emphysema (OR, 1.34; 95% CI, 1.06-1.69), symptom scores (OR, 1.10; 95% CI, 1.03-1.19 for each higher unit score), 6-min walk distance (OR, 0.92; 95% CI, 0.90-0.95 for each 30-m increase), body mass index, airflow obstruction, dyspnea, and exercise (BODE) index (OR, 1.07; 95% CI, 1.01-1.13 for each higher unit score), total exacerbations (OR, 1.13; 95% CI, 1.10-1.16 per exacerbation), and annual exacerbations (OR, 1.37; 95% CI, 1.21-1.55 per exacerbation). In multivariable modeling, age, black race, osteoporosis, prevalent hip and spine fracture, rheumatoid arthritis, and diabetes were associated with incident hip fracture. The presence of emphysema, 6-min walk distance, and total number of exacerbations added to traditional models improved risk discrimination (integrated discrimination improvement [IDI] values 0.001 [95% CI, 0.0003-0.002], 0.001 [95% CI, 0.0001-0.002], and 0.008 [95% CI, 0.003-0.013], corresponding to relative IDIs of 12.8%, 6.3%, and 34.6%, respectively). These findings suggest that the incorporation of lung-specific risk factors into fracture risk assessment tools may more accurately predict fracture risk in smokers. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jessica Bon
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine and VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seyed Mehdi Nouraie
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kenneth J Smith
- Department of Medicine, Division of General Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark T Dransfield
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Lung Health Center, Birmingham, AL, USA
| | - Merry-Lynn McDonald
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Lung Health Center, Birmingham, AL, USA
| | - Eric A Hoffman
- Department of Radiology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - John D Newell
- Department of Radiology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Alejandro P Comellas
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa, Iowa City, IA, USA
| | - Punam K Saha
- Department of Radiology, University of Iowa, Iowa City, IA, USA.,Department of Internal Medicine, University of Iowa, Iowa City, IA, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Russell P Bowler
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Elizabeth A Regan
- Department of Medicine, Division of Rheumatology, National Jewish Health, Denver, CO, USA
| |
Collapse
|
23
|
Gonzalez J, Rivera-Ortega P, Rodríguez-Fraile M, Restituto P, Colina I, Calleja MDLD, Alcaide AB, Campo A, Bertó J, Seijo L, Pérez-Warnisher MT, Zulueta JJ, Varo N, de-Torres JP. Exploring the Association Between Emphysema Phenotypes and Low Bone Mineral Density in Smokers with and without COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1823-1829. [PMID: 32801680 PMCID: PMC7401322 DOI: 10.2147/copd.s257918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022] Open
Abstract
Rationale Emphysema and osteoporosis are tobacco-related diseases. Many studies have shown that emphysema is a strong and independent predictor of low bone mineral density (BMD) in smokers; however, none of them explored its association with different emphysema subtypes. Objective To explore the association between the different emphysema subtypes and the presence of low bone mineral density in a population of active or former smokers with and without chronic obstructive pulmonary disease (COPD). Methods One hundred and fifty-three active and former smokers from a pulmonary clinic completed clinical questionnaires, pulmonary function tests, a low-dose chest computed tomography (LDCT) and a dual-energy absorptiometry (DXA) scans. Subjects were classified as having normal BMD or low BMD (osteopenia or osteoporosis). Emphysema was classified visually for its subtype and severity. Logistic regression analysis explored the relationship between the different emphysema subtypes and the presence of low BMD adjusting for other important factors. Results Seventy-five percent of the patients had low BMD (78 had osteopenia and 37 had osteoporosis). Emphysema was more frequent (66.1 vs 26.3%, p=<0.001) and severe in those with low BMD. Multivariable analysis adjusting for other significant cofactors (age, sex, FEV1, and severity of emphysema) showed that BMI (OR=0.91, 95% CI: 0.76–0.92) and centrilobular emphysema (OR=26.19, 95% CI: 1.71 to 399.44) were associated with low BMD. Conclusion Low BMD is highly prevalent in current and former smokers. BMI and centrilobular emphysema are strong and independent predictors of its presence, which suggests that they should be considered when evaluating smokers at risk for low BMD.
Collapse
Affiliation(s)
- Jessica Gonzalez
- Pulmonary Department, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | | | | | - Patricia Restituto
- Biochemistry Department, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Inmaculada Colina
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | | | - Ana B Alcaide
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Aránzazu Campo
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Juan Bertó
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Luis Seijo
- Pulmonary Department, Clínica Universidad de Navarra, Madrid, Spain
| | | | - Javier J Zulueta
- Pulmonary Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Nerea Varo
- Biochemistry Department, Clínica Universitaria de Navarra, Pamplona, Spain
| | | |
Collapse
|
24
|
Zhou Y, Liu QX, Li XM, Ma DD, Xing S, Feng JH, Zhang MH. Effects of ammonia exposure on growth performance and cytokines in the serum, trachea, and ileum of broilers. Poult Sci 2020; 99:2485-2493. [PMID: 32359584 PMCID: PMC7597540 DOI: 10.1016/j.psj.2019.12.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/23/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022] Open
Abstract
This study investigated the effects of ammonia (NH3) exposure (0, 15, 25, and 35 ppm) on growth performance and cytokines in the serum, trachea, and ileum of broilers. A total of 288 22-day-old male broiler chickens were assigned to 4 treatment groups with 6 replicates of 12 chickens for a 21-D trial period. Growth performance and cytokines (IL-1β, IL-6, and IL-10) concentrations in the serum, trachea, and ileum were measured in response to 3, 7, 14, or 21 D of exposure to NH3. Correlations between cytokines in the serum, trachea, and ileum and growth performance, and between tracheal and ileal cytokines, were also analyzed. Results showed that exposure to 15 ppm NH3 did not influence the growth performance, but exposure to both 25 ppm and 35 ppm NH3 decreased the growth performance compared to that of the control group. Exposure to 15 ppm NH3 for 3 D increased IL-6 concentrations and induced an inflammatory response in the trachea and ileum, whereas exposure to 15 ppm NH3 for 7 D increased IL-10 concentrations and induced an anti-inflammatory response in the ileum. Exposure to 25 ppm NH3 induced an inflammatory response in the serum, trachea, and ileum after 3 D and induced an anti-inflammatory response in the ileum after 7 D. Exposure to 35 ppm NH3 for 3 D induced both inflammatory and anti-inflammatory responses in the trachea and ileum. Furthermore, increases in cytokines in the serum, trachea, or ileum were accompanied by a decrease in BW, ADFI, ADG, and an increase of feed/gain (F/G) from 7 D to 21 D. In addition, tracheal cytokine, especially IL-1β, was positively correlated with ileal cytokine IL-1β. These results indicated that the low growth performance associated with NH3 exposure may be due in part to an increase in cytokines, and the inflammatory response in the trachea and ileum may be related to cross-talk by cytokines such as IL-6, IL-10, and, in particular, IL-1β.
Collapse
Affiliation(s)
- Ying Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qing Xiu Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiu Mei Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Dan Dan Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuang Xing
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jing Hai Feng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Min Hong Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
25
|
Suzuki S, Ishii M, Asakura T, Namkoong H, Okamori S, Yagi K, Kamata H, Kusumoto T, Kagawa S, Hegab AE, Yoda M, Horiuchi K, Hasegawa N, Betsuyaku T. ADAM17 protects against elastase-induced emphysema by suppressing CD62L + leukocyte infiltration in mice. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1172-L1182. [PMID: 32130031 DOI: 10.1152/ajplung.00214.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Pulmonary emphysema is a major manifestation of chronic obstructive pulmonary disease and is associated with chronic pulmonary inflammation caused by cigarette smoking, with contributions from immune cells such as neutrophils, macrophages, and lymphocytes. Although matrix metalloproteinases are well known to contribute to emphysema progression, the role of a disintegrin and metalloproteinase (ADAM) family proteins, other major metalloproteinases, in disease pathogenesis is largely unknown. ADAM17 is a major sheddase that cleaves various cell surface proteins, including CD62L, an adhesion molecule that plays a critical role in promoting the migration of immune cells to the site of inflammation. In the present study, we aimed to investigate the potential role of ADAM17 and CD62L in the development of elastase-induced emphysema. Control and Adam17flox/flox/Mx1-Cre (Adam17ΔMx1) mice (8-10 wk old) were intratracheally injected with 5 units of porcine pancreas elastase and monitored for 35 days after injection. Lung alveolar destruction was evaluated by analyzing the mean linear intercepts of lung tissue specimens and by histopathological examination. Mean linear intercepts data indicated that the degree of elastase-induced emphysema was significantly more severe in Adam17ΔMx1 mice. Furthermore, flow cytometry showed that CD62L+ neutrophil, CD62L+ macrophage, and CD62L+ B lymphocyte numbers were significantly increased in Adam17ΔMx1 mice. Moreover, the pharmacological depletion of CD62L+ cells with a CD62L-neutralizing antibody ameliorated the extent of emphysema in Adam17ΔMx1 mice. Collectively, these results suggest that ADAM17 possibly suppresses the progression of emphysema by proteolytically processing CD62L in immune cells and that ADAM17 and CD62L could be novel therapeutic targets for treating pulmonary emphysema.
Collapse
Affiliation(s)
- Shoji Suzuki
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Society of Promotion of Science, Tokyo, Japan
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Society of Promotion of Science, Tokyo, Japan
| | - Ho Namkoong
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Okamori
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Japan Society of Promotion of Science, Tokyo, Japan
| | - Kazuma Yagi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kamata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuya Kusumoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shizuko Kagawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ahmed E Hegab
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Yoda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Laboratory of Cell and Tissue Biology, Keio University School of Medicine, Tokyo, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Naoki Hasegawa
- Center for Infectious Diseases and Infection Control, Keio University School of Medicine, Tokyo, Japan
| | - Tomoko Betsuyaku
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
ADAM17 Activity and IL-6 Trans-Signaling in Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11111736. [PMID: 31694340 PMCID: PMC6895846 DOI: 10.3390/cancers11111736] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 02/08/2023] Open
Abstract
All ligands of the epidermal growth factor receptor (EGF-R) are transmembrane proteins, which need to be proteolytically cleaved in order to be systemically active. The major protease responsible for this cleavage is the membrane metalloprotease ADAM17, which also has been implicated in cleavage of TNFα and interleukin-6 (IL-6) receptor. It has been recently shown that in the absence of ADAM17, the main protease for EGF-R ligand processing, colon cancer formation is largely abrogated. Intriguingly, colon cancer formation depends on EGF-R activity on myeloid cells rather than on intestinal epithelial cells. A major activity of EGF-R on myeloid cells is the stimulation of IL-6 synthesis. Subsequently, IL-6 together with the ADAM17 shed soluble IL-6 receptor acts on intestinal epithelial cells via IL-6 trans-signaling to induce colon cancer formation, which can be blocked by the inhibitor of IL-6 trans-signaling, sgp130Fc. Blockade of IL-6 trans-signaling therefore offers a new therapeutic window downstream of the EGF-R for the treatment of colon cancer and possibly of other EGF-R related neoplastic diseases.
Collapse
|
27
|
Hayashi S, Matsuno Y, Tsunoda Y, Sakurai H, Kiwamoto T, Morishima Y, Ishii Y, Yoh K, Takahashi S, Hizawa N. Transcription Factor T-bet Attenuates the Development of Elastase-induced Emphysema in Mice. Am J Respir Cell Mol Biol 2019; 61:525-536. [DOI: 10.1165/rcmb.2018-0109oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - Keigyou Yoh
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
28
|
Saad M, Ruwanpura SM. Tissue Processing for Stereological Analyses of Lung Structure in Chronic Obstructive Pulmonary Disease. Methods Mol Biol 2018; 1725:155-162. [PMID: 29322416 DOI: 10.1007/978-1-4939-7568-6_13] [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] [Indexed: 03/23/2023]
Abstract
Quantitative data on lung structure, such as volume, surface area and length, are used for assessment of the functional performance of the lung during normal development and inflammatory-related diseases such as chronic obstructive pulmonary disorder (COPD), and carcinogenesis, in animal models. Stereology is considered as the gold standard to obtain quantitative data on lung structure, with a key advantage being to quantify irregular three-dimensional structures on the basis of measurement made on two-dimensional sections. Therefore, preservation of original tissue dimensions without shrinkage is vital for stereology.Three steps, fixation, sampling and embedding, are essential requirements to minimise tissue shrinkage to obtain theoretically unbiased estimates of stereological parameters of lung structures. Perfusion fixation by intratracheal instillation with 1.5% glutaraldehyde/1.5% formaldehyde at a pressure of 25 cm fluid column is considered as one of the best methods. A systematic uniform random sampling scheme is then applied to the fixed lung to ensure each and every part of the lung is analysed, irrespective of homogeneity or heterogeneity of the structural distribution. The sampled tissue sections are then embedded in glycol methacrylate to minimise further tissue shrinkage. Here we describe the accurate fixation, sampling and embedding for stereological methods to quantify lung structures in mice.
Collapse
Affiliation(s)
- Mohamed Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Saleela M Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
29
|
Vlahos R, Bozinovski S. Protocols to Evaluate Cigarette Smoke-Induced Lung Inflammation and Pathology in Mice. Methods Mol Biol 2018; 1725:53-63. [PMID: 29322408 DOI: 10.1007/978-1-4939-7568-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cigarette smoking is a major cause of chronic obstructive pulmonary disease (COPD). Inhalation of cigarette smoke causes inflammation of the airways, airway wall remodelling, mucus hypersecretion and progressive airflow limitation. Much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and infectious (viral and bacterial) exacerbations (AECOPD). Comorbidities, in particular skeletal muscle wasting, cardiovascular disease and lung cancer markedly impact on disease morbidity, progression and mortality. The mechanisms and mediators underlying COPD and its comorbidities are poorly understood and current COPD therapy is relatively ineffective. Many researchers have used animal modelling systems to explore the mechanisms underlying COPD, AECOPD and comorbidities of COPD with the goal of identifying novel therapeutic targets. Here we describe a mouse model that we have developed to define the cellular, molecular and pathological consequences of cigarette smoke exposure and the development of comorbidities of COPD.
Collapse
Affiliation(s)
- Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia.
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
30
|
Bradford E, Jacobson S, Varasteh J, Comellas AP, Woodruff P, O’Neal W, DeMeo DL, Li X, Kim V, Cho M, Castaldi PJ, Hersh C, Silverman EK, Crapo JD, Kechris K, Bowler RP. The value of blood cytokines and chemokines in assessing COPD. Respir Res 2017; 18:180. [PMID: 29065892 PMCID: PMC5655820 DOI: 10.1186/s12931-017-0662-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/09/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Blood biomarkers are increasingly used to stratify high risk chronic obstructive pulmonary disease (COPD) patients; however, there are fewer studies that have investigated multiple biomarkers and replicated in multiple large well-characterized cohorts of susceptible current and former smokers. METHODS We used two MSD multiplex panels to measure 9 cytokines and chemokines in 2123 subjects from COPDGene and 1117 subjects from SPIROMICS. These biomarkers included: interleukin (IL)-2, IL-6, IL-8, IL-10, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, eotaxin/CCL-11, eotaxin-3/CCL-26, and thymus and activation-regulated chemokine (TARC)/CCL-17. Regression models adjusted for clinical covariates were used to determine which biomarkers were associated with the following COPD phenotypes: airflow obstruction (forced expiratory flow at 1 s (FEV1%) and FEV1/forced vital capacity (FEV1/FVC), chronic bronchitis, COPD exacerbations, and emphysema. Biomarker-genotype associations were assessed by genome-wide association of single nucleotide polymorphisms (SNPs). RESULTS Eotaxin and IL-6 were strongly associated with airflow obstruction and accounted for 3-5% of the measurement variance on top of clinical variables. IL-6 was associated with progressive airflow obstruction over 5 years and both IL-6 and IL-8 were associated with progressive emphysema over 5 years. None of the biomarkers were consistently associated with chronic bronchitis or COPD exacerbations. We identified one novel SNP (rs9302690 SNP) that was associated with CCL17 plasma measurements. CONCLUSION When assessing smoking related pulmonary disease, biomarkers of inflammation such as IL-2, IL-6, IL-8, and eotaxin may add additional modest predictive value on top of clinical variables alone. TRIAL REGISTRATION COPDGene (ClinicalTrials.gov Identifier: NCT02445183 ). Subpopulations and Intermediate Outcomes Measures in COPD Study (SPIROMICS) ( ClinicalTrials.gov Identifier: NCT 01969344 ).
Collapse
Affiliation(s)
- Eric Bradford
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Sean Jacobson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Jason Varasteh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Alejandro P. Comellas
- University of Iowa, Internal Medicine, 200 Hawkins Dr C331-GH, Iowa City, IA 52242 USA
| | - Prescott Woodruff
- UCSF, Division of Pulmonary and Critical Care Medicine and Cardiovascular Research Institute, Box 0130, Rm HSE 1305, 513 Parnassus Ave, San Francisco, CA 94143 USA
| | - Wanda O’Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Dawn L. DeMeo
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
| | - Xingnan Li
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ USA
| | - Victor Kim
- Temple University School of Medicine, Pulmonary and Critical Care Medicine, 785 Parkinson Pavilion, 3401 North Broad Street, Philadelphia, PA 19140 USA
| | - Michael Cho
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Peter J. Castaldi
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
- Tufts Medical Center, ICRHPS, 800 Washington St, Box 63, Boston, MA 02111 USA
| | - Craig Hersh
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - James D. Crapo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, University of Colorado Denver, Colorado School of Public Health, Mail Stop B119, 13001 E. 17th Place, Aurora, CO 80045 USA
| | - Russell P. Bowler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, University of Colorado Anschutz Medical Campus, Research Building 2, 9th Floor, 12700 E. 19th Ave, Aurora, CO USA
| |
Collapse
|
31
|
Rodrigues Brandao-Rangel MA, Bachi ALL, Oliveira-Junior MC, Abbasi A, Silva-Renno A, Aparecida de Brito A, Ligeiro de Oliveira AP, Choqueta Toledo-Arruda A, Belvisi MG, Paula Vieira R. Exercise Inhibits the Effects of Smoke-Induced COPD Involving Modulation of STAT3. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6572714. [PMID: 29326759 PMCID: PMC5664289 DOI: 10.1155/2017/6572714] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/09/2017] [Accepted: 08/29/2017] [Indexed: 01/31/2023]
Abstract
PURPOSE Evaluate the participation of STAT3 in the effects of aerobic exercise (AE) in a model of smoke-induced COPD. METHODS C57Bl/6 male mice were divided into control, Exe, COPD, and COPD+Exe groups. Smoke were administered during 90 days. Treadmill aerobic training begun on day 61 until day 90. Pulmonary inflammation, systemic inflammation, the level of lung emphysema, and the airway remodeling were evaluated. Analysis of integral and phosphorylated expression of STAT3 by airway epithelial cells, peribronchial leukocytes, and parenchymal leukocytes was performed. RESULTS AE inhibited smoke-induced accumulation of total cells (p < 0.001), lymphocytes (p < 0.001), and neutrophils (p < 0.001) in BAL, as well as BAL levels of IL-1β (p < 0.001), CXCL1 (p < 0.001), IL-17 (p < 0.001), and TNF-α (p < 0.05), while increased the levels of IL-10 (p < 0.001). AE also inhibited smoke-induced increases in total leukocytes (p < 0.001), neutrophils (p < 0.05), lymphocytes (p < 0.001), and monocytes (p < 0.01) in blood, as well as serum levels of IL-1β (p < 0.01), CXCL1 (p < 0.01), IL-17 (p < 0.05), and TNF-α (p < 0.01), while increased the levels of IL-10 (p < 0.001). AE reduced smoke-induced emphysema (p < 0.001) and collagen fiber accumulation in the airways (p < 0.001). AE reduced smoke-induced STAT3 and phospho-STAT3 expression in airway epithelial cells (p < 0.001), peribronchial leukocytes (p < 0.001), and parenchymal leukocytes (p < 0.001). CONCLUSIONS AE reduces smoke-induced COPD phenotype involving STAT3.
Collapse
Affiliation(s)
- Maysa Alves Rodrigues Brandao-Rangel
- Nove de Julho University, São Paulo, SP, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of São José dos Campos Humanitas and Universidade Brasil, São Paulo, SP, Brazil
| | - Andre Luis Lacerda Bachi
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil
| | - Manoel Carneiro Oliveira-Junior
- Nove de Julho University, São Paulo, SP, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of São José dos Campos Humanitas and Universidade Brasil, São Paulo, SP, Brazil
| | - Asghar Abbasi
- Institute for Memory Impairments and Neurological Disorders (MIND Institute), University of California, Irvine, CA, USA
| | - Adriano Silva-Renno
- Nove de Julho University, São Paulo, SP, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of São José dos Campos Humanitas and Universidade Brasil, São Paulo, SP, Brazil
| | | | | | - Alessandra Choqueta Toledo-Arruda
- Laboratory of Experimental Therapeutics (LIM 20), Department of Medicine, School of Medicine, University of Sao Paulo, São Paulo, SP, Brazil
| | - Maria Gabriela Belvisi
- Respiratory Pharmacology Group, Airway Disease, National Heart and Lung Institute, Imperial College London, London, UK
| | - Rodolfo Paula Vieira
- Nove de Julho University, São Paulo, SP, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), School of Medical Sciences of São José dos Campos Humanitas and Universidade Brasil, São Paulo, SP, Brazil
| |
Collapse
|
32
|
Deoxyribonuclease 1 reduces pathogenic effects of cigarette smoke exposure in the lung. Sci Rep 2017; 7:12128. [PMID: 28935869 PMCID: PMC5608940 DOI: 10.1038/s41598-017-12474-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
Our aim was to investigate if deoxyribonuclease (DNase) 1 is a potential therapeutic agent to reduce pathogenic effects of cigarette smoke exposure in the lung. Cigarette smoke causes protease imbalance with excess production of proteases, which is a key process in the pathogenesis of emphysema. The mechanisms responsible for this effect are not well-defined. Our studies demonstrate both in vitro and in vivo that cigarette smoke significantly increases the expression of neutrophil and macrophage extracellular traps with coexpression of the pathogenic proteases, neutrophil elastase and matrix metalloproteinases 9 and 12. This response to cigarette smoke was significantly reduced by the addition of DNase 1, which also significantly decreased macrophage numbers and lung proteolysis. DNase 1, a treatment currently in clinical use, can diminish the pathogenic effects of cigarette smoke.
Collapse
|
33
|
Ruwanpura SM, McLeod L, Dousha LF, Seow HJ, Alhayyani S, Tate MD, Deswaerte V, Brooks GD, Bozinovski S, MacDonald M, Garbers C, King PT, Bardin PG, Vlahos R, Rose-John S, Anderson GP, Jenkins BJ. Therapeutic Targeting of the IL-6 Trans-Signaling/Mechanistic Target of Rapamycin Complex 1 Axis in Pulmonary Emphysema. Am J Respir Crit Care Med 2017; 194:1494-1505. [PMID: 27373892 DOI: 10.1164/rccm.201512-2368oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The potent immunomodulatory cytokine IL-6 is consistently up-regulated in human lungs with emphysema and in mouse emphysema models; however, the mechanisms by which IL-6 promotes emphysema remain obscure. IL-6 signals using two distinct modes: classical signaling via its membrane-bound IL-6 receptor (IL-6R), and trans-signaling via a naturally occurring soluble IL-6R. OBJECTIVES To identify whether IL-6 trans-signaling and/or classical signaling contribute to the pathogenesis of emphysema. METHODS We used the gp130F/F genetic mouse model for spontaneous emphysema and cigarette smoke-induced emphysema models. Emphysema in mice was quantified by various methods including in vivo lung function and stereology, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay was used to assess alveolar cell apoptosis. In mouse and human lung tissues, the expression level and location of IL-6 signaling-related genes and proteins were measured, and the levels of IL-6 and related proteins in sera from emphysematous mice and patients were also assessed. MEASUREMENTS AND MAIN RESULTS Lung tissues from patients with emphysema, and from spontaneous and cigarette smoke-induced emphysema mouse models, were characterized by excessive production of soluble IL-6R. Genetic blockade of IL-6 trans-signaling in emphysema mouse models and therapy with the IL-6 trans-signaling antagonist sgp130Fc ameliorated emphysema by suppressing augmented alveolar type II cell apoptosis. Furthermore, IL-6 trans-signaling-driven emphysematous changes in the lung correlated with mechanistic target of rapamycin complex 1 hyperactivation, and treatment of emphysema mouse models with the mechanistic target of rapamycin complex 1 inhibitor rapamycin attenuated emphysematous changes. CONCLUSIONS Collectively, our data reveal that specific targeting of IL-6 trans-signaling may represent a novel treatment strategy for emphysema.
Collapse
Affiliation(s)
- Saleela M Ruwanpura
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Louise McLeod
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Lovisa F Dousha
- 3 Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Huei J Seow
- 3 Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Sultan Alhayyani
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Michelle D Tate
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Virginie Deswaerte
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Gavin D Brooks
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Steven Bozinovski
- 3 Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia.,4 School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Martin MacDonald
- 5 Monash Lung and Sleep, Monash Medical Centre, Victoria, Australia; and
| | - Christoph Garbers
- 6 Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Paul T King
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,5 Monash Lung and Sleep, Monash Medical Centre, Victoria, Australia; and
| | - Philip G Bardin
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,5 Monash Lung and Sleep, Monash Medical Centre, Victoria, Australia; and
| | - Ross Vlahos
- 3 Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia.,4 School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Stefan Rose-John
- 6 Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Gary P Anderson
- 3 Lung Health Research Centre, Department of Pharmacology and Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Brendan J Jenkins
- 1 Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,2 Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
34
|
Mishra R, Foster D, Vasu VT, Thaikoottathil JV, Kosmider B, Chu HW, Bowler RP, Finigan JH. Cigarette Smoke Induces Human Epidermal Receptor 2-Dependent Changes in Epithelial Permeability. Am J Respir Cell Mol Biol 2017; 54:853-64. [PMID: 26600084 DOI: 10.1165/rcmb.2014-0437oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The airway epithelium constitutes a protective barrier against inhaled insults, such as viruses, bacteria, and toxic fumes, including cigarette smoke (CS). Maintenance of bronchial epithelial integrity is central for airway health, and defective epithelial barrier function contributes to the pathogenesis of CS-mediated diseases, such as chronic obstructive pulmonary disease. Although CS has been shown to increase epithelial permeability, current understanding of the mechanisms involved in CS-induced epithelial barrier disruption remains incomplete. We have previously identified that the receptor tyrosine kinase human epidermal receptor (HER) 2 growth factor is activated by the ligand neuregulin-1 and increases epithelial permeability in models of inflammatory acute lung injury. We hypothesized that CS activates HER2 and that CS-mediated changes in barrier function would be HER2 dependent in airway epithelial cells. We determined that HER2 was activated in whole lung, as well as isolated epithelial cells, from smokers, and that acute CS exposure resulted in HER2 activation in cultured bronchial epithelial cells. Mechanistic studies determined that CS-mediated HER2 activation is independent of neuregulin-1 but required upstream activation of the epidermal growth factor receptor. HER2 was required for CS-induced epithelial permeability as knockdown of HER2 blocked increases in permeability after CS. CS caused an increase in IL-6 production by epithelial cells that was dependent on HER2-mediated extracellular signal-regulated kinases (Erk) activation. Finally, blockade of IL-6 attenuated CS-induced epithelial permeability. Our data indicate that CS activates pulmonary epithelial HER2 and that HER2 is a central mediator of CS-induced epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Rangnath Mishra
- 1 Division Oncology, National Jewish Health, Denver, Colorado
| | - Daniel Foster
- 1 Division Oncology, National Jewish Health, Denver, Colorado
| | - Vihas T Vasu
- 1 Division Oncology, National Jewish Health, Denver, Colorado
| | | | - Beata Kosmider
- 2 Division Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado; and.,3 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| | - Hong Wei Chu
- 2 Division Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado; and.,3 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| | - Russell P Bowler
- 2 Division Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado; and.,3 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| | - James H Finigan
- 1 Division Oncology, National Jewish Health, Denver, Colorado.,2 Division Pulmonary and Critical Care Medicine, National Jewish Health, Denver, Colorado; and.,3 Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, Colorado
| |
Collapse
|
35
|
Craig JM, Scott AL, Mitzner W. Immune-mediated inflammation in the pathogenesis of emphysema: insights from mouse models. Cell Tissue Res 2017; 367:591-605. [PMID: 28164246 PMCID: PMC5366983 DOI: 10.1007/s00441-016-2567-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/21/2016] [Indexed: 12/31/2022]
Abstract
The cellular mechanisms that result in the initiation and progression of emphysema are clearly complex. A growing body of human data combined with discoveries from mouse models utilizing cigarette smoke exposure or protease administration have improved our understanding of emphysema development by implicating specific cell types that may be important for the pathophysiology of chronic obstructive pulmonary disease. The most important aspects of emphysematous damage appear to be oxidative or protease stress and sustained macrophage activation and infiltration of other immune cells leading to epithelial damage and cell death. Despite the identification of these associated processes and cell types in many experimental studies, the reasons why cigarette smoke and other pollutants result in unremitting damage instead of injury resolution are still uncertain. We propose an important role for macrophages in the sequence of events that lead and maintain this chronic tissue pathologic process in emphysema. This model involves chronic activation of macrophage subtypes that precludes proper healing of the lung. Further elucidation of the cross-talk between epithelial cells that release damage-associated signals and the cellular immune effectors that respond to these cues is a critical step in the development of novel therapeutics that can restore proper lung structure and function to those afflicted with emphysema.
Collapse
Affiliation(s)
- John M Craig
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe St., Baltimore, MD, USA
| | - Alan L Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe St., Baltimore, MD, USA.
| |
Collapse
|
36
|
Balic JJ, Garbers C, Rose-John S, Yu L, Jenkins BJ. Interleukin-11-driven gastric tumourigenesis is independent of trans-signalling. Cytokine 2017; 92:118-123. [PMID: 28160627 DOI: 10.1016/j.cyto.2017.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 11/28/2022]
Abstract
Deregulated gp130-dependent STAT3 signalling by the pleiotropic cytokine interleukin (IL)-11 has been implicated in the pathogenesis of gastric cancer (GC), the third most common cancer worldwide. While the IL-11-gp130-STAT3 signalling axis has traditionally been thought to exclusively use the membrane-bound IL-11 receptor (mIL-11R), recent evidence suggests that mIL-11R can be proteolytically cleaved to generate a soluble form (sIL-11R) which can elicit trans-signalling. Since the role of IL-11 trans-signalling in disease pathogenesis is unknown, here we have employed the IL-11-driven gp130F/F spontaneous model of GC to determine whether IL-11 trans-signalling promotes gastric tumourigenesis. sIL-11R protein was detectable in gastric tissue from GC patients, and sIL-11R levels were elevated in tumours of gp130F/F mice compared to matched non-tumours. Among candidate proteases associated with the generation of sIL-11R, ADAM10 and the related metalloprotease ADAM17 were significantly upregulated in tumours of both gp130F/F mice and GC patients compared to matched non-tumour tissues. The genetic blockade of IL-11 trans-signalling in gp130F/F mice upon the transgenic over-expression of the trans-signalling antagonist, sgp130Fc, failed to suppress gastric inflammation and associated tumour growth, and also had no effect on reducing hyper-activated STAT3 levels. Furthermore, a non-essential role for ADAM17 in IL-11-driven gastric tumourigenesis was supported by the observation that the tumour burden was unaffected in gp130F/F:Adam17ex/ex mice in which ADAM17 expression levels have been substantially reduced. Collectively, these findings suggest that classic signalling rather than trans-signalling is the mode by which IL-11 promotes gastric tumourigenesis.
Collapse
Affiliation(s)
- Jesse J Balic
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Christoph Garbers
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Olshausenstrasse 40, D-24098 Kiel, Germany
| | - Liang Yu
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Molecular Translational Science, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
37
|
Blockade of the IL-6 trans-signalling/STAT3 axis suppresses cachexia in Kras-induced lung adenocarcinoma. Oncogene 2016; 36:3059-3066. [DOI: 10.1038/onc.2016.437] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/09/2016] [Accepted: 10/11/2016] [Indexed: 12/23/2022]
|
38
|
Brooks GD, McLeod L, Alhayyani S, Miller A, Russell PA, Ferlin W, Rose-John S, Ruwanpura S, Jenkins BJ. IL6 Trans-signaling Promotes KRAS-Driven Lung Carcinogenesis. Cancer Res 2016; 76:866-76. [PMID: 26744530 DOI: 10.1158/0008-5472.can-15-2388] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/06/2015] [Indexed: 11/16/2022]
Abstract
Oncogenic KRAS mutations occur frequently in lung adenocarcinoma. The signaling pathways activated by IL6 promote Kras-driven lung tumorigenesis, but the basis for this cooperation is uncertain. In this study, we used the gp130(F/F) (Il6st) knock-in mouse model to examine the pathogenic contribution of hyperactivation of the STAT3 arm of IL6 signaling on KRAS-driven lung tumorigenesis. Malignant growths in the gp130(F/F):Kras(G12D) model displayed features of atypical adenomatous hyperplasia, adenocarcinoma in situ, and invasive adenocarcinoma throughout the lung, as compared with parental Kras(G12D) mice, where STAT3 was not hyperactivated. Among IL6 family cytokines, only IL6 was upregulated in the lung. Accordingly, normalization of pulmonary STAT3 activity, by genetic ablation of either Il6 or Stat3, suppressed the extent of lung cancer in the model. Mechanistic investigations revealed elevation in the lung of soluble IL6 receptor (sIL6R), the key driver of IL6 trans-signaling, and blocking this mechanism via interventions with an anti-IL6R antibody or the inhibitor sgp130Fc ameliorated lung cancer pathogenesis. Clinically, expression of IL6 and sIL6R was increased significantly in human specimens of lung adenocarcinoma or patient serum. Our results offer a preclinical rationale to clinically evaluate IL6 trans-signaling as a therapeutic target for the treatment of KRAS-driven lung adenocarcinoma.
Collapse
Affiliation(s)
- Gavin D Brooks
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Sultan Alhayyani
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Alistair Miller
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Prudence A Russell
- St Vincent's Hospital, Fitzroy, Victoria, Australia. Department of Pathology, Melbourne Medical School, Melbourne University, Parkville, Victoria, Australia
| | | | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Saleela Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
39
|
Tabata C, Tabata R, Takahashi Y, Nakamura K, Nakano T. Thalidomide prevents cigarette smoke extract-induced lung damage in mice. Int Immunopharmacol 2015; 25:511-7. [PMID: 25749497 DOI: 10.1016/j.intimp.2015.02.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a progressive airway obstruction that is not completely reversible and is mainly caused by smoking tobacco. COPD is a major cause of morbidity and mortality worldwide and there are currently no proven effective treatments. The pathogenesis of COPD involves several factors such as chronic inflammation, oxidative stress, and apoptosis. Cytokines play important roles in chronic inflammation. Thalidomide (Thal) has been used to treat multiple myeloma due to its inhibitory effects on IL-6-induced cell growth. We recently demonstrated that thalidomide (Thal) played important roles in cytokine-induced lung damage in a bleomycin-induced pulmonary fibrosis model in mice. We herein examined the preventative effects of Thal on cigarette smoke extract (CSE)-induced emphysematous changes in mice. We performed histological examinations and quantitative measurements of the expression of IL-1β and IL-6 mRNA, as well as apoptosis in CSE-induced mouse lung tissues treated with or without Thal. The results of the histological examination showed that Thal ameliorated CSE-induced emphysema in mice. It also inhibited the expression of IL-1β and IL-6 mRNA in mouse lung tissues. Thal decreased apoptosis in the mouse lung. In vitro studies revealed that Thal decreased 1) the expression of IL-1β and IL-6 in human lung epithelial cells, and 2) CSE-induced apoptosis and the inhibition of cell growth, which may be the underlying mechanisms for the preventative effects of Thal on emphysema. These results provide a rationale for exploring the clinical use of Thal for COPD.
Collapse
Affiliation(s)
| | - Rie Tabata
- Department of Internal Medicine, Hyogo Prefectural Tsukaguchi Hospital, Japan
| | - Yuta Takahashi
- Department of Pharmacology 1, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo, Japan
| | - Kazuki Nakamura
- Department of Pharmacology 1, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo, Japan
| | | |
Collapse
|
40
|
He ZH, Chen P, Chen Y, He SD, Ye JR, Zhang HL, Cao J. Comparison between cigarette smoke-induced emphysema and cigarette smoke extract-induced emphysema. Tob Induc Dis 2015; 13:6. [PMID: 25814921 PMCID: PMC4374374 DOI: 10.1186/s12971-015-0033-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 03/15/2015] [Indexed: 12/18/2022] Open
Abstract
Background and objective Emphysema is the main pathological feature of COPD and also is the focus of the related research. Although several emphysema animal models have been established, exact comparison of findings is seldom. The present study aimed to compare cigarette smoke (CS) exposure-induced emphysema model and intraperitoneal injection of cigarette smoke extract (CSE)-induced emphysema model to evaluate the effectiveness of the two different modeling methods. Methods Six-week-old male C57BL/6 J mice were used and randomly divided into two groups: CS exposure and intraperitoneal injection of CSE. Each group was subdivided into two subgroups: control and CS or CSE. Lung function, mean linear intercept (MLI), destructive index (DI), apoptotic index (AI), total and differential cells count in broncholavolar lavage fluid (BALF), SOD and IL-6 concentration in serum were measured. Results Compared with their respective controls, lung function was significantly decreased in CS and CSE groups (P < 0.01); MLI, DI, and AI of lung tissue were significantly higher in CS and CSE groups (P < 0.01); total number of leukocytes, the number and percentage of neutrophils (NEUs), and the number of macrophages (MAC) in BALF were significantly higher in CS and CSE groups (P < 0.01); SOD concentration in serum was significantly decreased in CS and CSE groups (P < 0.01); IL-6 concentration in serum was significantly increased in in CS and CSE groups (P < 0.01). There was no significant difference between CS group and CSE group in any of the parameters described above. Conclusions Both CS exposure and intraperitoneal injection of CSE could induce emphysema and the effectiveness of the two different modeling methods were equal.
Collapse
Affiliation(s)
- Zhi-Hui He
- Intensive Care Unit, the Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China ; Division of Respiratory Disease, Department of Internal Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Sheng-Dong He
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Ji-Ru Ye
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Hong-Liang Zhang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011 China
| | - Jun Cao
- Department of Respiratory Medicine, Hunan Provincial People's Hospital, Changsha, Hunan 410005 China
| |
Collapse
|
41
|
Vlahos R, Bozinovski S. Preclinical murine models of Chronic Obstructive Pulmonary Disease. Eur J Pharmacol 2015; 759:265-71. [PMID: 25818750 DOI: 10.1016/j.ejphar.2015.03.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/03/2015] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a major incurable global health burden and is the 4th leading cause of death worldwide. It is believed that an exaggerated inflammatory response to cigarette smoke causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T lymphocytes are prominent, leads to oxidative stress, emphysema, small airway fibrosis and mucus hypersecretion. Much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and infectious (viral and bacterial) exacerbations (AECOPD). Comorbidities, defined as other chronic medical conditions, in particular skeletal muscle wasting and cardiovascular disease markedly impact on disease morbidity, progression and mortality. The mechanisms and mediators underlying COPD and its comorbidities are poorly understood and current COPD therapy is relatively ineffective. Thus, there is an obvious need for new therapies that can prevent the induction and progression of COPD and effectively treat AECOPD and comorbidities of COPD. Given that access to COPD patients can be difficult and that clinical samples often represent a "snapshot" at a particular time in the disease process, many researchers have used animal modelling systems to explore the mechanisms underlying COPD, AECOPD and comorbidities of COPD with the goal of identifying novel therapeutic targets. This review highlights the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and the recent advances in modelling infectious exacerbations and comorbidities of COPD.
Collapse
Affiliation(s)
- Ross Vlahos
- School of Health Sciences, Health Innovations Research Institute, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia; Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Steven Bozinovski
- School of Health Sciences, Health Innovations Research Institute, RMIT University, PO Box 71, Bundoora, VIC 3083, Australia; Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
42
|
Ruwanpura S, McLeod L, Anderson G, Valhos R, Rose-John S, Jenkins BJ. 161. Cytokine 2014. [DOI: 10.1016/j.cyto.2014.07.168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Fallica J, Boyer L, Kim B, Serebreni L, Varela L, Hamdan O, Wang L, Simms T, Damarla M, Kolb TM, Bucala R, Mitzner W, Hassoun PM, Damico R. Macrophage migration inhibitory factor is a novel determinant of cigarette smoke-induced lung damage. Am J Respir Cell Mol Biol 2014; 51:94-103. [PMID: 24490973 DOI: 10.1165/rcmb.2013-0371oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cigarette smoke (CS) is the most common cause of chronic obstructive pulmonary diseases (COPD), including emphysema. CS exposure impacts all cell types within the airways and lung parenchyma, causing alveolar tissue destruction through four mechanisms: (1) oxidative stress; (2) inflammation; (3) protease-induced degradation of the extracellular matrix; and (4) enhanced alveolar epithelial and endothelial cell (EC) apoptosis. Studies in human pulmonary ECs demonstrate that macrophage migration inhibitory factor (MIF) antagonizes CS-induced apoptosis. Here, we used human microvascular ECs, an animal model of emphysema (mice challenged with chronic CS), and patient serum samples to address both the capacity of CS to alter MIF expression and the effects of MIF on disease severity. We demonstrate significantly reduced serum MIF levels in patients with COPD. In the murine model, chronic CS exposure resulted in decreased MIF mRNA and protein expression in the intact lung. MIF deficiency (Mif(-/-)) potentiated the toxicity of CS exposure in vivo via increased apoptosis of ECs, resulting in enhanced CS-induced tissue remodeling. This was linked to MIF's capacity to protect against double-stranded DNA damage and suppress p53 expression. Taken together, MIF appears to antagonize CS-induced toxicity in the lung and resultant emphysematous tissue remodeling by suppressing EC DNA damage and controlling p53-mediated apoptosis, highlighting a critical role of MIF in EC homeostasis within the lung.
Collapse
Affiliation(s)
- Jonathan Fallica
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Aida Y, Shibata Y, Abe S, Inoue S, Kimura T, Igarashi A, Yamauchi K, Nunomiya K, Kishi H, Nemoto T, Sato M, Sato-Nishiwaki M, Nakano H, Sato K, Kubota I. Inhibition of elastase-pulmonary emphysema in dominant-negative MafB transgenic mice. Int J Biol Sci 2014; 10:882-94. [PMID: 25170302 PMCID: PMC4147222 DOI: 10.7150/ijbs.8737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 07/30/2014] [Indexed: 11/06/2022] Open
Abstract
Alveolar macrophages (AMs) play important roles in the pathogenesis of chronic obstructive pulmonary disease (COPD). We previously demonstrated upregulation of the transcription factor MafB in AMs of mice exposed to cigarette smoke. The aim of this study was to elucidate the roles of MafB in the development of pulmonary emphysema. Porcine pancreatic elastase was administered to wild-type (WT) and dominant-negative (DN)-MafB transgenic (Tg) mice in which MafB activity was suppressed only in macrophages. We measured the mean linear intercept and conducted cell differential analysis of bronchoalveolar lavage (BAL) cells, surface marker analysis using flow cytometry, and immunohistochemical staining using antibodies to matrix metalloproteinase (MMP)-9 and MMP-12. Airspace enlargement of the lungs was suppressed significantly in elastase-treated DN-MafB Tg mice compared with treated WT mice. AMs with projected pseudopods were decreased in DN-MafB Tg mice. The number of cells intermediately positive for F4/80 and weakly or intermediately positive for CD11b, which are considered cell subsets of matured AMs, decreased in the BAL of DN-MafB Tg mice. Furthermore, MMP-9 and -12 were significantly downregulated in BAL cells of DN-MafB Tg mice. Because MMPs exacerbate emphysema, MafB may be involved in pulmonary emphysema development through altered maturation of macrophages and MMP expression.
Collapse
Affiliation(s)
- Yasuko Aida
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yoko Shibata
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Shuichi Abe
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Sumito Inoue
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Tomomi Kimura
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Akira Igarashi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Keiko Yamauchi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Keiko Nunomiya
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Hiroyuki Kishi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takako Nemoto
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Masamichi Sato
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Michiko Sato-Nishiwaki
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Hiroshi Nakano
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Kento Sato
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Isao Kubota
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| |
Collapse
|
45
|
Magnitude of influenza virus replication and cell damage is associated with interleukin-6 production in primary cultures of human tracheal epithelium. Respir Physiol Neurobiol 2014; 202:16-23. [PMID: 25064661 DOI: 10.1016/j.resp.2014.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/15/2014] [Accepted: 07/15/2014] [Indexed: 01/22/2023]
Abstract
Primary cultures of human tracheal epithelium were infected with influenza viruses to examine the relationships between the magnitude of viral replication and infection-induced cell damage and cytokine production in airway epithelial cells. Infection with four strains of the type A influenza virus increased the detached cell number and lactate dehydrogenase (LDH) levels in the supernatants. The detached cell number and LDH levels were related to the viral titers and interleukin (IL)-6 levels and the nuclear factor kappa B (NF-κB) p65 activation. Treatment of the cells with an anti-IL-6 receptor antibody and an NF-κB inhibitor, caffeic acid phenethyl ester, reduced the detached cell number, viral titers and the LDH levels and improved cell viability after infection with the pandemic influenza virus [A/Sendai-H/N0633/2009 (H1N1) pdm09]. A caspase-3 inhibitor, benzyloxycarbonyl-DEVD-fluoromethyl ketone, reduced the detached cell number and viral titers. Influenza viral infection-induced cell damage may be partly related to the magnitude of viral replication, NF-κB-p65-mediated IL-6 production and caspase-3 activation.
Collapse
|
46
|
Yamaya M, Nadine L, Kubo H, Saito K, Saito R, Nishimura H. The effects of neuraminidase inhibitors on the release of oseltamivir-sensitive and oseltamivir-resistant influenza viruses from primary cultures of human tracheal epithelium. J Med Virol 2014; 87:25-34. [PMID: 24838828 DOI: 10.1002/jmv.23974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2014] [Indexed: 11/07/2022]
Abstract
Defining the effects of neuraminidase inhibitors on influenza virus infection may provide important information for the treatment of patients. The effects of neuraminidase inhibitors have been examined using various methods, including viral release from kidney cells. However, the effects of neuraminidase inhibitors on viral release from primary cultures of human tracheal epithelial cells, which retain functions of the original tissues, have not been studied. The effects of neuraminidase inhibitors on the replication of the pandemic influenza virus [A/Sendai-H/N0633/2009 (H1N1) pdm09] and the seasonal influenza virus [A/Sendai-H/216/2009 (H1N1)] that was isolated during the 2008-2009 season were examined. The virus stocks were generated by infecting tracheal cells with the pandemic or seasonal influenza virus. Four types of inhibitors (oseltamivir, zanamivir, laninamivir, and peramivir) reduced pandemic viral titers and concentrations of the cytokines interleukin-6 and tumor necrosis factor-α in supernatants and viral RNA in cells. However, oseltamivir did not reduce seasonal viral titers, cytokine concentrations and viral RNA, and the 50% inhibitory concentration (IC50 ) of oseltamivir for neuraminidase activity in the seasonal virus was 300-fold higher than that observed for the pandemic influenza virus. The seasonal influenza virus had an oseltamivir-resistant genotype. The magnitude of the IC50 values of the neuraminidase inhibitors for the seasonal influenza virus was inversely related to the magnitude of the inhibitory effects on viral release. These methods for measuring the release of virus and inflammatory cytokines from primary cultures of human tracheal epithelium may provide useful information regarding the effects of neuraminidase inhibitors on influenza viruses.
Collapse
Affiliation(s)
- Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Leung JM, Liu JC, Mtambo A, Ngan D, Nashta N, Guillemi S, Harris M, Lima VD, Mattman A, Shaipanich T, Raju R, Hague C, Leipsic JA, Sin DD, Montaner JS, Man SP. The determinants of poor respiratory health status in adults living with human immunodeficiency virus infection. AIDS Patient Care STDS 2014; 28:240-7. [PMID: 24742270 DOI: 10.1089/apc.2013.0373] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The increased longevity afforded by combination antiretroviral therapy in developed countries has led to an increased concern regarding senescence-related diseases in patients with human immunodeficiency virus (HIV) infection. Previous epidemiologic analyses have demonstrated an increased risk of chronic obstructive pulmonary disease, as well as a significant burden of respiratory symptoms in HIV-infected patients. We performed the St. George's Respiratory Questionnaire (SGRQ) in 199 HIV-positive men, and determined the predominant factors contributing to poor respiratory-related health status. In univariate analyses, worse SGRQ scores were associated with respiratory-related variables such as greater smoking pack-year history (p=0.028), lower forced expiratory volume in 1 second (FEV1) (p<0.001), and worse emphysema severity as quantified by computed tomographic imaging (p=0.017). In addition, HIV-specific variables, such as a history of plasma viral load >100,000 copies/mL (p=0.043), lower nadir CD4 cell count (p=0.040), and current CD4 cell count ≤350 cells/μL (p=0.005), as well as elevated levels of inflammatory markers, specifically plasma interleukin (IL)-6 (p=0.002) and alpha-1 antitrypsin (p=0.005) were also associated with worse SGRQ scores. In a multiple regression model, FEV1, current CD4 count ≤350 cells/μL, and IL-6 levels remained significant contributors to reduced respiratory-related health status. HIV disease activity as measured by HIV-related immunosuppression in conjunction with the triggering of key inflammatory pathways may be important determinants of worse respiratory health status among HIV-infected individuals. Limitations of this analysis include the absence of available echocardiograms, diffusion capacity and lung volume testing, and an all-male cohort due to the demographics of the clinic population.
Collapse
Affiliation(s)
| | - Joseph C. Liu
- UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Andy Mtambo
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
| | - David Ngan
- UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Negar Nashta
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
| | - Silvia Guillemi
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
- Department of Family Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of HIV/AIDS, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marianne Harris
- AIDS Research Program, St. Paul's Hospital, Vancouver, BC, Canada
- Department of Family Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of HIV/AIDS, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Viviane D. Lima
- BC Centre for Excellence in HIV/AIDS, St. Paul's Hospital, Vancouver, BC, Canada
| | - Andre Mattman
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tawimas Shaipanich
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| | - Rekha Raju
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Cameron Hague
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Jonathon A. Leipsic
- Department of Radiology and Diagnostic Imaging, St. Paul's Hospital, Vancouver, BC, Canada
| | - Don D. Sin
- UBC James Hogg Research Centre, Vancouver, BC, Canada
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| | - Julio S. Montaner
- BC Centre for Excellence in HIV/AIDS, St. Paul's Hospital, Vancouver, BC, Canada
| | - S.F. Paul Man
- UBC James Hogg Research Centre, Vancouver, BC, Canada
- UBC Department of Medicine and Division of Respiratory Medicine, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
48
|
Differential involvement of gp130 signalling pathways in modulating tobacco carcinogen-induced lung tumourigenesis. Oncogene 2014; 34:1510-9. [DOI: 10.1038/onc.2014.99] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 02/19/2014] [Accepted: 03/10/2014] [Indexed: 12/11/2022]
|
49
|
Ruwanpura SM, McLeod L, Brooks GD, Bozinovski S, Vlahos R, Longano A, Bardin PG, Anderson GP, Jenkins BJ. IL-6/Stat3-driven pulmonary inflammation, but not emphysema, is dependent on interleukin-17A in mice. Respirology 2014; 19:419-27. [DOI: 10.1111/resp.12243] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/25/2013] [Accepted: 12/15/2013] [Indexed: 01/12/2023]
Affiliation(s)
- Saleela M. Ruwanpura
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Gavin D. Brooks
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| | - Steven Bozinovski
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Ross Vlahos
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Anthony Longano
- Pathology; Monash Medical Centre; Clayton Victoria Australia
| | - Philip G. Bardin
- Respiratory and Sleep Medicine; Monash Medical Centre; Clayton Victoria Australia
| | - Gary P. Anderson
- Department of Medicine and Pharmacology; The University of Melbourne; Parkville Victoria Australia
| | - Brendan J. Jenkins
- Centre for Innate Immunity and Infectious Diseases; Monash Institute of Medical Research; Monash University; Clayton Victoria Australia
| |
Collapse
|
50
|
Vlahos R, Bozinovski S. Recent advances in pre-clinical mouse models of COPD. Clin Sci (Lond) 2014; 126:253-65. [PMID: 24144354 PMCID: PMC3878607 DOI: 10.1042/cs20130182] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 07/17/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023]
Abstract
COPD (chronic obstructive pulmonary disease) is a major incurable global health burden and will become the third largest cause of death in the world by 2020. It is currently believed that an exaggerated inflammatory response to inhaled irritants, in particular cigarette smoke, causes progressive airflow limitation. This inflammation, where macrophages, neutrophils and T-cells are prominent, leads to oxidative stress, emphysema, small airways fibrosis and mucus hypersecretion. The mechanisms and mediators that drive the induction and progression of chronic inflammation, emphysema and altered lung function are poorly understood. Current treatments have limited efficacy in inhibiting chronic inflammation, do not reverse the pathology of disease and fail to modify the factors that initiate and drive the long-term progression of disease. Therefore there is a clear need for new therapies that can prevent the induction and progression of COPD. Animal modelling systems that accurately reflect disease pathophysiology continue to be essential to the development of new therapies. The present review highlights some of the mouse models used to define the cellular, molecular and pathological consequences of cigarette smoke exposure and whether they can be used to predict the efficacy of new therapeutics for COPD.
Collapse
Key Words
- acute exacerbations of chronic obstructive pulmonary disease (aecopd)
- chronic obstructive pulmonary disease (copd)
- emphysema
- inflammation
- skeletal muscle wasting
- smoking
- aecopd, acute exacerbations of copd
- bal, bronchoalveolar lavage
- balf, bal fluid
- copd, chronic obstructive pulmonary disease
- gm-csf, granulocyte/macrophage colony-stimulating factor
- gold, global initiative on chronic obstructive lung disease
- gpx, glutathione peroxidase
- hdac, histone deacetylation
- il, interleukin
- ltb4, leukotriene b4
- mapk, mitogen-activated protein kinase
- mcp-1, monocyte chemotactic protein-1
- mmp, matrix metalloproteinase
- ne, neutrophil elastase
- nf-κb, nuclear factor κb
- nrf2, nuclear erythroid-related factor 2
- o2•−, superoxide radical
- onoo−, peroxynitrite
- pde, phosphodiesterase
- pi3k, phosphoinositide 3-kinase
- ros, reactive oxygen species
- rv, rhinovirus
- slpi, secretory leucocyte protease inhibitor
- sod, superoxide dismutase
- tgf-β, transforming growth factor-β
- timp, tissue inhibitor of metalloproteinases
- tnf-α, tumour necrosis factor-α
- v/q, ventilation/perfusion
Collapse
Affiliation(s)
- Ross Vlahos
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Steven Bozinovski
- *Lung Health Research Centre, Department of Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|