1
|
Zailani H, Satyanarayanan SK, Liao WC, Su KP, Chang JPC. Omega-3 Polyunsaturated Fatty Acids in Chronic Obstructive Pulmonary Disease Patients with COVID-19: A Review. Curr Nutr Rep 2025; 14:12. [PMID: 39760917 DOI: 10.1007/s13668-024-00599-9] [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] [Accepted: 11/06/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE OF THE REVIEW Mounting evidence indicates that individuals with chronic obstructive pulmonary disease (COPD) face a heightened risk of severe outcomes upon contracting coronavirus disease 2019 (COVID-19). Current medications for COVID-19 often carry side effects, necessitating alternative therapies with improved tolerance. This review explores the biological mechanisms rendering COPD patients more susceptible to severe COVID-19 and investigates the potential of omega-3 polyunsaturated fatty acids (n-3 PUFAs) in mitigating the severity of COVID-19 in COPD patients. RECENT FINDINGS Current evidence indicates that COPD patients are at an increased risk of severe COVID-19 due to factors including compromised pulmonary function, dysregulated inflammation, weakened immune response, increased oxidative stress, elevated expression of angiotensin-converting enzyme (ACE2) receptors in the lungs, and genetic predispositions. Remarkably, n-3 PUFAs exhibit the potential in ameliorating the clinical outcomes of COPD patients with COVID-19 by modulating inflammation, reinforcing the body's antioxidant defenses, reducing viral entry and replication, and enhancing immunity. N-3 PUFAs hold potential for improving COVID-19 outcomes in patients with COPD. However, there has been limited investigation into the therapeutic effects of n-3 PUFAs in enhancing clinical outcomes for COPD patients. Rigorous clinical studies are essential to evaluate the impact of n-3 PUFAs on COPD patients with concurrent COVID-19 infection.
Collapse
Grants
- MOST 109-2320-B-038-057- MY3, 110-2321-B-006-004, 110-2811-B-039-507, 110-2320-B-039-048-MY2,110-2320-B-039- 047-MY3, 110-2813-C-039-327-B, 110-2314-B-039-029-MY3, 111-2321-B-006-008, and NSTC 111-2314-B-039-041-MY3 Ministry of Science and Technology, Taiwan
- MOST 109-2320-B-038-057- MY3, 110-2321-B-006-004, 110-2811-B-039-507, 110-2320-B-039-048-MY2,110-2320-B-039- 047-MY3, 110-2813-C-039-327-B, 110-2314-B-039-029-MY3, 111-2321-B-006-008, and NSTC 111-2314-B-039-041-MY3 Ministry of Science and Technology, Taiwan
- ANHRF 109-31, 109-40, 110-13, 110-26, 110-44, 110-45, 111-27, 111-28, 111-47, 111-48, and 111-52 An-Nan Hospital, China Medical University, Tainan, Taiwan
- CMRC-CMA-2 Higher Education Sprout Project by the Ministry of Education, Taiwan
- CMRC-CMA-2 Higher Education Sprout Project by the Ministry of Education, Taiwan
- CMU 110- AWARD-02, 110-N-17, 1110-SR-73 China Medical University, Taiwan
- CMU 110- AWARD-02, 110-N-17, 1110-SR-73 China Medical University, Taiwan
- DMR-106-101, 106-227, 109-102, 109-244, 110-124, 111-245, 112-097, 112-086, 112-109 and DMR-HHC-109-11, HHC-109-12, HHC-110-10, and HHC-111-8 China Medical University Hospital
- DMR-106-101, 106-227, 109-102, 109-244, 110-124, 111-245, 112-097, 112-086, 112-109 and DMR-HHC-109-11, HHC-109-12, HHC-110-10, and HHC-111-8 China Medical University Hospital
Collapse
Affiliation(s)
- Halliru Zailani
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Nutrition, China Medical University, Taichung, Taiwan
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Senthil Kumaran Satyanarayanan
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, China
| | - Wei-Chih Liao
- Division of Pulmonary and Critical Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
| | - Kuan-Pin Su
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Jane Pei-Chen Chang
- Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan.
- College of Medicine, China Medical University, Taichung, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Child and Adolescent Psychiatry Division, Department of Psychiatry, China Medical University Hospital, No. 2 Yu-Der Rd, North District, Taichung, 404, Taiwan.
| |
Collapse
|
2
|
Lv K, Li Q, Jiang N, Chen Q. Role of TRIM29 in disease: What is and is not known. Int Immunopharmacol 2025; 147:113983. [PMID: 39755113 DOI: 10.1016/j.intimp.2024.113983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Tripartite motif-containing proteins (TRIMs), comprising the greatest subfamily of E3 ubiquitin ligases with approximately 80 members of this family, are widely distributed in mammalian cells. TRIMs actively participate in ubiquitination of target proteins, a type of post-translational modification associated with protein degradation and other functions. Tripartite motif-containing protein 29 (TRIM29), a member of the TRIM family, differs from other members of this family in that it lacks the RING finger structural domain containing cysteine and histidine residues that mediates DNA binding, protein-protein interactions, and ubiquitin ligase, at its N-terminus. The expression of TRIM29 was initially found to be associated with cancer and diabetic nephropathy progression, and antiviral immunity which is triggered by virus-derived nucleic acids binding to pattern recognition receptors (PRRs) on immune cells. Recently, TRIM29 has also been explored as a diagnostic biomarker and therapeutic target for some immune-related diseases. Here, we review the functions of TRIM29 in the progression of diseases and the inherent mechanisms, as well as the remaining gaps in the literature. A thorough understanding of the detailed regulatory mechanisms of TRIM29 will ultimately facilitate the development of different therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Kunying Lv
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China.
| |
Collapse
|
3
|
Wang J, Luo Q, Gu T, An F, Zhou Y, Min Y, Zhang R, Jiang Y. Serum-Derived Exosomal TBX2-AS1 Exacerbates COPD by Altering the M1/M2 Ratio of Macrophages through Regulating the miR-423-5p/miR-23b-3p Axis. Immunol Invest 2024:1-25. [PMID: 39589066 DOI: 10.1080/08820139.2024.2434692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
OBJECTIVE To investigate the mechanism of serum exosomes in chronic obstructive pulmonary disease (COPD), especially the effect of lncRNA TBX2-AS1 on macrophage polarization. METHODS Screen differentially expressed genes through bioinformatics analysis, detect the expression of related molecules in clinical samples and cell experiments, construct a mouse model and conduct functional rescue experiments, using various experimental techniques such as RT - qPCR, Western Blot, flow cytometry, ELISA, and luciferase reporter assay. RESULTS TBX2-AS1 is highly expressed in the serum and serum exosomes of COPD patients, and it can promote macrophage M1 polarization and inhibit M2 polarization; it exerts its role by negatively regulating the miR-423-5p/miR-23b - 3p axis, where miR-423-5p inhibits CELSR2 expression to prevent M1 polarization, and miR-23b-3p inhibits NEK6 expression to promote M2 polarization; in vivo experiments, down-regulation of CELSR2/NEK6 can reverse the promoting effect of COPD serum exosomes on lung injury and inflammation. CONCLUSION COPD serum exosomes deliver TBX2-AS1 to macrophages, regulate the miR-423-5p-CELSR2/miR-23b-3p-NEK6 pathway, affect macrophage polarization, and exacerbate the progression of COPD, providing new directions and potential targets for the diagnosis and treatment of COPD.
Collapse
Affiliation(s)
- JinHai Wang
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qing Luo
- Department of Internal Medicine, People's Hospital of Hainan Tibetan Autonomous Prefecture, Qinghai, Hainan Tibetan Autonomous Prefecture, China
| | - TiJun Gu
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - FenQin An
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YunZheng Zhou
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YePing Min
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - RuiRen Zhang
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YiMing Jiang
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| |
Collapse
|
4
|
Wen Y, Chen Q, Wang H, Xie S, Chen H, Yao W, Zhang L, Sun W, Wen J, Yang X, Chung KF, Zhang Q, Tao A, Yan J. Contribution of IL-17C-mediated macrophage polarization to Type 17 inflammation in neutrophilic asthma. Cell Commun Signal 2024; 22:557. [PMID: 39568050 PMCID: PMC11580697 DOI: 10.1186/s12964-024-01937-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND IL-17C has been described in a variety of inflammatory diseases driven by neutrophils. However, the role of IL-17C in neutrophilic asthma has not been completely characterized. METHODS The level of IL-17C in asthmatic patients and mice was assessed. Il-17c-deficient mice or mice treated with exogenous rmIL-17C were performed for OVA/CFA-induced asthmatic mice model. Pulmonary inflammation was evaluated by histological analysis, flow cytometry and cytokine analysis. Il-17re-overexpressed Raw264.7 were used in vitro to investigate the role of IL-17C in macrophage polarization. RESULTS Here, we show IL-17C were increased in serum or plasma from asthmatic patients and OVA/CFA-induced asthma mice. In the OVA/CFA-induced model, exogenous rmIL-17C aggravated neutrophil- and Type 17-dominated inflammation and promoted M1 macrophage differentiation, whereas deficiency of Il-17c reversed the pro-inflammatory phenotypes and inhibited the expansion of M1 macrophages. In vitro, IL-17C in synergy with IFN-γ induced STAT1 activation in Il-17re overexpressed Raw264.7 to upregulate M1-related genes expression, and promoted pro-inflammatory M1 polymerization, whereas IL-17C in contrast to the effect of IL-4 inhibited STAT6 activation, to reduce Raw264.7 differentiation to M2 macrophage and functional M2-related genes expression. CONCLUSIONS IL-17C promotes allergic inflammation via M1 polarization of pulmonary macrophages in neutrophilic asthma. Modulation of the IL-17C/IL-17RE axis represents a novel therapeutic target in neutrophilic asthma.
Collapse
Affiliation(s)
- Yuhuan Wen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Qile Chen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Hao Wang
- Department of Hematology, Guangzhou First People's HospitalInstitute of Blood Transfusion and Hematology, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiyun Xie
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Honglv Chen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenruo Yao
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Le Zhang
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Weimin Sun
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Junjie Wen
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaojing Yang
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College & Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Ailin Tao
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Jie Yan
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
5
|
Zhu F, Gan W, Liu H, Chen W, Zeng X. Risk factors for renal progression in patients with CKD and coexisting COPD. Int Urol Nephrol 2024:10.1007/s11255-024-04227-x. [PMID: 39400674 DOI: 10.1007/s11255-024-04227-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Chronic diseases rarely occur in isolation, and chronic kidney disease (CKD) is no exception. There has been considerable research on the interplay between the heart and kidneys, but studies on the relationship between the lungs and kidneys are less common. The interaction between pulmonary and renal functions in areas such as acid-base metabolism, chronic inflammation, and bone metabolism is increasingly gaining clinical attention. METHOD In this cohort study, we examined 480 patients with stages 3-4 CKD and COPD (GOLD stages 1 and 2) to identify risk factors that contribute to the progression of renal function to a composite endpoint, which includes a 40% decline in estimated glomerular filtration rate (eGFR) and the onset of end-stage renal disease during follow-up periods. A Cox proportional hazards regression model was used to investigate the risk factors associated with the timing of renal event endpoints in the study population. Additionally, the restricted cubic spline method was used to explore the relationship between quantitative variables and survival risk. RESULTS Our study included 480 eligible patients with an average follow-up period of 21.41 ± 14.90 months, during which 224 individuals (46.7%) experienced the composite renal endpoints. Multivariable Cox regression analysis revealed that systolic blood pressure (SBP) [1.01 (1.00-1.02), p = 0.002], hemoglobin (Hb) [HR 0.89 (0.83-0.96), p = 0.002], albumin (Alb) [0.96 (0.93-0.99), p = 0.009], and edema [1.73 (1.29-2.33), p < 0.001] were independent risk factors for the renal endpoints. CONCLUSION The adjusted multivariable Cox regression analysis demonstrated that elevated SBP and edema were factors that promoted the occurrence of composite endpoints, while higher levels of Hb and Alb were protective factors.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Wenyuan Gan
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Hui Liu
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Wenli Chen
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China.
| | - Xingruo Zeng
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China.
| |
Collapse
|
6
|
Xiang Y, Jiang Z, Yang Z, Gong S, Niu W. Ephedrine attenuates LPS-induced M1 polarization of alveolar macrophages via the PKM2-mediated glycolysis. Toxicol Res (Camb) 2024; 13:tfae166. [PMID: 39399212 PMCID: PMC11465183 DOI: 10.1093/toxres/tfae166] [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: 07/21/2024] [Revised: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024] Open
Abstract
Background Asthma is one of chronic inflammatory lung diseases in world. The important role of macrophage polarization and glycolysis in lung inflammation has attracted considerable attention. Ephedrine (EP) is a compound isolated from Ephedra and plays a regulatory role in inflammatory response, but its role in asthma and mechanism involved are not clear. Therefore, the purpose of this study was to investigate the molecular mechanism and effect of EP on lipopolysaccharide (LPS)-induced alveolar macrophage polarization and glycolysis. Methods We investigated the expression of Tnf-a, Nos2, Il10, and Arg1 using RT-PCR, as well as PKM2 and LDHA protein expression with Western blot. A CCK-8 assay was performed to determine the viability of the cells. The extracellular acidification rate (ECAR), ATP and lactate level were detected using commercial kits. Results The results revealed that EP alleviated LPS-induced NR8383 cell glycolysis and M1 polarization. Further studies found that EP enhanced the effect of 2-DG on NR8383 cell glycolysis and M1 polarization. More importantly, PKM2 inhibitor alleviated LPS-induced NR8383 cell glycolysis and M1 polarization. In addition, EP alleviated LPS-induced NR8383 cell glycolysis and M1 polarization by targeting PKM2. Conclusion It is suggested that EP alleviates LPS-induced glycolysis and M1 polarization in NR8383 cells by regulating PKM2, thereby alleviating lung injury, suggesting the involvment of alveolar macrophage polarization and glycolysis in the role of EP in asthma.
Collapse
Affiliation(s)
- Yijin Xiang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Zaifeng Jiang
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Zhangjiang Hi-TechPark, Pudong New Area, Shanghai, China
| | - Zhigang Yang
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Shaomin Gong
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| | - Weiran Niu
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai 200032, China
| |
Collapse
|
7
|
Choi K. The Role of Macrophages in Airway Disease Focusing on Porcine Reproductive and Respiratory Syndrome Virus and the Treatment with Antioxidant Nanoparticles. Viruses 2024; 16:1563. [PMID: 39459897 PMCID: PMC11512392 DOI: 10.3390/v16101563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Lung macrophage cells play a critical role in various lung diseases, and their state can change depending on the progression of the disease by inducing either an inflammatory or anti-inflammatory state. In this review, the potential therapeutic effects of treatment with antioxidant nanoparticles in air-borne diseases focusing on porcine reproductive and respiratory virus (PRRSV), considering reactive oxygen species (ROS) as one of the factors that regulate M1 and M2 macrophages in the inflammatory and anti-inflammatory states, respectively, was described. In addition, the author examines the status of protein structure research on CD163 (one of the markers of anti-inflammatory M2 macrophages) in human and veterinary lung diseases.
Collapse
MESH Headings
- Animals
- Porcine respiratory and reproductive syndrome virus/immunology
- Porcine respiratory and reproductive syndrome virus/physiology
- Antioxidants/pharmacology
- Swine
- Nanoparticles/chemistry
- Porcine Reproductive and Respiratory Syndrome/immunology
- Porcine Reproductive and Respiratory Syndrome/virology
- Porcine Reproductive and Respiratory Syndrome/drug therapy
- Humans
- Reactive Oxygen Species/metabolism
- Macrophages/immunology
- Macrophages/virology
- Macrophages/drug effects
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/immunology
- Macrophages, Alveolar/virology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/drug effects
- Receptors, Cell Surface/metabolism
- Antigens, CD/metabolism
- Antigens, CD/immunology
Collapse
Affiliation(s)
- Kyuhyung Choi
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea;
- Bundang New York Animal Hospital, Seongnam 13637, Republic of Korea
| |
Collapse
|
8
|
Lai S, Guo Z. Stem cell therapies for chronic obstructive pulmonary disease: mesenchymal stem cells as a promising treatment option. Stem Cell Res Ther 2024; 15:312. [PMID: 39300523 DOI: 10.1186/s13287-024-03940-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Chronic obstructive pulmonary disease(COPD) is an inflammatory disease characterized by the progressive and irreversible structural and functional damage of lung tissue. Although COPD is a significant global disease burden, the available treatments only ameliorate the symptoms, but cannot reverse lung damage. Researchers in regenerative medicine have examined the use of stem cell transplantation for treatment of COPD and other diseases because these cells have the potential for unlimited self-renewal and the ability to undergo directed differentiation. Stem cells are typically classified as embryonic stem cells, induced pluripotent stem cells, and adult stem cells (which includes mesenchymal stem cells [MSCs]), each with its own advantages and disadvantages regarding applications in regenerative medicine. Although the heterogeneity and susceptibility to senescence of MSCs make them require careful consideration for clinical applications. However, the low tumourigenicity and minimal ethical concerns of MSCs make them appear to be excellent candidates. This review summarizes the characteristics of various stem cell types and describes their therapeutic potential in the treatment of COPD, with a particular emphasis on MSCs. We aim to facilitate subsequent in-depth research and preclinical applications of MSCs by providing a comprehensive overview.
Collapse
Affiliation(s)
- Sumei Lai
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| | - Zhifeng Guo
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
9
|
Ashique S, Mishra N, Mantry S, Garg A, Kumar N, Gupta M, Kar SK, Islam A, Mohanto S, Subramaniyan V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03392-1. [PMID: 39196392 DOI: 10.1007/s00210-024-03392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
A significant number of deaths and disabilities worldwide are brought on by inflammatory lung diseases. Many inflammatory lung disorders, including chronic respiratory emphysema, resistant asthma, resistance to steroids, and coronavirus-infected lung infections, have severe variants for which there are no viable treatments; as a result, new treatment alternatives are needed. Here, we emphasize how oxidative imbalance contributes to the emergence of provocative lung problems that are challenging to treat. Endogenic antioxidant systems are not enough to avert free radical-mediated damage due to the induced overproduction of ROS. Pro-inflammatory mediators are then produced due to intracellular signaling events, which can harm the tissue and worsen the inflammatory response. Overproduction of ROS causes oxidative stress, which causes lung damage and various disease conditions. Invasive microorganisms or hazardous substances that are inhaled repeatedly can cause an excessive amount of ROS to be produced. By starting signal transduction pathways, increased ROS generation during inflammation may cause recurrent DNA damage and apoptosis and activate proto-oncogenes. This review provides information about new targets for conducting research in related domains or target factors to prevent, control, or treat such inflammatory oxidative stress-induced inflammatory lung disorders.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, 713212, India.
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, MP, 474005, India
| | - Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Sanjeeb Kumar Kar
- Department of Pharmaceutical Chemistry, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
10
|
Baraldo S, Bonato M, Cassia S, Casolari P, De Ferrari L, Tiné M, Baraldi F, Bigoni T, Riccio AM, Braido F, Saetta M, Papi A, Contoli M. Expression of human Interferon Regulatory Factor 3 (IRF-3) in alveolar macrophages relates to clinical and functional traits in COPD. Respir Res 2024; 25:315. [PMID: 39160551 PMCID: PMC11334339 DOI: 10.1186/s12931-024-02952-6] [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: 12/19/2023] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a frequent cause of morbidity and mortality. Dysregulated and enhanced immune-inflammatory responses have been described in COPD. Recent data showed impaired immune responses and, in particular, of interferon (IFNs) signaling pathway in these patients. AIM To evaluate in peripheral lung of COPD patients, the expression of some of the less investigated key components of the innate immune responses leading to IFN productions including: IFN-receptors (IFNAR1/IFNAR2), IRF-3 and MDA-5. Correlations with clinical traits and with the inflammatory cell profile have been assessed. METHODS Lung specimens were collected from 58 subjects undergoing thoracic surgery: 22 COPD patients, 21 smokers with normal lung function (SC) and 15 non-smoker controls (nSC). The expression of IFNAR1, IFNAR2, IRF-3 and MDA-5, of eosinophils and activated NK cells (NKp46+) were quantified in the peripheral lung by immunohistochemistry. RESULTS A significant increase of IRF-3 + alveolar macrophages were observed in COPD and SC compared with nSC subjects. However, in COPD patients, the lower the levels of IRF-3 + alveolar macrophages the lower the FEV1 and the higher the exacerbation rate. The presence of chronic bronchitis (CB) was also associated with low levels of IRF-3 + alveolar macrophages. NKp46 + cells, but not eosinophils, were increased in COPD patients compared to nSC patients (p < 0.0001). CONCLUSIONS Smoking is associated with higher levels of innate immune response as showed by higher levels of IRF-3 + alveolar macrophages and NKp46 + cells. In COPD, exacerbation rates, severe airflow obstruction and CB were associated with lower levels of IRF-3 expression, suggesting that innate immune responses characterize specific clinical traits of the disease.
Collapse
Affiliation(s)
- Simonetta Baraldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Pulmonology Unit, Ca' Foncello Hospital, Azienda Unità Locale Socio-Sanitaria 2 Marca Trevigiana, Treviso, Italy
| | - Sebastiano Cassia
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Paolo Casolari
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Laura De Ferrari
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mariaenrica Tiné
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Federico Baraldi
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Tommaso Bigoni
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Anna Maria Riccio
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Fulvio Braido
- Respiratory Clinic, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Alberto Papi
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
- Azienda Ospedaliera Universitaria Ferrara and AUSL, Ferrara, Italy
| | - Marco Contoli
- Section of Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy.
- Azienda Ospedaliera Universitaria Ferrara and AUSL, Ferrara, Italy.
| |
Collapse
|
11
|
Alateeq R, Akhtar A, De Luca SN, Chan SMH, Vlahos R. Apocynin Prevents Cigarette Smoke-Induced Anxiety-Like Behavior and Preserves Microglial Profiles in Male Mice. Antioxidants (Basel) 2024; 13:855. [PMID: 39061923 PMCID: PMC11274253 DOI: 10.3390/antiox13070855] [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: 05/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death globally and is primarily caused by cigarette smoking (CS). Neurocognitive comorbidities such as anxiety and cognitive impairments are common among people with COPD. CS-induced lung inflammation and oxidative stress may "spill-over" into the systemic circulation, driving the onset of these comorbidities. We investigated whether a prophylactic treatment with the NADPH Oxidase 2 (NOX2) inhibitor, apocynin, could prevent CS-induced neurocognitive impairments. Adult male BALB/c mice were exposed to CS (9 cigarettes/day, 5 days/week) or room air (sham) for 8 weeks with co-administration of apocynin (5 mg/kg, intraperitoneal injection once daily) or vehicle (0.01% DMSO in saline). Following 7 weeks of CS exposure, mice underwent behavioral testing to assess recognition and spatial memory (novel object recognition and Y maze, respectively) and anxiety-like behaviors (open field and elevated plus maze). Mice were then euthanized, and blood, lungs, and brains were collected. Apocynin partially improved CS-induced lung neutrophilia and reversed systemic inflammation (C-reactive protein) and oxidative stress (malondialdehyde). Apocynin exerted an anxiolytic effect in CS-exposed mice, which was associated with restored microglial profiles within the amygdala and hippocampus. Thus, targeting oxidative stress using apocynin can alleviate anxiety-like behaviors and could represent a novel strategy for managing COPD-related anxiety disorders.
Collapse
Affiliation(s)
| | | | | | | | - Ross Vlahos
- Respiratory Research Group, Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, VIC 3083, Australia; (R.A.); (A.A.); (S.N.D.L.)
| |
Collapse
|
12
|
Akdeniz YS, Özkan S. New markers in chronic obstructive pulmonary disease. Adv Clin Chem 2024; 123:1-63. [PMID: 39181619 DOI: 10.1016/bs.acc.2024.06.001] [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: 08/27/2024]
Abstract
Chronic obstructive pulmonary disease (COPD), a global healthcare and socioeconomic burden, is a multifaceted respiratory disorder that results in substantial decline in health status and life quality. Acute exacerbations of the disease contribute significantly to increased morbidity and mortality. Consequently, the identification of reliable and effective biomarkers for rapid diagnosis, prediction, and prognosis of exacerbations is imperative. In addition, biomarkers play a crucial role in monitoring responses to therapeutic interventions and exploring innovative treatment strategies. Although established markers such as CRP, fibrinogen and neutrophil count are routinely used, a universal marker is lacking. Fortunately, an increasing number of studies based on next generation analytics have explored potential biomarkers in COPD. Here we review those advances and the need for standardized validation studies in the appropriate clinical setting.
Collapse
Affiliation(s)
- Yonca Senem Akdeniz
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye.
| | - Seda Özkan
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye
| |
Collapse
|
13
|
Alwani S, Wasan EK, Badea I. Solid Lipid Nanoparticles for Pulmonary Delivery of Biopharmaceuticals: A Review of Opportunities, Challenges, and Delivery Applications. Mol Pharm 2024; 21:3084-3102. [PMID: 38828798 DOI: 10.1021/acs.molpharmaceut.4c00128] [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: 06/05/2024]
Abstract
Biopharmaceuticals such as nucleic acids, proteins, and peptides constitute a new array of treatment modalities for chronic ailments. Invasive routes remain the mainstay of administering biopharmaceuticals due to their labile nature in the biological environment. However, it is not preferred for long-term therapy due to the lack of patient adherence and clinical suitability. Therefore, alternative routes of administration are sought to utilize novel biopharmaceutical therapies to their utmost potential. Nanoparticle-mediated pulmonary delivery of biologics can facilitate both local and systemic disorders. Solid lipid nanoparticles (SLNs) afford many opportunities as pulmonary carriers due to their physicochemical stability and ability to incorporate both hydrophilic and hydrophobic moieties, thus allowing novel combinatorial drug/gene therapies. These applications include pulmonary infections, lung cancer, and cystic fibrosis, while systemic delivery of biomolecules, like insulin, is also attractive for the treatment of chronic ailments. This Review explores physiological and particle-associated factors affecting pulmonary delivery of biopharmaceuticals. It compares the advantages and limitations of SLNs as pulmonary nanocarriers along with design improvements underway to overcome these limitations. Current research illustrating various SLN designs to deliver proteins, peptides, plasmids, oligonucleotides, siRNA, and mRNA is also summarized.
Collapse
Affiliation(s)
- Saniya Alwani
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ellen K Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| |
Collapse
|
14
|
Kim GD, Lim EY, Shin HS. Macrophage Polarization and Functions in Pathogenesis of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2024; 25:5631. [PMID: 38891820 PMCID: PMC11172060 DOI: 10.3390/ijms25115631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), the major leading cause of mortality worldwide, is a progressive and irreversible respiratory condition characterized by peripheral airway and lung parenchymal inflammation, accompanied by fibrosis, emphysema, and airflow limitation, and has multiple etiologies, including genetic variance, air pollution, and repetitive exposure to harmful substances. However, the precise mechanisms underlying the pathogenesis of COPD have not been identified. Recent multiomics-based evidence suggests that the plasticity of alveolar macrophages contributes to the onset and progression of COPD through the coordinated modulation of numerous transcription factors. Therefore, this review focuses on understanding the mechanisms and functions of macrophage polarization that regulate lung homeostasis in COPD. These findings may provide a better insight into the distinct role of macrophages in COPD pathogenesis and perspective for developing novel therapeutic strategies targeting macrophage polarization.
Collapse
Affiliation(s)
- Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (G.-D.K.); (E.Y.L.)
| | - Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (G.-D.K.); (E.Y.L.)
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; (G.-D.K.); (E.Y.L.)
- Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
15
|
Mirra D, Esposito R, Spaziano G, Sportiello L, Panico F, Squillante A, Falciani M, Cerqua I, Gallelli L, Cione E, D’Agostino B. MicroRNA Monitoring in Human Alveolar Macrophages from Patients with Smoking-Related Lung Diseases: A Preliminary Study. Biomedicines 2024; 12:1050. [PMID: 38791013 PMCID: PMC11118114 DOI: 10.3390/biomedicines12051050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/18/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease that is commonly considered to be a potent driver of non-small cell lung cancer (NSCLC) development and related mortality. A growing body of evidence supports a role of the immune system, mainly played by alveolar macrophages (AMs), in key axes regulating the development of COPD or NSCLC phenotypes in response to harmful agents. MicroRNAs (miRNAs) are small non-coding RNAs that influence most biological processes and interfere with several regulatory pathways. The purpose of this study was to assess miRNA expression patterns in patients with COPD, NSCLC, and ever- or never-smoker controls to explore their involvement in smoking-related diseases. Bronchoalveolar lavage (BAL) specimens were collected from a prospective cohort of 43 sex-matched subjects to determine the expressions of hsa-miR-223-5p, 16-5p, 20a-5p, -17-5p, 34a-5p and 106a-5p by RT-PCR. In addition, a bioinformatic analysis of miRNA target genes linked to cancer was performed. Distinct and common miRNA expression levels were identified in each pathological group, suggesting their possible role as an index of NSCLC or COPD microenvironment. Moreover, we identified miRNA targets linked to carcinogenesis using in silico analysis. In conclusion, this study identified miRNA signatures in AMs, allowing us to understand the molecular mechanisms underlying smoking-related conditions and potentially providing new insights for diagnosis or pharmacological treatment.
Collapse
Affiliation(s)
- Davida Mirra
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (B.D.)
| | - Renata Esposito
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (B.D.)
| | - Giuseppe Spaziano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (B.D.)
| | - Liberata Sportiello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy;
- Department of Experimental Medicine-Section of Pharmacology “L. Donatelli”, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Francesca Panico
- Department of Health Sciences, University of “Magna Graecia”, 88100 Catanzaro, Italy; (F.P.); (L.G.)
| | | | - Maddalena Falciani
- Pulmonary and Critical Care Medicine, Ospedale Scarlato, 84018 Scafati, Italy;
| | - Ida Cerqua
- Department of Pharmacy, University Federico II of Naples, Via D. Montesano 49, 80131 Naples, Italy;
| | - Luca Gallelli
- Department of Health Sciences, University of “Magna Graecia”, 88100 Catanzaro, Italy; (F.P.); (L.G.)
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Bruno D’Agostino
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (B.D.)
| |
Collapse
|
16
|
Yehia D, Leung C, Sin DD. Clinical utilization of airway inflammatory biomarkers in the prediction and monitoring of clinical outcomes in patients with chronic obstructive pulmonary disease. Expert Rev Mol Diagn 2024; 24:409-421. [PMID: 38635513 DOI: 10.1080/14737159.2024.2344777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) accounts for 545 million people living with chronic respiratory disorders and is the third leading cause of morbidity and mortality around the world. COPD is a progressive disease, characterized by episodes of acute worsening of symptoms such as cough, dyspnea, and sputum production. AREAS COVERED Airway inflammation is a prominent feature of COPD. Chronic airway inflammation results in airway structural remodeling and emphysema. Persistent airway inflammation is a treatable trait of COPD and plays a significant role in disease development and progression. In this review, the authors summarize the current and emerging biomarkers that reveal the heterogeneity of airway inflammation subtypes, clinical outcomes, and therapeutic response in COPD. EXPERT OPINION Airway inflammation can be broadly categorized as eosinophilic (type 2 inflammation) and non-eosinophilic (non-type 2 inflammation) in COPD. Currently, blood eosinophil counts are incorporated in clinical practice guidelines to identify COPD patients who are at a higher risk of exacerbations and lung function decline, and who are likely to respond to inhaled corticosteroids. As new therapeutics are being developed for the chronic management of COPD, it is essential to identify biomarkers that will predict treatment response.
Collapse
Affiliation(s)
- Dina Yehia
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Clarus Leung
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Kim JW, Kim JH, Kim CY, Jeong JS, Ko JW, Kim TW. Korean Red Ginseng suppresses emphysematous lesions induced by cigarette smoke condensate through inhibition of macrophage-driven apoptosis pathways. J Ginseng Res 2024; 48:181-189. [PMID: 38465217 PMCID: PMC10920012 DOI: 10.1016/j.jgr.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/12/2023] [Accepted: 11/02/2023] [Indexed: 03/12/2024] Open
Abstract
Background Cigarette smoke is generally accepted as a major contributor to chronic obstructive pulmonary disease (COPD), which is characterized by emphysematous lesions. In this study, we investigated the protective effects of Korean Red Ginseng (KRG) against cigarette smoke condensate (CSC)-induced emphysema. Methods Mice were instilled with 50 mg/kg of CSC intranasally once a week for 4 weeks, KRG was administered to the mice once daily for 4 weeks at doses of 100 or 300 mg/kg, and dexamethasone (DEX, positive control) was administered to the mice once daily for 2 weeks at 3 mg/kg. Results KRG markedly decreased the macrophage population in bronchoalveolar lavage fluid and reduced emphysematous lesions in the lung tissues. KRG suppressed CSC-induced apoptosis as revealed by terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling staining and Caspase 3 immunohistochemistry. Additionally, KRG effectively inhibited CSC-mediated activation of Bcl-2-associated X protein/Caspase 3 signaling, followed by the induction of cell survival signaling, including vascular endothelial growth factor/phosphoinositide 3-kinase/protein kinase B in vivo and in vitro. The DEX group also showed similar improved results in vivo and in vitro. Conclusion Taken together, KRG effectively inhibits macrophage-mediated emphysema induced by CSC exposure, possibly via the suppression of pro-apoptotic signaling, which results in cell survival pathway activation. These findings suggest that KRG has therapeutic potential for the prevention of emphysema in COPD patients.
Collapse
Affiliation(s)
- Jeong-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| | - Jin-Hwa Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| | - Chang-Yeop Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| | - Ji-Soo Jeong
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| | - Je-Won Ko
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| | - Tae-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, 99 Daehak-ro, Daejeon, Republic of Korea
| |
Collapse
|
18
|
Xiong K, Ao K, Wei W, Dong J, Li J, Yang Y, Tang B, Li Y. Periodontitis aggravates COPD through the activation of γδ T cell and M2 macrophage. mSystems 2024; 9:e0057223. [PMID: 38214520 PMCID: PMC10878042 DOI: 10.1128/msystems.00572-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic systemic inflammatory disease with high morbidity and mortality. Periodontitis exacerbates COPD progression; however, the immune mechanisms by which periodontitis affects COPD remain unclear. Here, by constructing periodontitis and COPD mouse models, we demonstrated that periodontitis and COPD could mutually aggravate disease progression. For the first time, we found that the progression was associated with the activation of γδ T cells and M2 macrophages, and M2 polarization of macrophages was affected by γδ T cells activation. In the lung tissues of COPD with periodontitis, the activation of γδ T cells finally led to the increase of IL 17 and IFN γ expression and M2 macrophage polarization. Furthermore, we found that the periodontitis-associated bacteria Porphyromonas gingivalis (P. gingivalis) promoted the activation of γδ T cells and M2 macrophages ex vivo. The data from clinical bronchoalveolar lavage fluid (BALF) samples were consistent with the in vivo and ex vivo experiments. For the first time, our results identified the crucial role of γδ T-M2 immune mechanism in mediating periodontitis-promoted COPD progression. Therefore, targeting at periodontitis treatment and the γδ T-M2 immune mechanism might provide a new practical strategy for COPD prevention or control.IMPORTANCEPeriodontitis exacerbates chronic obstructive pulmonary disease (COPD) progression. For the first time, the current study identified that the impact of periodontitis on COPD progression was associated with the activation of γδ T cells and M2 macrophages and that M2 polarization of macrophages was affected by γδ T cells activation. The results indicated that targeting at periodontitis treatment and the γδ T-M2 immune mechanism might provide a new practical strategy for COPD prevention or control.
Collapse
Affiliation(s)
- Kaixin Xiong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Keping Ao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Jiajia Dong
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jia Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yutao Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Boyu Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Conservation Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Noh M, Sim JY, Kim J, Ahn JH, Min HY, Lee JU, Park JS, Jeong JY, Lee JY, Lee SY, Lee HJ, Park CS, Lee HY. Particulate matter-induced metabolic recoding of epigenetics in macrophages drives pathogenesis of chronic obstructive pulmonary disease. JOURNAL OF HAZARDOUS MATERIALS 2024; 464:132932. [PMID: 37988864 DOI: 10.1016/j.jhazmat.2023.132932] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/19/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a group of illnesses associated with unresolved inflammation in response to toxic environmental stimuli. Persistent exposure to PM is a major risk factor for COPD, but the underlying mechanism remains unclear. Using our established mouse model of PM-induced COPD, we find that repeated PM exposure provokes macrophage-centered chronic inflammation and COPD development. Mechanistically, chronic PM exposure induces transcriptional downregulation of HAAO, KMO, KYNU, and QPRT in macrophages, which are the enzymes of de novo NAD+ synthesis pathway (kynurenine pathway; KP), via elevated chromatin binding of the CCCTC-binding factor (CTCF) near the transcriptional regulatory regions of the enzymes. Subsequent reduction of NAD+ and SIRT1 function increases histone acetylation, resulting in elevated expression of pro-inflammatory genes in PM-exposed macrophages. Activation of SIRT1 by nutraceutical resveratrol mitigated PM-induced chronic inflammation and COPD development. In agreement, increased levels of histone acetylation and decreased expression of KP enzymes were observed in pulmonary macrophages of COPD patients. We newly provide an evidence that dysregulated NAD+ metabolism and consecutive SIRT1 deficiency significantly contribute to the pathological activation of macrophages during PM-mediated COPD pathogenesis. Additionally, targeting PM-induced intertwined metabolic and epigenetic reprogramming in macrophages is an effective strategy for COPD treatment.
Collapse
Affiliation(s)
- Myungkyung Noh
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jeong Yeon Sim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jisung Kim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Natural Products Research Institute, Seoul National University, Seoul 08826, South Korea
| | - Jong-Uk Lee
- Department of Medical Bioscience, Graduate School, Soonchunhyang University, 22, Soonchunhyang-ro, Asan 31538, South Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Jae Young Lee
- Department of Environmental and Safety Engineering, Ajou University, Suwon 16499, South Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Gyeonggi do, South Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
20
|
Zhou X, Zhang C, Yang S, Yang L, Luo W, Zhang W, Zhang X, Chao J. Macrophage-derived MMP12 promotes fibrosis through sustained damage to endothelial cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132733. [PMID: 37816293 DOI: 10.1016/j.jhazmat.2023.132733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/16/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023]
Abstract
Macrophages are essential for the maintenance of endothelial cell function. However, the potential impact and mechanisms of crosstalk between macrophages and endothelial cells during silicosis progression remain unexplored. To fill this knowledge gap, a mouse model of silicosis was established. Single cell sequencing, spatial transcriptome sequencing, western blotting, immunofluorescence staining, tube-forming and wound healing assays were used to explore the effects of silicon dioxide on macrophage-endothelial interactions. To investigate the mechanism of macrophage-mediated fibrosis, MMP12 was specifically inactivated using siRNA and pharmacological approaches, and macrophages were depleted using disodium chlorophosphite liposomes. Compared to the normal saline group, the silica dust group showed altered macrophage-endothelial interactions. Matrix metalloproteinase family member MMP12 was identified as a key mediator of the altered function of macrophage-endothelial interactions after silica exposure, which was accompanied by pro-inflammatory macrophage activation and fibrotic progression. By using ablation strategies, macrophage-derived MMP12 was shown to mediate endothelial cell dysfunction by accumulating on the extracellular matrix. During the inflammatory phase of silicosis, MMP12 secreted by pro-inflammatory macrophages caused decreased endothelial cell viability, reduced migration, decreased trans-endothelial resistance and increased permeability; while during the fibrotic phase, macrophage-derived MMP12 sustained endothelial cell injury through accumulation on the extracellular matrix.
Collapse
Affiliation(s)
- Xinbei Zhou
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Cong Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Shaoqi Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Liliang Yang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Wei Luo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China; Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Wei Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Xinxin Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, China; Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China; School of Medicine, Xizang Minzu University, Xianyang, Shanxi, 712082, China.
| |
Collapse
|
21
|
Lee J, Mohammad N, Lu Y, Oshins R, Aranyos A, Brantly M. Alpha-defensins inhibit ERK/STAT3 signaling during monocyte-macrophage differentiation and impede macrophage function. Respir Res 2023; 24:309. [PMID: 38082274 PMCID: PMC10714504 DOI: 10.1186/s12931-023-02605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Alpha-1-antitrypsin deficiency (AATD) is a genetic disorder associated with a 5-tenfold decrease in lung levels of alpha-1-antitrypsin (AAT) and an increased risk for obstructive lung disease. α-defensins are cationic broad-spectrum cytotoxic and pro-inflammatory peptides found in the azurophilic granules of neutrophils. The concentration of α-defensins is less than 30 nM in the bronchoalveolar lavage fluid of healthy controls but is up to 6 μM in AATD individuals with significant lung function impairment. Alveolar macrophages are generally classified into pro-inflammatory (M1) or anti-inflammatory (M2) subsets that play distinct roles in the initiation and resolution of inflammation. Therefore, monocyte-macrophage differentiation should be tightly controlled to maintain lung integrity. In this study, we determined the effect of α-defensins on monocyte-macrophage differentiation and identified the molecular mechanism of this effect. The results of this study demonstrate that 2.5 μM of α-defensins inhibit the phosphorylation of ERK1/2 and STAT3 and suppress the expression of M2 macrophage markers, CD163 and CD206. In addition, a scratch assay shows that the high concentration of α-defensins inhibits cell movement by ~ 50%, and the phagocytosis assay using flow cytometry shows that α-defensins significantly reduce the bacterial phagocytosis rate of monocyte-derived macrophages (MDMs). To examine whether exogenous AAT is able to alleviate the inhibitory effect of α-defensins on macrophage function, we incubated MDMs with AAT prior to α-defensin treatment and demonstrate that AAT improves the migratory ability and phagocytic ability of MDMs compared with MDMs incubated only with α-defensins. Taken together, this study suggests that a high concentration of α-defensins inhibits the activation of ERK/STAT3 signaling, negatively regulates the expression of M2 macrophage markers, and impairs innate immune function of macrophages.
Collapse
Affiliation(s)
- Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Naweed Mohammad
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Yuanqing Lu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Regina Oshins
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Alek Aranyos
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Mark Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
22
|
Zhang M, Lu H, Xie L, Liu X, Cun D, Yang M. Inhaled RNA drugs to treat lung diseases: Disease-related cells and nano-bio interactions. Adv Drug Deliv Rev 2023; 203:115144. [PMID: 37995899 DOI: 10.1016/j.addr.2023.115144] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/07/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
In recent years, RNA-based therapies have gained much attention as biomedicines due to their remarkable therapeutic effects with high specificity and potency. Lung diseases offer a variety of currently undruggable but attractive targets that could potentially be treated with RNA drugs. Inhaled RNA drugs for the treatment of lung diseases, including asthma, chronic obstructive pulmonary disease, cystic fibrosis, and acute respiratory distress syndrome, have attracted more and more attention. A variety of novel nanoformulations have been designed and attempted for the delivery of RNA drugs to the lung via inhalation. However, the delivery of RNA drugs via inhalation poses several challenges. It includes protection of the stability of RNA molecules, overcoming biological barriers such as mucus and cell membrane to the delivery of RNA molecules to the targeted cytoplasm, escaping endosomal entrapment, and circumventing unwanted immune response etc. To address these challenges, ongoing researches focus on developing innovative nanoparticles to enhance the stability of RNA molecules, improve cellular targeting, enhance cellular uptake and endosomal escape to achieve precise delivery of RNA drugs to the intended lung cells while avoiding unwanted nano-bio interactions and off-target effects. The present review first addresses the pathologic hallmarks of different lung diseases, disease-related cell types in the lung, and promising therapeutic targets in these lung cells. Subsequently we highlight the importance of the nano-bio interactions in the lung that need to be addressed to realize disease-related cell-specific delivery of inhaled RNA drugs. This is followed by a review on the physical and chemical characteristics of inhaled nanoformulations that influence the nano-bio interactions with a focus on surface functionalization. Finally, the challenges in the development of inhaled nanomedicines and some key aspects that need to be considered in the development of future inhaled RNA drugs are discussed.
Collapse
Affiliation(s)
- Mengjun Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China; School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Haoyu Lu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China
| | - Liangkun Xie
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China
| | - Xulu Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China.
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016 Shenyang, China; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
23
|
Rahman M, Sompa SI, Introna M, Upadhyay S, Ganguly K, Palmberg L. Lipid from electronic cigarette-aerosol both with and without nicotine induced pro-inflammatory macrophage polarization and disrupted phagocytosis. J Inflamm (Lond) 2023; 20:39. [PMID: 37978397 PMCID: PMC10655339 DOI: 10.1186/s12950-023-00367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
Clinical cases and experimental evidence revealed that electronic cigarettes (ECIG) induce serious adverse health effects, but underlying mechanisms remain to be fully uncovered. Based on recent exploratory evidence, investigating the effects of ECIG on macrophages can broadly define potential mechanisms by focusing on the effect of ECIG exposure with or without nicotine. Here we investigated the effect of ECIG-aerosol exposure on macrophages (MQ) phenotype, inflammatory response, and function of macrophages.MQ were cultured at air liquid interface and exposed to ECIG-aerosol. Oxidative stress was determined by reactive oxygen species (ROS), heat shock protein 60 (HSP60), glutathione peroxidase (GPx) and heme oxygenase1 (HMOX1). Lipid accumulation and lipid peroxidation were defined by lipid staining and level of malondialdehyde (MDA) respectively. MQ polarization was identified by surface expression markers CD86, CD11C and CD206 as well as pro-inflammatory and anti-inflammatory cytokines in gene and protein level. Phagocytosis of E. coli by MQ was investigated by fluorescence-based phagocytosis assay.ECIG-aerosol exposure in presence or absence of nicotine induced oxidative stress evidenced by ROS, HSP60, GPx, GPx4 and HMOX1 upregulation in MQ. ECIG-aerosol exposure induced accumulation of lipids and the lipid peroxidation product MDA in MQ. Pro-inflammatory MQ (M1) markers CD86 and CD11C but not anti-inflammatory MQ (M2) marker CD206 were upregulated in response to ECIG-aerosol exposure. In addition, ECIG induced pro-inflammatory cytokines IL-1beta and IL-8 in gene level and IL-6, IL-8, and IL-1beta in protein level whereas ECIG exposure downregulated anti-inflammatory cytokine IL-10 in protein level. Phagocytosis activity of MQ was downregulated by ECIG exposure. shRNA mediated lipid scavenger receptor 'CD36' silencing inhibited ECIG-aerosol-induced pro-inflammatory MQ polarization and recovered phagocytic activity of MQ.ECIG exposure alters lung lipid homeostasis and thus induced inflammation by inducing M1 type MQ and impair phagocytic function, which could be a potential cause of ECIG-induced lung inflammation in healthy and inflammatory exacerbation in disease condition.
Collapse
Affiliation(s)
- Mizanur Rahman
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Shanzina Iasmin Sompa
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Micol Introna
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
24
|
Liu Y, Rajeevan H, Simonov M, Lee S, Wilson FP, Desir GV, Vinetz JM, Yan X, Wang Z, Clark BJ, Possick JD, Price C, Lutchmansingh DD, Ortega H, Zaeh S, Gomez JVL, Cohn L, Gautam S, Chupp GL. Differences in Mortality Among Patients With Asthma and COPD Hospitalized With COVID-19. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:3383-3390.e3. [PMID: 37454926 PMCID: PMC10787810 DOI: 10.1016/j.jaip.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/14/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND It remains unclear whether patients with asthma and/or chronic obstructive pulmonary disease (COPD) are at increased risk for severe coronavirus disease 2019 (COVID-19). OBJECTIVE Compare in-hospital COVID-19 outcomes among patients with asthma, COPD, and no airway disease. METHODS A retrospective cohort study was conducted on 8,395 patients admitted with COVID-19 between March 2020 and April 2021. Airway disease diagnoses were defined using International Classification of Diseases, 10th Revision codes. Mortality and sequential organ failure assessment (SOFA) scores were compared among groups. Logistic regression analysis was used to identify and adjust for confounding clinical features associated with mortality. RESULTS The median SOFA score in patients without airway disease was 0.32 and mortality was 11%. In comparison, asthma patients had lower SOFA scores (median 0.15; P < .01) and decreased mortality, even after adjusting for age, diabetes, and other confounders (odds ratio 0.65; P = .01). Patients with COPD had higher SOFA scores (median 0.86; P < .01) and increased adjusted odds of mortality (odds ratio 1.40; P < .01). Blood eosinophil count of 200 cells/μL or greater, a marker of type 2 inflammation, was associated with lower mortality across all groups. Importantly, patients with asthma showed improved outcomes even after adjusting for eosinophilia, indicating that noneosinophilic asthma was associated with protection as well. CONCLUSIONS COVID-19 severity was increased in patients with COPD and decreased in those with asthma, eosinophilia, and noneosinophilic asthma, independent of clinical confounders. These findings suggest that COVID-19 severity may be influenced by intrinsic immunological factors in patients with airway diseases, such as type 2 inflammation.
Collapse
Affiliation(s)
- Yunqing Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn
| | - Haseena Rajeevan
- Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, Conn; Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Michael Simonov
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Seohyuk Lee
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - F Perry Wilson
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn; Clinical and Translational Research Accelerator, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Gary V Desir
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Xiting Yan
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn; Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, Conn
| | - Brian J Clark
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Jennifer D Possick
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Christina Price
- Section of Allergy and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Denyse D Lutchmansingh
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Hector Ortega
- Clinical Development, Nexstone Immunology, San Diego, Calif
| | - Sandra Zaeh
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Jose Villa-Lobos Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Lauren Cohn
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Samir Gautam
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
25
|
Petraroia I, Ghidotti P, Bertolini G, Pontis F, Roz L, Balsamo M, Suatoni P, Pastorino U, Ferretti AM, Sozzi G, Fortunato O. Extracellular vesicles from subjects with COPD modulate cancer initiating cells phenotype through HIF-1α shuttling. Cell Death Dis 2023; 14:681. [PMID: 37838700 PMCID: PMC10576796 DOI: 10.1038/s41419-023-06212-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a risk factor for lung cancer development. COPD induces activation of hypoxia-induced signaling, causing remodeling of surrounding microenvironmental cells also modulating the release and cargo of their extracellular vesicles (EVs). We aimed to evaluate the potential role of circulating EVs from COPD subjects in lung cancer onset. Plasma-EVs were isolated by ultracentrifugation from heavy smoker volunteers with (COPD-EVs) or without (heavy smoker-EVs, HS-EV) COPD and characterized following MISEV guidelines. Immortalized human bronchial epithelial cells (CDK4, hTERT-HBEC3-KT), genetically modified with different oncogenic alterations commonly found in lung cancer (sh-p53, KRASV12), were used to test plasma-EVs pro-tumorigenic activity in vitro. COPD-EVs mainly derived from immune and endothelial cells. COPD-EVs selectively increased the subset of CD133+CXCR4+ metastasis initiating cells (MICs) in HBEC-sh-p53-KRASV12high cells and stimulated 3D growth, migration/invasion, and acquisition of mesenchymal traits. These effects were not observed in HBEC cells bearing single oncogenic mutation (sh-p53 or KRASV12). Mechanistically, hypoxia-inducible factor 1-alpha (HIF-1α) transferred from COPD-EVs triggers CXCR4 pathway activation that in turn mediates MICs expansion and acquisition of pro-tumorigenic effects. Indeed, HIF-1α inhibition or CXCR4 silencing prevented the acquisition of malignant traits induced by COPD-EVs alone. Hypoxia recapitulates the effects observed with COPD-EVs in HBEC-sh-p53-KRASV12high cells. Notably, higher levels of HIF-1α were observed in EVs from COPD subjects who subsequently developed cancer compared to those who remained cancer-free. Our findings support a role of COPD-EVs to promote the expansion of MICs in premalignant epithelial cells through HIF-1α-CXCR4 axis activation thereby potentially sustaining lung cancer progression.
Collapse
Affiliation(s)
- Ilaria Petraroia
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Patrizia Ghidotti
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Giulia Bertolini
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.
| | - Francesca Pontis
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Luca Roz
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Melissa Balsamo
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Paola Suatoni
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Ugo Pastorino
- Thoracic Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Gabriella Sozzi
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Orazio Fortunato
- Epigenomics and biomarkers of solid tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
26
|
Błach J, Siedliński M, Sydor W. Immunology in COPD and the use of combustible cigarettes and heated tobacco products. Eur J Med Res 2023; 28:397. [PMID: 37794516 PMCID: PMC10548761 DOI: 10.1186/s40001-023-01374-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/18/2023] [Indexed: 10/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common chronic respiratory diseases, characterised by high morbidity and mortality. COPD is characterised by a progressive decline of lung function caused by chronic inflammatory reactions in the lung tissue due to continual exposure to harmful molecules by inhalation. As prevention plays a very important role in COPD, quitting smoking is the most important factor in reducing the decline in lung function. Unfortunately, many people are unable to break their nicotine addiction. This paper summarises current knowledge about combustible cigarettes (CSs) and alternative tobacco products such as heated tobacco products (HTPs) in COPD. The paper focuses on the immunological aspects of COPD and the influence of tobacco products on lung tissue immunology. There are differences in research results between HTPs and CSs in favour of HTPs. More long-term studies are needed to look at the effects of HTPs, especially in COPD. However, there is no doubt that it would be best for patients to give up their nicotine addiction completely.
Collapse
Affiliation(s)
- Justyna Błach
- Department of Clinical Immunology, UCH, Cracow, Poland.
| | - Mateusz Siedliński
- Department of Internal Medicine and Rural Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Wojciech Sydor
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
27
|
Zheng W, Zhou Z, Guo X, Zuo X, Zhang J, An Y, Zheng H, Yue Y, Wang G, Wang F. Efferocytosis and Respiratory Disease. Int J Mol Sci 2023; 24:14871. [PMID: 37834319 PMCID: PMC10573909 DOI: 10.3390/ijms241914871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Cells are the smallest units that make up living organisms, which constantly undergo the processes of proliferation, differentiation, senescence and death. Dead cells need to be removed in time to maintain the homeostasis of the organism and keep it healthy. This process is called efferocytosis. If the process fails, this may cause different types of diseases. More and more evidence suggests that a faulty efferocytosis process is closely related to the pathological processes of respiratory diseases. In this review, we will first introduce the process and the related mechanisms of efferocytosis of the macrophage. Secondly, we will propose some methods that can regulate the function of efferocytosis at different stages of the process. Next, we will discuss the role of efferocytosis in different lung diseases and the related treatment approaches. Finally, we will summarize the drugs that have been applied in clinical practice that can act upon efferocytosis, in order to provide new ideas for the treatment of lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guoqiang Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.Z.); (Z.Z.); (X.G.); (X.Z.); (J.Z.); (Y.A.); (H.Z.); (Y.Y.)
| | - Fang Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.Z.); (Z.Z.); (X.G.); (X.Z.); (J.Z.); (Y.A.); (H.Z.); (Y.Y.)
| |
Collapse
|
28
|
Staitieh BS, Hu X, Yeligar SM, Auld SC. Paired ATAC- and RNA-seq offer insight into the impact of HIV on alveolar macrophages: a pilot study. Sci Rep 2023; 13:15276. [PMID: 37714998 PMCID: PMC10504379 DOI: 10.1038/s41598-023-42644-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
People with HIV remain at greater risk for both infectious and non-infectious pulmonary diseases even after antiretroviral therapy initiation and CD4 cell count recovery. These clinical risks reflect persistent HIV-mediated defects in innate and adaptive immunity, including in the alveolar macrophage, a key innate immune effector in the lungs. In this proof-of-concept pilot study, we leveraged paired RNA-seq and ATAC-seq analyses of human alveolar macrophages obtained with research bronchoscopy from people with and without HIV to highlight the potential for recent methodologic advances to generate novel hypotheses about biological pathways that may contribute to impaired pulmonary immune function in people with HIV. In addition to 35 genes that were differentially expressed in macrophages from people with HIV, gene set enrichment analysis identified six gene sets that were differentially regulated. ATAC-seq analysis revealed 115 genes that were differentially accessible for people with HIV. Data-driven integration of the findings from these complementary, high-throughput techniques using xMWAS identified distinct clusters involving lipoprotein lipase and inflammatory pathways. By bringing together transcriptional and epigenetic data, this analytic approach points to several mechanisms, including previously unreported pathways, that warrant further exploration as potential mediators of the increased risk of pulmonary disease in people with HIV.
Collapse
Affiliation(s)
- Bashar S Staitieh
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, 615 Michael St NE, Ste 200, Atlanta, GA, 30322, USA
- Grady Health System, Atlanta, GA, USA
| | - Xin Hu
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, 615 Michael St NE, Ste 200, Atlanta, GA, 30322, USA
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Samantha M Yeligar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, 615 Michael St NE, Ste 200, Atlanta, GA, 30322, USA
- Veterans Affairs Atlanta Healthcare System, Decatur, GA, USA
| | - Sara C Auld
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Emory University, 615 Michael St NE, Ste 200, Atlanta, GA, 30322, USA.
- Departments of Epidemiology and Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
29
|
Bedford R, Smith G, Rothwell E, Martin S, Medhane R, Casentieri D, Daunt A, Freiberg G, Hollings M. A multi-organ, lung-derived inflammatory response following in vitro airway exposure to cigarette smoke and next-generation nicotine delivery products. Toxicol Lett 2023; 387:35-49. [PMID: 37774809 DOI: 10.1016/j.toxlet.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Despite increasing use of in vitro models that closely resemble in vivo human biology, their application in understanding downstream effects of airway toxicity, such as inflammation, are at an early stage. In this study, we used various assays to examine the inflammatory response induced in MucilAir™ tissues and A549 cells exposed to three products known to induce toxicity. Reduced barrier integrity was observed in tissues following exposure to each product, with reduced viability and increased cytotoxicity also shown. Similar changes in viability were also observed in A549 cells. Furthermore, whole cigarette smoke (CS) induced downstream phenotypic THP-1 changes and endothelial cell adhesion, an early marker of atherosclerosis. In contrast, exposure to next-generation delivery product (NGP) aerosol did not induce this response. Cytokine, histological and RNA analysis highlighted increased biomarkers linked to inflammatory pathways and immune cell differentiation following exposure to whole cigarette smoke, including GM-CSF, IL-1β, cleaved caspase-3 and cytochrome P450 enzymes. As a result of similar observations in human airway inflammation, we propose that our exposure platform could act as a representative model for studying such events in vitro. Furthermore, this model could be used to test the inflammatory or anti-inflammatory impact posed by inhaled compounds delivered to the lung.
Collapse
Affiliation(s)
- R Bedford
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - G Smith
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - E Rothwell
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - S Martin
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - R Medhane
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - D Casentieri
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - A Daunt
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - G Freiberg
- Labcorp Early Development Laboratories Limited, Eye, UK
| | - M Hollings
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| |
Collapse
|
30
|
Joshi I, Devine AJ, Joshi R, Smith NJ, Varisco BM. A titratable murine model of progressive emphysema using tracheal porcine pancreatic elastase. Sci Rep 2023; 13:15259. [PMID: 37709810 PMCID: PMC10502133 DOI: 10.1038/s41598-023-41527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Progressive emphysema often leads to end-stage lung disease. Most mouse models of emphysema are typically modest (i.e. cigarette smoke exposure), and changes over time are difficult to quantify. The tracheal porcine pancreatic elastase model (PPE) produces severe injury, but the literature is conflicted as to whether emphysema improves, is stable, or progresses over time. We hypothesized a threshold of injury below which repair would occur and above which emphysema would be stable or progress. We treated 8-week-old C57BL6 mixed sex mice with 0, 0.5, 2, or 4 activity units of PPE in 100 µL PBS and performed lung stereology at 21 and 84 days. There were no significant differences in weight gain or mouse health. Despite minimal emphysema at 21-days in the 0.5 units group (2.8 µm increased mean linear intercept, MLI), MLI increased by 4.6 µm between days 21 and 84 (p = 0.0007). In addition to larger MLI at 21 days in 2- and 4-unit groups, MLI increases from day 21 to 84 were 17.2 and 34 µm respectively (p = 0.002 and p = 0.0001). Total lung volume increased, and alveolar surface area decreased with time and injury severity. Contrary to our hypothesis, we found no evidence of alveolar repair over time. Airspace destruction was both progressive and accelerative. Future mechanistic studies in lung immunity, mechano-biology, senescence, and cell-specific changes may lead to novel therapies to slow or halt progressive emphysema in humans.
Collapse
Affiliation(s)
- Imani Joshi
- College of Arts and Sciences, Xavier University, Cincinnati, OH, USA
| | - Andrew J Devine
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rashika Joshi
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Noah J Smith
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian M Varisco
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- University of Arkansas for Medical Sciences, 1 Children's Way Slot 663, Little Rock, AR, 72202, USA.
| |
Collapse
|
31
|
Strizova Z, Benesova I, Bartolini R, Novysedlak R, Cecrdlova E, Foley L, Striz I. M1/M2 macrophages and their overlaps - myth or reality? Clin Sci (Lond) 2023; 137:1067-1093. [PMID: 37530555 PMCID: PMC10407193 DOI: 10.1042/cs20220531] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 08/03/2023]
Abstract
Macrophages represent heterogeneous cell population with important roles in defence mechanisms and in homoeostasis. Tissue macrophages from diverse anatomical locations adopt distinct activation states. M1 and M2 macrophages are two polarized forms of mononuclear phagocyte in vitro differentiation with distinct phenotypic patterns and functional properties, but in vivo, there is a wide range of different macrophage phenotypes in between depending on the microenvironment and natural signals they receive. In human infections, pathogens use different strategies to combat macrophages and these strategies include shaping the macrophage polarization towards one or another phenotype. Macrophages infiltrating the tumours can affect the patient's prognosis. M2 macrophages have been shown to promote tumour growth, while M1 macrophages provide both tumour-promoting and anti-tumour properties. In autoimmune diseases, both prolonged M1 activation, as well as altered M2 function can contribute to their onset and activity. In human atherosclerotic lesions, macrophages expressing both M1 and M2 profiles have been detected as one of the potential factors affecting occurrence of cardiovascular diseases. In allergic inflammation, T2 cytokines drive macrophage polarization towards M2 profiles, which promote airway inflammation and remodelling. M1 macrophages in transplantations seem to contribute to acute rejection, while M2 macrophages promote the fibrosis of the graft. The view of pro-inflammatory M1 macrophages and M2 macrophages suppressing inflammation seems to be an oversimplification because these cells exploit very high level of plasticity and represent a large scale of different immunophenotypes with overlapping properties. In this respect, it would be more precise to describe macrophages as M1-like and M2-like.
Collapse
Affiliation(s)
- Zuzana Strizova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Iva Benesova
- Department of Immunology, Second Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Robin Bartolini
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Rene Novysedlak
- Third Department of Surgery, First Faculty of Medicine, Charles University and University Hospital Motol, V Uvalu 84, 15006, Prague, Czech Republic
| | - Eva Cecrdlova
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lily Koumbas Foley
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, U.K
| | - Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
32
|
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
|
33
|
Papakonstantinou GI, Psalla D, Pourlis A, Stylianaki I, Athanasiou LV, Tzika E, Meletis E, Kostoulas P, Maragkakis G, Christodoulopoulos G, Papaioannou N, Papatsiros VG. Histopathological Pulmonary Lesions in 1st-Day Newborn Piglets Derived from PRRSV-1 MLV Vaccinated Sows at the Last Stage of Gestation. Life (Basel) 2023; 13:1609. [PMID: 37511984 PMCID: PMC10381811 DOI: 10.3390/life13071609] [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: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Modified live virus (MLV) vaccines for the control of porcine respiratory and reproductive syndrome virus (PRRSV) have been associated with the vertical and horizontal transmission of vaccine viruses. The present study aimed to describe pathological lung lesions in piglets born by gilts vaccinated with PRRSV-1 MLV. In total, 25 gilts were vaccinated at late gestation (100th day) and were divided into five groups according to the different vaccines (Vac) used: no vaccine-control group, Vac-1-strain DV, Vac-2-strain VP-046 BIS, Vac-3-strain 94881, Vac-4-strain 96V198. Within the first 0-9 h of the farrowing, blood samples were collected from all newborn piglets and lung samples were exanimated grossly, histopathologically and with scanning electron microscopy. PRRSV (RT-PCR-positive) and antibodies were detected in the serum of piglets from gilts vaccinated with Vac-2. In these piglets, moderate to severe interstitial pneumonia with thickened alveolar septa was noticed. Type II pneumocyte hyperplasia was also observed. The rest of the trial piglets showed unremarkable lung lesions. Phylogenetic analysis revealed the 98.7% similarity of the PRRSV field strain (GR 2019-1) to the PRRS MLV vaccine strain VP-046 BIS. In conclusion, the Vac-2 PRRSV vaccine strain can act as an infectious strain when vaccination is administrated at late gestation, causing lung lesions.
Collapse
Affiliation(s)
- Georgios I. Papakonstantinou
- Clinic of Medicine, Faculty of Veterinary Medicine, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (G.I.P.); (L.V.A.); (G.M.)
| | - Dimitra Psalla
- Laboratory of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.P.); (I.S.); (N.P.)
| | - Aris Pourlis
- Laboratory of Anatomy, Histology & Embryology, Veterinary School, University of Thessaly, 43100 Karditsa, Greece;
| | - Ioanna Stylianaki
- Laboratory of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.P.); (I.S.); (N.P.)
| | - Labrini V. Athanasiou
- Clinic of Medicine, Faculty of Veterinary Medicine, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (G.I.P.); (L.V.A.); (G.M.)
| | - Eleni Tzika
- Farm Animals Clinic, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54627 Thessaloniki, Greece;
| | - Eleftherios Meletis
- Laboratory of Epidemiology & Artificial Intelligence, Faculty of Public Health, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (E.M.); (P.K.)
| | - Polychronis Kostoulas
- Laboratory of Epidemiology & Artificial Intelligence, Faculty of Public Health, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (E.M.); (P.K.)
| | - George Maragkakis
- Clinic of Medicine, Faculty of Veterinary Medicine, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (G.I.P.); (L.V.A.); (G.M.)
| | - Georgios Christodoulopoulos
- Department of Animal Science, Agricultural University of Athens, 75 Iera Odos Street, Votanikos, 11855 Athens, Greece;
| | - Nikolaos Papaioannou
- Laboratory of Pathology, Faculty of Health Sciences, School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.P.); (I.S.); (N.P.)
| | - Vasileios G. Papatsiros
- Clinic of Medicine, Faculty of Veterinary Medicine, School of Health Sciences, University of Thessaly, 43100 Karditsa, Greece; (G.I.P.); (L.V.A.); (G.M.)
| |
Collapse
|
34
|
Shields PG, Ying KL, Brasky TM, Freudenheim JL, Li Z, McElroy JP, Reisinger SA, Song MA, Weng DY, Wewers MD, Whiteman NB, Yang Y, Mathé EA. A Pilot Cross-Sectional Study of Immunological and Microbiome Profiling Reveals Distinct Inflammatory Profiles for Smokers, Electronic Cigarette Users, and Never-Smokers. Microorganisms 2023; 11:1405. [PMID: 37374908 PMCID: PMC10303504 DOI: 10.3390/microorganisms11061405] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/11/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Smokers (SM) have increased lung immune cell counts and inflammatory gene expression compared to electronic cigarette (EC) users and never-smokers (NS). The objective of this study is to further assess associations for SM and EC lung microbiomes with immune cell subtypes and inflammatory gene expression in samples obtained by bronchoscopy and bronchoalveolar lavage (n = 28). RNASeq with the CIBERSORT computational algorithm were used to determine immune cell subtypes, along with inflammatory gene expression and microbiome metatranscriptomics. Macrophage subtypes revealed a two-fold increase in M0 (undifferentiated) macrophages for SM and EC users relative to NS, with a concordant decrease in M2 (anti-inflammatory) macrophages. There were 68, 19, and 1 significantly differentially expressed inflammatory genes (DEG) between SM/NS, SM/EC users, and EC users/NS, respectively. CSF-1 and GATA3 expression correlated positively and inversely with M0 and M2 macrophages, respectively. Correlation profiling for DEG showed distinct lung profiles for each participant group. There were three bacteria genera-DEG correlations and three bacteria genera-macrophage subtype correlations. In this pilot study, SM and EC use were associated with an increase in undifferentiated M0 macrophages, but SM differed from EC users and NS for inflammatory gene expression. The data support the hypothesis that SM and EC have toxic lung effects influencing inflammatory responses, but this may not be via changes in the microbiome.
Collapse
Affiliation(s)
- Peter G. Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Kevin L. Ying
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Theodore M. Brasky
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Jo L. Freudenheim
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, NY 14261, USA
| | - Zihai Li
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Joseph P. McElroy
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sarah A. Reisinger
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Y. Weng
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Mark D. Wewers
- Pulmonary and Critical Care Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Noah B. Whiteman
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Yiping Yang
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
| | - Ewy A. Mathé
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, OH 43210, USA; (K.L.Y.)
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institute of Health, Rockville, MD 20892, USA
| |
Collapse
|
35
|
Kotlyarov S. The Role of Smoking in the Mechanisms of Development of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2023; 24:8725. [PMID: 37240069 PMCID: PMC10217854 DOI: 10.3390/ijms24108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Tobacco smoking is a major cause of chronic obstructive pulmonary disease (COPD) and atherosclerotic cardiovascular disease (ASCVD). These diseases share common pathogenesis and significantly influence each other's clinical presentation and prognosis. There is increasing evidence that the mechanisms underlying the comorbidity of COPD and ASCVD are complex and multifactorial. Smoking-induced systemic inflammation, impaired endothelial function and oxidative stress may contribute to the development and progression of both diseases. The components present in tobacco smoke can have adverse effects on various cellular functions, including macrophages and endothelial cells. Smoking may also affect the innate immune system, impair apoptosis, and promote oxidative stress in the respiratory and vascular systems. The purpose of this review is to discuss the importance of smoking in the mechanisms underlying the comorbid course of COPD and ASCVD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
36
|
Shirata M, Ito I, Jo T, Iwao T, Oi I, Hamao N, Nishioka K, Yamana H, Nagase T, Yasunaga H, Hirai T. Factors Associated With the Development of Bacterial Pneumonia Related to Seasonal Influenza Virus Infection: A Study Using a Large-scale Health Insurance Claim Database. Open Forum Infect Dis 2023; 10:ofad222. [PMID: 37234515 PMCID: PMC10205552 DOI: 10.1093/ofid/ofad222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Background Influenza-related bacterial pneumonia is a leading complication of influenza infection. However, the differences in the incidence rates and risk factors associated with concomitant viral/bacterial pneumonia (CP) and secondary bacterial pneumonia following influenza (SP) remain unclear. This study aimed to clarify the incidence rates of CP and SP following seasonal influenza and identify factors associated with their development. Methods This retrospective cohort study was conducted using the JMDC Claims Database, a health insurance claims database in Japan. All patients aged <75 years who developed influenza during 2 consecutive epidemic seasons, 2017/2018 and 2018/2019, were analyzed. CP was defined as bacterial pneumonia diagnosed between 3 days before and 6 days after the date of influenza diagnosis, and SP was defined as pneumonia diagnosed 7-30 days after the date of diagnosis. Multivariable logistic regression analyses were performed to identify factors associated with the development of CP and SP. Results Among the 10 473 014 individuals registered in the database, 1 341 355 patients with influenza were analyzed. The average age at diagnosis (SD) was 26.6 (18.6) years. There were 2901 (0.22%) and 1262 (0.09%) patients who developed CP and SP, respectively. Age 65-74 years, asthma, chronic bronchitis/emphysema, cardiovascular disease, renal disease, malignant tumor, and immunosuppression were significant risk factors for both CP and SP, whereas cerebrovascular disease, neurological disease, liver disease, and diabetes were risk factors specific to CP development. Conclusions The results determined the incidence rates of CP and SP and identified their risk factors, such as older age and comorbidities.
Collapse
Affiliation(s)
- Masahiro Shirata
- Correspondence: Isao Ito, MD, PhD, Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto 606-8507, Japan (); or Masahiro Shirata, MD, Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto 606-8507, Japan ()
| | - Isao Ito
- Correspondence: Isao Ito, MD, PhD, Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto 606-8507, Japan (); or Masahiro Shirata, MD, Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo, Kyoto 606-8507, Japan ()
| | - Taisuke Jo
- Department of Health Services Research, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohide Iwao
- Institute for Advancement of Clinical and Translational Science, Kyoto University Hospital, Kyoto, Japan
| | - Issei Oi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuyoshi Hamao
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kensuke Nishioka
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hayato Yamana
- Department of Health Services Research, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, Tokyo, Japan
| | | |
Collapse
|
37
|
Wu X, Li RF, Lin ZS, Xiao C, Liu B, Mai KL, Zhou HX, Zeng DY, Cheng S, Weng YC, Zhao J, Chen RF, Jiang HM, Chen LP, Deng LZ, Xie PF, Yang WM, Xia XS, Yang ZF. Coinfection with influenza virus and non-typeable Haemophilus influenzae aggregates inflammatory lung injury and alters gut microbiota in COPD mice. Front Microbiol 2023; 14:1137369. [PMID: 37065141 PMCID: PMC10098174 DOI: 10.3389/fmicb.2023.1137369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023] Open
Abstract
BackgroundAcute exacerbation of chronic obstructive pulmonary disease (AECOPD) is associated with high mortality rates. Viral and bacterial coinfection is the primary cause of AECOPD. How coinfection with these microbes influences host inflammatory response and the gut microbiota composition is not entirely understood.MethodsWe developed a mouse model of AECOPD by cigarette smoke exposure and sequential infection with influenza H1N1 virus and non-typeable Haemophilus influenzae (NTHi). Viral and bacterial titer was determined using MDCK cells and chocolate agar plates, respectively. The levels of cytokines, adhesion molecules, and inflammatory cells in the lungs were measured using Bio-Plex and flow cytometry assays. Gut microbiota was analyzed using 16S rRNA gene sequencing. Correlations between cytokines and gut microbiota were determined using Spearman’s rank correlation coefficient test.ResultsCoinfection with H1N1 and NTHi resulted in more severe lung injury, higher mortality, declined lung function in COPD mice. H1N1 enhanced NTHi growth in the lungs, but NTHi had no effect on H1N1. In addition, coinfection increased the levels of cytokines and adhesion molecules, as well as immune cells including total and M1 macrophages, neutrophils, monocytes, NK cells, and CD4 + T cells. In contrast, alveolar macrophages were depleted. Furthermore, coinfection caused a decline in the diversity of gut bacteria. Muribaculaceae, Lactobacillus, Akkermansia, Lachnospiraceae, and Rikenella were further found to be negatively correlated with cytokine levels, whereas Bacteroides was positively correlated.ConclusionCoinfection with H1N1 and NTHi causes a deterioration in COPD mice due to increased lung inflammation, which is correlated with dysbiosis of the gut microbiota.
Collapse
Affiliation(s)
- Xiao Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Run-Feng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
| | - Zheng-Shi Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Bin Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kai-Lin Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | | | - De-You Zeng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Sha Cheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Yun-Ceng Weng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rui-Feng Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hai-Ming Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li-Ping Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling-Zhu Deng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pei-Fang Xie
- The Affiliated Anning First Hospital and Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Wei-Min Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
- Wei-Min Yang,
| | - Xue-Shan Xia
- The Affiliated Anning First Hospital and Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- Xue-Shan Xia,
| | - Zi-Feng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Laboratory, Guangzhou, China
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, Guangzhou, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
- *Correspondence: Zi-Feng Yang,
| |
Collapse
|
38
|
Sun Z, Lin J, Zhang T, Sun X, Wang T, Duan J, Yao K. Combining bioinformatics and machine learning to identify common mechanisms and biomarkers of chronic obstructive pulmonary disease and atrial fibrillation. Front Cardiovasc Med 2023; 10:1121102. [PMID: 37057099 PMCID: PMC10086368 DOI: 10.3389/fcvm.2023.1121102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundPatients with chronic obstructive pulmonary disease (COPD) often present with atrial fibrillation (AF), but the common pathophysiological mechanisms between the two are unclear. This study aimed to investigate the common biological mechanisms of COPD and AF and to search for important biomarkers through bioinformatic analysis of public RNA sequencing databases.MethodsFour datasets of COPD and AF were downloaded from the Gene Expression Omnibus (GEO) database. The overlapping genes common to both diseases were screened by WGCNA analysis, followed by protein-protein interaction network construction and functional enrichment analysis to elucidate the common mechanisms of COPD and AF. Machine learning algorithms were also used to identify key biomarkers. Co-expression analysis, “transcription factor (TF)-mRNA-microRNA (miRNA)” regulatory networks and drug prediction were performed for key biomarkers. Finally, immune cell infiltration analysis was performed to evaluate further the immune cell changes in the COPD dataset and the correlation between key biomarkers and immune cells.ResultsA total of 133 overlapping genes for COPD and AF were obtained, and the enrichment was mainly focused on pathways associated with the inflammatory immune response. A key biomarker, cyclin dependent kinase 8 (CDK8), was identified through screening by machine learning algorithms and validated in the validation dataset. Twenty potential drugs capable of targeting CDK8 were obtained. Immune cell infiltration analysis revealed the presence of multiple immune cell dysregulation in COPD. Correlation analysis showed that CDK8 expression was significantly associated with CD8+ T cells, resting dendritic cell, macrophage M2, and monocytes.ConclusionsThis study highlights the role of the inflammatory immune response in COPD combined with AF. The prominent link between CDK8 and the inflammatory immune response and its characteristic of not affecting the basal expression level of nuclear factor kappa B (NF-kB) make it a possible promising therapeutic target for COPD combined with AF.
Collapse
Affiliation(s)
- Ziyi Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jianguo Lin
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tianya Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiaoning Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tianlin Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jinlong Duan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kuiwu Yao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Eye Hospital China Academy of Chinese Medical Sciences, China Academy of Chinese Medical Sciences, Beijing, China
- Correspondence: Kuiwu Yao
| |
Collapse
|
39
|
Ohashi K, Hayashida A, Nozawa A, Matsumura K, Ito S. Human vasculature-on-a-chip with macrophage-mediated endothelial activation: The biological effect of aerosol from heated tobacco products on monocyte adhesion. Toxicol In Vitro 2023; 89:105582. [PMID: 36933580 DOI: 10.1016/j.tiv.2023.105582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Heated tobacco products (HTPs) are expected to have the potential to reduce risks of smoking-associated cardiovascular disease (CVD). However, mechanism-based investigations of the effect of HTPs on atherosclerosis remain insufficient and further studies under human-relevant situations are desired for deeper understanding of the reduced risk potential of HTPs. In this study, we first developed an in vitro model of monocyte adhesion by considering macrophage-derived proinflammatory cytokine-mediated endothelial activation using an organ-on-a-chip (OoC), which provided great opportunities to mimic major aspects of human physiology. Then biological activities of aerosol from three different types of HTPs in terms of monocyte adhesion were compared with that of cigarette smoke (CS). Our model showed that the effective concentration ranges of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were close to the actual condition in CVD pathogenesis. The model also showed that monocyte adhesion was less induced by each HTP aerosol than CS, which may be caused by less proinflammatory cytokine secretion. In summary, our vasculature-on-a-chip model assessed the difference in biological effects between cigarettes and HTPs, and suggested a reduced risk potential of HTPs for atherosclerosis.
Collapse
Affiliation(s)
- Kazuhiro Ohashi
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Ayaka Hayashida
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Atsuko Nozawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Kazushi Matsumura
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| | - Shigeaki Ito
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama 227-8512, Kanagawa, Japan.
| |
Collapse
|
40
|
Biological and Genetic Mechanisms of COPD, Its Diagnosis, Treatment, and Relationship with Lung Cancer. Biomedicines 2023; 11:biomedicines11020448. [PMID: 36830984 PMCID: PMC9953173 DOI: 10.3390/biomedicines11020448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most prevalent chronic adult diseases, with significant worldwide morbidity and mortality. Although long-term tobacco smoking is a critical risk factor for this global health problem, its molecular mechanisms remain unclear. Several phenomena are thought to be involved in the evolution of emphysema, including airway inflammation, proteinase/anti-proteinase imbalance, oxidative stress, and genetic/epigenetic modifications. Furthermore, COPD is one main risk for lung cancer (LC), the deadliest form of human tumor; formation and chronic inflammation accompanying COPD can be a potential driver of malignancy maturation (0.8-1.7% of COPD cases develop cancer/per year). Recently, the development of more research based on COPD and lung cancer molecular analysis has provided new light for understanding their pathogenesis, improving the diagnosis and treatments, and elucidating many connections between these diseases. Our review emphasizes the biological factors involved in COPD and lung cancer, the advances in their molecular mechanisms' research, and the state of the art of diagnosis and treatments. This work combines many biological and genetic elements into a single whole and strongly links COPD with lung tumor features.
Collapse
|
41
|
Kim JH, Kim JW, Kim CY, Jeong JS, Ko JW, Kim TW. Green tea extract ameliorates macrophage-driven emphysematous lesions in chronic obstructive pulmonary disease induced by cigarette smoke condensate. Phytother Res 2023; 37:1366-1376. [PMID: 36729048 DOI: 10.1002/ptr.7745] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/14/2022] [Accepted: 01/21/2023] [Indexed: 02/03/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is an important lung disease characterized by complicated symptoms including emphysema. We aimed to explore the mechanisms underlying the protective effect of green tea extract (GTE) on cigarette smoke condensate (CSC)-induced emphysema by demonstrating the reduction of macrophage-induced protease expression through GTE treatment in vivo and in vitro. Mice were intranasally administered 50 mg/kg CSC once a week for 4 weeks, and doses of 100 or 300 mg/kg GTE were administered orally once daily for 4 weeks. GTE significantly reduced macrophage counts in bronchoalveolar lavage fluid and emphysematous lesions in lung tissues in CSC-exposed mice. In addition, GTE suppressed CSC-induced extracellular signal-regulated kinase (ERK)/activator protein (AP)-1 phosphorylation followed by matrix metalloproteinases (MMP)-9 expression as revealed by western blotting, immunohistochemistry, and zymography in CSC-instilled mice. These underlying mechanisms related to reduced protease expression were confirmed in NCI-H292 cells stimulated by CSC. Taken together, GTE effectively inhibits macrophage-driven emphysematous lesions induced by CSC treatment, and these protective effects of GTE are closely related to the ERK/AP-1 signaling pathway, followed by a reduced protease/antiprotease imbalance. These results suggest that GTE can be used as a supplementary agent for the prevention of emphysema progression in COPD patients.
Collapse
Affiliation(s)
- Jin-Hwa Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Chang-Yeop Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Ji-Soo Jeong
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Je-Won Ko
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Tae-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
42
|
Identification of TRPC6 as a Novel Diagnostic Biomarker of PM-Induced Chronic Obstructive Pulmonary Disease Using Machine Learning Models. Genes (Basel) 2023; 14:genes14020284. [PMID: 36833211 PMCID: PMC9957274 DOI: 10.3390/genes14020284] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) was the third most prevalent cause of mortality worldwide in 2010; it results from a progressive and fatal deterioration of lung function because of cigarette smoking and particulate matter (PM). Therefore, it is important to identify molecular biomarkers that can diagnose the COPD phenotype to plan therapeutic efficacy. To identify potential novel biomarkers of COPD, we first obtained COPD and the normal lung tissue gene expression dataset GSE151052 from the NCBI Gene Expression Omnibus (GEO). A total of 250 differentially expressed genes (DEGs) were investigated and analyzed using GEO2R, gene ontology (GO) functional annotation, and Kyoto Encyclopedia of Genes and Genomes (KEGG) identification. The GEO2R analysis revealed that TRPC6 was the sixth most highly expressed gene in patients with COPD. The GO analysis indicated that the upregulated DEGs were mainly concentrated in the plasma membrane, transcription, and DNA binding. The KEGG pathway analysis indicated that the upregulated DEGs were mainly involved in pathways related to cancer and axon guidance. TRPC6, one of the most abundant genes among the top 10 differentially expressed total RNAs (fold change ≥ 1.5) between the COPD and normal groups, was selected as a novel COPD biomarker based on the results of the GEO dataset and analysis using machine learning models. The upregulation of TRPC6 was verified in PM-stimulated RAW264.7 cells, which mimicked COPD conditions, compared to untreated RAW264.7 cells by a quantitative reverse transcription polymerase chain reaction. In conclusion, our study suggests that TRPC6 can be regarded as a potential novel biomarker for COPD pathogenesis.
Collapse
|
43
|
Zhang J, Cheng H, Xue M, Xiong Y, Zhu Y, Björkegren JLM, Zhang Z, Chen J, Shi Z, Hao K. Effects of chronic electronic cigarettes exposure in inducing respiratory function decline and pulmonary tissue injury - A direct comparison to combustible cigarettes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114426. [PMID: 36525947 DOI: 10.1016/j.ecoenv.2022.114426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Electronic cigarette (e-cig) use is increasing worldwide, especially among young individuals. Spirometry measures airflow obstruction and is the primary tool for diagnosing/monitoring respiratory diseases in clinical settings. This study aims to assess the effects of chronic e-cig exposure on spirometric traits, and directly compare to conventional combustible-cigarette (c-cig). METHODS We employed an e- and c-cig aerosol generation system that resembled human smoking/vaping scenario. Fifty 6-week old C57BL/6 mice were equally divided into five groups and exposed to clean air (control), e-cig aerosol (low- and high-dose), and c-cig aerosol (low- and high-dose), respectively, for 10 weeks. Afterwards, growth trajectory, spirometry and pulmonary pathology were analyzed. RESULTS Both e- and c-cig exposure slowed down growth and weight gain. Low dose e-cig exposure (1 h exposure per day) resulted in minimal respiratory function damage. At high dose (2 h exposure per day), e-cig exposure deteriorated 7 spirometry traits but by a smaller magnitude than c-cig exposure. For example, comparing to clean air controls, high dose e- and c-cig exposure increased inspiratory resistance by 24.3% (p = 0.026) and 66.7% (p = 2.6e-5), respectively. Low-dose e-cig exposure increased alveolar macrophage count but did not lead to airway remodeling. In contrast, even low-dose c-cig caused alveoli break down and thickening of the small airway, hallmarks of airway obstructive disease. CONCLUSIONS We conducted well-controlled animal exposure experiments assessing chronic e-cig exposure's effects on spirometry traits. Further, mechanistic study characterized airway remodeling, alveolar tissue lesion and inflammation induced by e- and c-cig exposure. Our findings provided scientific and public health insights on e-cig's health consequences, especially in adolescent users.
Collapse
Affiliation(s)
- Jushan Zhang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China; College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mo Xue
- Smoore Research Institute, Smoore International, Shenzhen, China
| | - Yuming Xiong
- Smoore Research Institute, Smoore International, Shenzhen, China
| | - Yujie Zhu
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Johan L M Björkegren
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Zhongyang Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhiqiang Shi
- Smoore Research Institute, Smoore International, Shenzhen, China
| | - Ke Hao
- College of Environmental Science and Engineering, Tongji University, Shanghai, China; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
44
|
Polverino F, Mirra D, Yang CX, Esposito R, Spaziano G, Rojas-Quintero J, Sgambato M, Piegari E, Cozzolino A, Cione E, Gallelli L, Capuozzo A, Santoriello C, Berrino L, de- Torres JP, Hackett TL, Polverino M, D’Agostino B. Similar programmed death ligand 1 (PD-L1) expression profile in patients with mild COPD and lung cancer. Sci Rep 2022; 12:22402. [PMID: 36575294 PMCID: PMC9792927 DOI: 10.1038/s41598-022-26650-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Programmed Death Ligand 1 (PD-L1) is crucial in regulating the immunological tolerance in non-small cell lung cancer (NSCLC). Alveolar macrophage (AM)-derived PD-L1 binds to its receptor, PD-1, on surveilling lymphocytes, leading to lymphocyte exhaustion. Increased PD-L1 expression is associated with cigarette smoke (CS)-exposure. However, the PD-L1 role in CS-associated lung diseases associated with NSCLC, such as chronic obstructive pulmonary disease (COPD), is still unclear. In two different cohorts of ever smokers with COPD or NSCLC, and ever and never smoker controls, we evaluated PD-L1 expression: (1) via cutting-edge digital spatial proteomic and transcriptomic profiling (Geomx) of formalin-fixed paraffin-embedded (FFPE) lung tissue sections (n = 19); and (2) via triple immunofluorescence staining of bronchoalveolar lavage (BAL) AMs (n = 83). PD-L1 mRNA expression was also quantified in BAL AMs exposed to CS extract. PD-L1 expression was increased in the bronchiolar wall, parenchyma, and vascular wall from mild-moderate (GOLD 1-2) COPD patients compared to severe-very severe (GOLD 3-4) COPD patients and controls. Within all the COPD patients, PD-L1 protein expression was associated with upregulation of genes involved in tumor progression and downregulation of oncosuppressive genes, and strongly directly correlated with the FEV1% predicted, indicating higher PD-L1 expression in the milder vs. more severe COPD stages. In bronchioles, PD-L1 levels were strongly directly correlated with the number of functionally active AMs. In BAL, we confirmed that AMs from patients with both GOLD 1-2 COPD and NSCLC had the highest and similar, PD-L1 expression levels versus all the other groups, independently from active cigarette smoking. Intriguingly, AMs from patients with more severe COPD had reduced AM PD-L1 expression compared to patients with mild COPD. Acute CS extract stimulation increased PD-L1 mRNA expression only in never-and not in ever-smoker AMs. Lungs from patients with mild COPD and NSCLC are characterized by a similar strong PD-L1 expression signature in bronchioles and functionally active AMs compared to patients with severe COPD and controls. Active smoking does not affect PD-L1 levels. These observations represent a new resource in understanding the innate immune mechanisms underlying the link between COPD and lung cancer onset and progression and pave the way to future studies focused on the mechanisms by which CS promotes tumorigenesis and COPD.
Collapse
Affiliation(s)
- F. Polverino
- grid.39382.330000 0001 2160 926XPulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77030 USA
| | - D. Mirra
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - C. X. Yang
- grid.17091.3e0000 0001 2288 9830University of British Columbia, Vancouver, Canada
| | - R. Esposito
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - G. Spaziano
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - J. Rojas-Quintero
- grid.39382.330000 0001 2160 926XPulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX 77030 USA
| | - M. Sgambato
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - E. Piegari
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - A. Cozzolino
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - E. Cione
- grid.7778.f0000 0004 1937 0319University of Calabria, Rende, Italy
| | - L. Gallelli
- grid.411489.10000 0001 2168 2547University of Catanzaro, Catanzaro, Italy
| | | | | | - L. Berrino
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - J. P. de- Torres
- grid.410356.50000 0004 1936 8331Queen’s University, Hamilton, Canada
| | - T. L. Hackett
- grid.17091.3e0000 0001 2288 9830University of British Columbia, Vancouver, Canada
| | | | - B. D’Agostino
- grid.9841.40000 0001 2200 8888University of Campania “Luigi Vanvitelli”, Caserta, Italy
| |
Collapse
|
45
|
Japiassu KB, Fay F, Marengo A, Louaguenouni Y, Cailleau C, Denis S, Chapron D, Tsapis N, Nascimento TL, Lima EM, Fattal E. Interplay between mucus mobility and alveolar macrophage targeting of surface-modified liposomes. J Control Release 2022; 352:15-24. [PMID: 36209941 DOI: 10.1016/j.jconrel.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
Alveolar macrophages play a crucial role in the initiation and resolution of the immune response in the lungs. Pro-inflammatory M1 alveolar macrophages are an interesting target for treating inflammatory and infectious pulmonary diseases. One commune targeting strategy is to use nanoparticles conjugated with hyaluronic acid, which interact with CD44 overexpressed on the membrane of those cells. Unfortunately, this coating strategy may be countered by the presence on the surface of the nanoparticles of a poly(ethylene glycol) corona employed to improve nanoparticles' diffusion in the lung mucus. This study aims to measure this phenomenon by comparing the behavior in a murine lung inflammation model of three liposomal platforms designed to represent different poly(ethylene glycol) and hyaluronic acid densities (Liposome-PEG, Liposome-PEG-HA and Liposome-HA). In this work, the liposomes were obtained by a one-step ethanol injection method. Their interaction with mucin and targeting ability toward pro-inflammatory macrophages were then investigated in vitro and in vivo in a LPS model of lung inflammation. In vitro, poly(ethylene glycol) free HA-liposomes display a superior targeting efficiency toward M1 macrophages, while the addition of poly(ethylene glycol) induces better mucus mobility. Interestingly in vivo studies revealed that the three liposomes showed distinct cell specificity with alveolar macrophages demonstrating an avidity for poly(ethylene glycol) free HA-liposomes, while neutrophils favored PEGylated liposomes exempt of HA. Those results could be explained by the presence of two forces exercising a balance between mucus penetration and receptor targeting. This study corroborates the importance of considering the site of action and the targeted cells when designing nanoparticles to treat lung diseases.
Collapse
Affiliation(s)
- Kamila Bohne Japiassu
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France; Center for RD&I in Pharmaceutical Nano/Technology (FarmaTec), Federal University of Goias, Goiania, 74605-220, Goias, Brazil
| | - Francois Fay
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Alessandro Marengo
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Younès Louaguenouni
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Catherine Cailleau
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Stéphanie Denis
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - David Chapron
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Nicolas Tsapis
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France
| | - Thais Leite Nascimento
- Center for RD&I in Pharmaceutical Nano/Technology (FarmaTec), Federal University of Goias, Goiania, 74605-220, Goias, Brazil
| | - Eliana Martins Lima
- Center for RD&I in Pharmaceutical Nano/Technology (FarmaTec), Federal University of Goias, Goiania, 74605-220, Goias, Brazil
| | - Elias Fattal
- University Paris-Saclay, CNRS, Institut Galien Paris-Saclay (UMR 8612), Châtenay-Malabry, France.
| |
Collapse
|
46
|
Tejwani V, Woo H, Liu C, Tillery AK, Gassett AJ, Kanner RE, Hoffman EA, Martinez FJ, Woodruff PG, Barr RG, Fawzy A, Koehler K, Curtis JL, Freeman CM, Cooper CB, Comellas AP, Pirozzi C, Paine R, Tashkin D, Krishnan JA, Sack C, Putcha N, Paulin LM, Zusman M, Kaufman JD, Alexis NE, Hansel NN. Black carbon content in airway macrophages is associated with increased severe exacerbations and worse COPD morbidity in SPIROMICS. Respir Res 2022; 23:310. [PMID: 36376879 PMCID: PMC9664618 DOI: 10.1186/s12931-022-02225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Airway macrophages (AM), crucial for the immune response in chronic obstructive pulmonary disease (COPD), are exposed to environmental particulate matter (PM), which they retain in their cytoplasm as black carbon (BC). However, whether AM BC accurately reflects environmental PM2.5 exposure, and can serve as a biomarker of COPD outcomes, is unknown. METHODS We analyzed induced sputum from participants at 7 of 12 sites SPIROMICS sites for AM BC content, which we related to exposures and to lung function and respiratory outcomes. Models were adjusted for batch (first vs. second), age, race (white vs. non-white), income (<$35,000, $35,000~$74,999, ≥$75,000, decline to answer), BMI, and use of long-acting beta-agonist/long-acting muscarinic antagonists, with sensitivity analysis performed with inclusion of urinary cotinine and lung function as covariates. RESULTS Of 324 participants, 143 were current smokers and 201 had spirometric-confirmed COPD. Modeled indoor fine (< 2.5 μm in aerodynamic diameter) particulate matter (PM2.5) and urinary cotinine were associated with higher AM BC. Other assessed indoor and ambient pollutant exposures were not associated with higher AM BC. Higher AM BC was associated with worse lung function and odds of severe exacerbation, as well as worse functional status, respiratory symptoms and quality of life. CONCLUSION Indoor PM2.5 and cigarette smoke exposure may lead to increased AM BC deposition. Black carbon content in AMs is associated with worse COPD morbidity in current and former smokers, which remained after sensitivity analysis adjusting for cigarette smoke burden. Airway macrophage BC, which may alter macrophage function, could serve as a predictor of experiencing worse respiratory symptoms and impaired lung function.
Collapse
Affiliation(s)
- Vickram Tejwani
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, A90, 44195, Cleveland, OH, USA.
| | - Han Woo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chen Liu
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Anna K Tillery
- Center for Environmental Medicine, Asthma, and Lung Biology, Division of Allergy and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda J Gassett
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Richard E Kanner
- Division of Respiratory, Critical Care and Occupational Medicine, University of Utah, Salt Lake City, UT, USA
| | - Eric A Hoffman
- Department of Radiology, Medicine and Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Fernando J Martinez
- Division of Pulmonology and Critical Care Medicine, Weill-Cornell Medical Center, Cornell University, New York, NY, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - R Graham Barr
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Ashraf Fawzy
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kirsten Koehler
- Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey L Curtis
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Christine M Freeman
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Christopher B Cooper
- Division of Pulmonary and Critical Care Medicine, University of California Los Angeles Medical Center, Los Angeles, CA, USA
| | - Alejandro P Comellas
- Division of Pulmonary, Critical Care, and Occupational Medicine, College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Robert Paine
- University of Utah Hospital, Salt Lake City, UT, USA
| | - Donald Tashkin
- Division of Pulmonary and Critical Care Medicine, University of California Los Angeles Medical Center, Los Angeles, CA, USA
| | - Jerry A Krishnan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Coralynn Sack
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Laura M Paulin
- Pulmonary/Critical Care, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Marina Zusman
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Joel D Kaufman
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA, USA
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, Division of Allergy and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
47
|
Rajabi H, Mortazavi D, Konyalilar N, Aksoy GT, Erkan S, Korkunc SK, Kayalar O, Bayram H, Rahbarghazi R. Forthcoming complications in recovered COVID-19 patients with COPD and asthma; possible therapeutic opportunities. Cell Commun Signal 2022; 20:173. [PMID: 36320055 PMCID: PMC9623941 DOI: 10.1186/s12964-022-00982-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/01/2022] [Indexed: 11/21/2022] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been growing swiftly worldwide. Patients with background chronic pulmonary inflammations such as asthma or chronic obstructive pulmonary diseases (COPD) are likely to be infected with this virus. Of note, there is an argument that COVID-19 can remain with serious complications like fibrosis or other pathological changes in the pulmonary tissue of patients with chronic diseases. Along with conventional medications, regenerative medicine, and cell-based therapy could be alternative approaches to compensate for organ loss or restore injured sites using different stem cell types. Owing to unique differentiation capacity and paracrine activity, these cells can accelerate the healing procedure. In this review article, we have tried to scrutinize different reports related to the harmful effects of SARS-CoV-2 on patients with asthma and COPD, as well as the possible therapeutic effects of stem cells in the alleviation of post-COVID-19 complications. Video abstract.
Collapse
Affiliation(s)
- Hadi Rajabi
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Deniz Mortazavi
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Nur Konyalilar
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Gizem Tuse Aksoy
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Sinem Erkan
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Seval Kubra Korkunc
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Ozgecan Kayalar
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Hasan Bayram
- Koç University Research Centre for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey.
- Department of Pulmonary Medicine, School of Medicine, Koç University, Istanbul, Turkey.
| | - Reza Rahbarghazi
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
48
|
Li Q, Cheng Y, Zhang Z, Bi Z, Ma X, Wei Y, Wei X. Inhibition of ROCK ameliorates pulmonary fibrosis by suppressing M2 macrophage polarisation through phosphorylation of STAT3. Clin Transl Med 2022; 12:e1036. [PMID: 36178087 PMCID: PMC9523675 DOI: 10.1002/ctm2.1036] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Emerging evidence provides mechanistic insights into the pathogenesis of pulmonary fibrosis (PF), and rare anti-PF therapeutic method has promising effect in its treatment. Rho-associated coiled-coil kinases (ROCK) inhibition significantly ameliorates bleomycin-induced PF and decreases macrophage infiltration, but the mechanism remains unclear. We established bleomycin and radiation-induced PF to identify the activity of WXWH0265, a newly designed unselective ROCK inhibitor in regulating macrophages. METHODS Bleomycin-induced PF was induced by intratracheal instillation and radiation-induced PF was induced by bilateral thoracic irradiation. Histopathological techniques (haematoxylin and eosin, Masson's trichrome and immunohistochemistry) and hydroxyproline were used to evaluate PF severity. Western blot, quantitative real-time reverse transcription-polymerase chain reaction and flow cytometry were performed to explore the underlying mechanisms. Bone marrow-derived macrophages (BMDMs) were used to verify their therapeutic effect. Clodronate liposomes were applied to deplete macrophages and to identify the therapeutic effect of WXWH0265. RESULTS Therapeutic administration of ROCK inhibitor ameliorates bleomycin-induced PF by inhibiting M2 macrophages polarisation. ROCK inhibitor showed no significant anti-fibrotic effect in macrophages-depleted mice. Treatment with WXWH0265 demonstrated superior protection effect in bleomycin-induced PF compared with positive drugs. In radiation-induced PF, ROCK inhibitor effectively ameliorated PF. Fibroblasts co-cultured with supernatant from various M2 macrophages phenotypes revealed that M2 macrophages stimulated by interleukin-4 promoted extracellular matrix production. Polarisation of M2 macrophages was inhibited by ROCK inhibitor treatment in vitro. The p-signal transducer and activator of transcription 3 (STAT3) in lung tissue and BMDMs was significantly decreased in PF in vivo and vitro after treated with ROCK inhibitors. CONCLUSION Inhibiting ROCK could significantly attenuate bleomycin- and radiation-induced PF by regulating the macrophages polarisation via phosphorylation of STAT3. WXWH0265 is a kind of efficient unselective ROCK inhibitor in ameliorating PF. Furthermore, the results provide empirical evidence that ROCK inhibitor, WXWH0265 is a potential drug to prevent the development of PF.
Collapse
Affiliation(s)
- Qingfang Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Yuan Cheng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Zhe Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Xuelei Ma
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
| |
Collapse
|
49
|
Marzec JM, Nadadur SS. Inflammation resolution in environmental pulmonary health and morbidity. Toxicol Appl Pharmacol 2022; 449:116070. [PMID: 35618031 PMCID: PMC9872158 DOI: 10.1016/j.taap.2022.116070] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/04/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023]
Abstract
Inflammation and resolution are dynamic processes comprised of inflammatory activation and neutrophil influx, followed by mediator catabolism and efferocytosis. These critical pathways ensure a return to homeostasis and promote repair. Over the past decade research has shown that diverse mediators play a role in the active process of resolution. Specialized pro-resolving mediators (SPMs), biosynthesized from fatty acids, are released during inflammation to facilitate resolution and are deficient in a variety of lung disorders. Failed resolution results in remodeling and cellular deposition through pro-fibrotic myofibroblast expansion that irreversibly narrows the airways and worsens lung function. Recent studies indicate environmental exposures may perturb and deregulate critical resolution pathways. Environmental xenobiotics induce lung inflammation and generate reactive metabolites that promote oxidative stress, injuring the respiratory mucosa and impairing gas-exchange. This warrants recognition of xenobiotic associated molecular patterns (XAMPs) as new signals in the field of inflammation biology, as many environmental chemicals generate free radicals capable of initiating the inflammatory response. Recent studies suggest that unresolved, persistent inflammation impacts both resolution pathways and endogenous regulatory mediators, compromising lung function, which over time can progress to chronic lung disease. Chronic ozone (O3) exposure overwhelms successful resolution, and in susceptible individuals promotes asthma onset. The industrial contaminant cadmium (Cd) bioaccumulates in the lung to impair resolution, and recurrent inflammation can result in chronic obstructive pulmonary disease (COPD). Persistent particulate matter (PM) exposure increases systemic cardiopulmonary inflammation, which reduces lung function and can exacerbate asthma, COPD, and idiopathic pulmonary fibrosis (IPF). While recurrent inflammation underlies environmentally induced pulmonary morbidity and may drive the disease process, our understanding of inflammation resolution in this context is limited. This review aims to explore inflammation resolution biology and its role in chronic environmental lung disease(s).
Collapse
Affiliation(s)
- Jacqui M Marzec
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Srikanth S Nadadur
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
50
|
Herman KD, Wright CG, Marriott HM, McCaughran SC, Bowden KA, Collins MO, Renshaw SA, Prince LR. The EGFR/ErbB inhibitor neratinib modifies the neutrophil phosphoproteome and promotes apoptosis and clearance by airway macrophages. Front Immunol 2022; 13:956991. [PMID: 35967296 PMCID: PMC9371615 DOI: 10.3389/fimmu.2022.956991] [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: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/05/2022] Open
Abstract
Dysregulated neutrophilic inflammation can be highly destructive in chronic inflammatory diseases due to prolonged neutrophil lifespan and continual release of histotoxic mediators in inflamed tissues. Therapeutic induction of neutrophil apoptosis, an immunologically silent form of cell death, may be beneficial in these diseases, provided that the apoptotic neutrophils are efficiently cleared from the tissue. Previous research in our group identified ErbB inhibitors as able to induce neutrophil apoptosis and reduce neutrophilic inflammation both in vitro and in vivo. Here, we extend that work using a clinical ErbB inhibitor, neratinib, which has the potential to be repurposed in inflammatory diseases. We show that neratinib reduces neutrophilic migration o an inflammatory site in zebrafish larvae. Neratinib upregulates efferocytosis and reduces the number of persisting neutrophil corpses in mouse models of acute, but not chronic, lung injury, suggesting that the drug may have therapeutic benefits in acute inflammatory settings. Phosphoproteomic analysis of human neutrophils shows that neratinib modifies the phosphorylation of proteins regulating apoptosis, migration, and efferocytosis. This work identifies a potential mechanism for neratinib in treating acute lung inflammation by upregulating the clearance of dead neutrophils and, through examination of the neutrophil phosphoproteome, provides important insights into the mechanisms by which this may be occurring.
Collapse
Affiliation(s)
- Kimberly D. Herman
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and The Bateson Centre, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Carl G. Wright
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Helen M. Marriott
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Sam C. McCaughran
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Kieran A. Bowden
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Mark O. Collins
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease and The Bateson Centre, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Lynne R. Prince
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|