1
|
Hu B, Yuan L, Zhang Y, Deng W, Zhong H, Miao C, Wang C, Cai J. Dietary Vitamin C Intake Affects Lung Function Through White Blood Cell. Food Sci Nutr 2025; 13:e70299. [PMID: 40370419 PMCID: PMC12076004 DOI: 10.1002/fsn3.70299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025] Open
Abstract
As an antioxidant, vitamin C has been increasingly used in the treatment of various pulmonary diseases in recent years. However, the mechanism by which vitamin C affects lung function remains unclear to this day. Given its low cost and low risk, vitamin C is highly suitable for widespread use as a conventional treatment, making research into its mechanisms of influencing lung function necessary. Considering the potential association between vitamin C and white blood cells (WBCs), it may influence lung function by affecting white blood count (WBC). The potential impacts of WBCs on the lungs may include damage to the lung parenchyma through proteases released by these cells, as well as the effects of inflammatory factors on alveolar epithelial cells, among other mechanisms. This study aims to explore the potential relationship between dietary vitamin C intake, WBC, and lung function through a cross-sectional study. This cross-sectional study included data from 15,738 participants in the National Health and Nutrition Examination Survey (NHANES) from three time periods: 2007-2008, 2009-2010, and 2011-2012. Parallel mediation analysis was conducted using a multivariable logistic regression model to assess the relationships between dietary vitamin C intake, WBC, and lung function. Following the cross-sectional study, we further incorporated Mendelian randomization (MR) analysis to strengthen the validity of the findings. The results of this cross-sectional study showed that dietary vitamin C intake was negatively associated with WBC (p < 0.05, β < 0), while WBC was also negatively associated with lung function. In contrast, dietary vitamin C intake was positively associated with lung function, with a significant positive mediation effect (p < 0.05, β > 0). These findings suggest that vitamin C may influence lung function by modulating WBC levels. The study may reveal part of the mechanism through which vitamin C affects lung function, specifically through the mediation of WBC. The roles of inflammation and proteases could be potential underlying mechanisms. However, further research is required to clarify the biochemical mechanisms. This study provides a reference for the clinical use of vitamin C in the treatment of related pulmonary diseases and promotes further research into its broader effects.
Collapse
Affiliation(s)
- Biao Hu
- Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
- Guangzhou Medical UniversityGuangzhouChina
| | - Lu Yuan
- Guangzhou Medical UniversityGuangzhouChina
| | | | | | | | | | | | - Jiaxin Cai
- Department of RadiologyThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
2
|
Kotlyarov S, Oskin D. The Role of Inflammation in the Pathogenesis of Comorbidity of Chronic Obstructive Pulmonary Disease and Pulmonary Tuberculosis. Int J Mol Sci 2025; 26:2378. [PMID: 40141021 PMCID: PMC11942565 DOI: 10.3390/ijms26062378] [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: 01/11/2025] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The comorbid course of chronic obstructive pulmonary disease (COPD) and pulmonary tuberculosis is an important medical and social problem. Both diseases, although having different etiologies, have many overlapping relationships that mutually influence their course and prognosis. The aim of the current review is to discuss the role of different immune mechanisms underlying inflammation in COPD and pulmonary tuberculosis. These mechanisms are known to involve both the innate and adaptive immune system, including various cellular and intercellular interactions. There is growing evidence that immune mechanisms involved in the pathogenesis of both COPD and tuberculosis may jointly contribute to the tuberculosis-associated obstructive pulmonary disease (TOPD) phenotype. Several studies have reported prior tuberculosis as a risk factor for COPD. Therefore, the study of the mechanisms that link COPD and tuberculosis is of considerable clinical interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Dmitry Oskin
- Department of Infectious Diseases and Phthisiology, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
3
|
Fraser DD, Roy S, Kuruc M, Quintero M, Van Nynatten LR, Cepinskas G, Zheng H, Soherwardy A, Roy D. Functional mass spectrometry indicates anti-protease and complement activity increase with COVID-19 severity. Exp Biol Med (Maywood) 2025; 250:10308. [PMID: 39949890 PMCID: PMC11813650 DOI: 10.3389/ebm.2025.10308] [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: 07/06/2024] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
Investigations on some innate immunity proteins can yield misleading information, as investigators often rely on static measurements and assume a direct correlation to function. As protein function is often not directly proportional to protein abundance, and mechanistic pathways are interconnected and under constant feedback regulatory control, functional analysis is required. In this study, we used functional mass spectrometry to measure anti-protease and complement activity in plasma obtained from coronavirus disease 2019 (COVID-19) patients. Our data suggests that within 48 h of hospital admission, COVID-19 patients undergo a protease storm with significantly elevated neutrophil elastase (p < 0.001) and lymphocyte granzyme B (p < 0.01), while, anti-protease activity is significantly increased, including alpha-1 antitrypsin (AAT; p < 0.001) and alpha-1-antichymotrypsin (ACT; p < 0.001). Concurrently, the ratio of C3a to C3beta activity significantly decreased with increasing COVID-19 severity, suggesting more complement activation (Mild COVID-19 p < 0.05; Severe COVID-19 p < 0.001). Activity levels of AAT, ACT and C3a/C3beta remained unchanged over 10 hospital days. Our data suggests that COVID-19 is associated with both a protease storm and complement activation, with the former somewhat balanced with increased anti-protease activity. Evaluation of the AAT/ACT ratio and C3a/C3beta ratio indicated that COVID-19 severity is associated with both neutrophil elastase neutralization and complement activation.
Collapse
Affiliation(s)
- Douglas D. Fraser
- London Health Sciences Centre Research Institute, London, ON, Canada
- Pediatrics, Western University, London, ON, Canada
| | - Swapan Roy
- Biotech Support Group LLC, Monmouth Junction, NJ, United States
| | - Matt Kuruc
- Biotech Support Group LLC, Monmouth Junction, NJ, United States
| | | | | | - Gediminas Cepinskas
- London Health Sciences Centre Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
| | - Haiyan Zheng
- Rutgers Center for Integrative Proteomics, Rutgers University, Piscataway, NJ, United States
| | - Amenah Soherwardy
- Rutgers Center for Integrative Proteomics, Rutgers University, Piscataway, NJ, United States
| | - Devjit Roy
- Nathan Littauer Hospital, Gloversville, NY, United States
| |
Collapse
|
4
|
Ahmed NT, Kummarapurugu AB, Zheng S, Bulut G, Kang L, Batheja A, Hawkridge A, Voynow JA. Neutrophil Elastase Targets Select Proteins on Human Blood-Monocyte-Derived Macrophage Cell Surfaces. Int J Mol Sci 2024; 25:13038. [PMID: 39684750 DOI: 10.3390/ijms252313038] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Neutrophil elastase (NE) has been reported to be a pro-inflammatory stimulus for macrophages. The aim of the present study was to determine the impact of NE exposure on the human macrophage proteome and evaluate its impact on pro-inflammatory signals. Human blood monocytes from healthy volunteers were differentiated to macrophages and then exposed to either 500 nM of NE or control vehicle for 2 h in triplicate. Label-free quantitative proteomics analysis identified 41 differentially expressed proteins in the NE versus control vehicle datasets. A total of 26 proteins were downregulated and of those, 21 were cell surface proteins. Importantly, four of the cell surface proteins were proteoglycans: neuropilin 1 (NRP1), syndecan 2 (SDC2), glypican 4 (GPC4), and CD99 antigen-like protein 2 (CD99L2) along with neuropilin 2 (NRP2), CD99 antigen (CD99), and endoglin (ENG) which are known interactors. Additional NE-targeted proteins related to macrophage function were also measured including CD40, CD48, SPINT1, ST14, and MSR1. Collectively, this study provides a comprehensive unbiased view of selective NE-targeted cell surface proteins in chronically inflamed lungs.
Collapse
Affiliation(s)
- Nadia Tasnim Ahmed
- School of Pharmacy, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | | | - Shuo Zheng
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| | - Gamze Bulut
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| | - Le Kang
- Department of Biostatistics, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Aashish Batheja
- School of Medicine, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Adam Hawkridge
- School of Pharmacy, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Judith A Voynow
- Children's Hospital of Richmond at VCU, Richmond, VA 23298, USA
| |
Collapse
|
5
|
Zheng Y, Hu J, Chen J, Wang H, Zhao Z, Zhu H, Li Z, Wang N, Chen X, Liu M, Luo Z, Zhang S, Zhang H, Xuan X, Li X, Xue L, Wang G, Wu J. Association between dust exposure and lung function levels in steelworkers: mediation analysis of inflammatory biomarkers. Int Arch Occup Environ Health 2024; 97:971-980. [PMID: 39306641 DOI: 10.1007/s00420-024-02101-y] [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: 06/17/2024] [Accepted: 08/26/2024] [Indexed: 11/14/2024]
Abstract
PURPOSE This investigation aimed to examine the mediating effect of inflammatory biomarkers on the relationship between dust exposure and lung function levels among steelworkers. METHODS The study comprised 2,315 front-line workers employed at an iron and steel company in Tangshan, who underwent occupational health assessments through cluster sampling. Demographic and lifestyle data were collected via a self-administered questionnaire, while physical examinations measured parameters such as height and weight. Lung function was assessed using a portable pulmonary function tester (CHEST). Blood cell counts were uniformly analyzed using a Mindray fully automated biochemistry analyzer (BS-800). Inflammatory biomarkers, including leukocyte count, neutrophil count, lymphocyte count, and platelet count, were assessed, and the neutrophil-to-lymphocyte ratio and systemic immune inflammation index were computed. Generalized linear models and Spearman rank correlation analyses were employed to explore the interplay among dust exposure, inflammatory biomarkers, and alterations in lung function. A mediation analysis model was constructed to elucidate how inflammatory biomarkers mediate the relationship between dust exposure and lung function levels. RESULTS After adjusting for covariates, dust exposure was significantly associated with reduced lung function levels, with statistically significant differences observed between dust-exposed and non-exposed groups across various lung function indicators (P < 0.001). In the dust-exposed group, inflammatory biomarkers were elevated, showing significant correlations with FVC and FEV1 (P < 0.05). However, the correlation between FEV1/FVC and various inflammatory biomarkers was insignificant (P > 0.05). Mediation analysis revealed that white blood cells and neutrophils partially mediated the association between dust exposure and FVC, with proportions of 1.75% and 1.09%, respectively. Similarly, white blood cells, neutrophils, and the systemic immune inflammation index partially mediated the association between dust exposure and FEV1, with proportions of 1.15%, 0.82%, and 0.82%, respectively. CONCLUSION In conclusion, dust exposure poses a risk for decreased lung function levels. Inflammatory biomarkers derived from blood cells offer a valuable and easily obtainable means of identifying changes in lungfunction levels. Among these biomarkers, white blood cells, neutrophils, and the systemic immune inflammation index significantly mediate the association between dust exposure and lung function levels, although further exploration is needed to understand their underlying mechanisms.
Collapse
Affiliation(s)
- Yizhan Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Jiaqi Hu
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Jiaqi Chen
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Huan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Ziqi Zhao
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Hongmin Zhu
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Zheng Li
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Nan Wang
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Xinyang Chen
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Mingyue Liu
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Zhenghao Luo
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Shangmingzhu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Haoruo Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Xiaoqing Xuan
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Xiaoming Li
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Ling Xue
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China
| | - Guoli Wang
- School of Emergency Management and Safety Engineering, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China.
| | - Jianhui Wu
- Department of Epidemiology and Health Statistics, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China.
- Hebei Key Laboratory of Coal Health and Safety, School of Public Health, North China University of Science and Technology, No. 21 Bohai Avenue, Caofeidian New Town, Tangshan, 063210, China.
| |
Collapse
|
6
|
Yuan Q, Xiao LW, Zhang Y, Li L, Xia T, Xu Q, Xing SG, Wang LS. Inverted U-Shaped relationship Between Systemic Immune-Inflammation Index and Pulmonary Function: A Large Population-Based Study in US Adults. Int J Chron Obstruct Pulmon Dis 2024; 19:1971-1987. [PMID: 39247667 PMCID: PMC11379031 DOI: 10.2147/copd.s471068] [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: 05/27/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Background Systemic immune-inflammation index (SII) is a novel comprehensive inflammatory marker. Inflammation is associated with impaired lung function. We aimed to explore the possible relationship between SII and lung function to examine the potential of SII in predicting lung function decline. Methods A cross-sectional survey was conducted using the data of the NHANES from 2007 to 2012. Multiple linear regression models were used to analyze the linear relationship between SII and pulmonary functions. Sensitivity analyses, subgroup analyses, and interaction tests were used to examine the robustness of this relationship across populations. Fitted smooth curves and threshold effect analysis were used to describe the nonlinear relationships. Results A total of 10,125 patients were included in this study. After adjusting for all covariates, multiple linear regression model analysis showed that high Log2-SII level was significantly associated with decreased FVC(β, -23.4061; 95% CI, -42.2805- -4.5317), FEV1(β, -46.7730; 95% CI, -63.3371- -30.2089), FEV1%(β, -0.7923; 95% CI, -1.1635- -0.4211), FEV1/FVC(β, -0.6366; 95% CI, -0.8328- -0.4404) and PEF(β, -121.4468; 95% CI,-164.1939- -78.6998). The negative correlation between Log2-SII and pulmonary function indexes remained stable in trend test and stratified analysis. Inverted U-shaped relationships between Log2-SII and FVC, FEV1, FEV1%, and PEF were observed, while a negative linear correlation existed between FEV1/FVC and Log2-SII. The cutoff values of the nonlinear relationship between Log2-SII and FVC, FEV1, FEV1%, PEF were 8.3736, 8.0688, 8.3745, and 8.5255, respectively. When SII exceeded the critical value, the lung function decreased significantly. Conclusion This study found a close correlation between SII and pulmonary function indicators. This study investigated the SII threshold when lung functions began to decline in the overall population. SII may become a promising serological indicator for predicting lung function decline. However, prospective studies were needed further to establish the causal relationship between these two factors.
Collapse
Affiliation(s)
- Qian Yuan
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Long-Wu Xiao
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Yao Zhang
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Long Li
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Teng Xia
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Qing Xu
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Shi-Gui Xing
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Liu-Shun Wang
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| |
Collapse
|
7
|
Tu Y, Jiang T, Wu Y, Luo J, Sun S, Liu C, Yu P, Chen A, Ji H, Wan Y, Yu L, Shi L. Difference of Antrochoanal Polyps Between Children and Adults in the Chinese Population. Laryngoscope 2024; 134:2093-2099. [PMID: 37916785 DOI: 10.1002/lary.31154] [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: 05/25/2023] [Revised: 09/18/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
OBJECTIVE This study aims to find the difference in clinical and immunopathological characteristics between children and adults with antrochoanal polyps (ACPs) in the Chinese population. METHODS The clinical data of 69 patients diagnosed with ACPs were retrospectively analyzed. Cytokine levels in 16 controls and 40 ACPs tissues were determined by quantitative real-time polymerase chain reaction (qPCR). The expression of matrix metalloproteinase (MMP)-9 was measured using qPCR, immunofluorescent staining, and western blot. RESULTS There were 51 (73.9%) children (<18 years old) and 18 (26.1%) adults (≥18 years old). The sex ratio differed significantly between the two groups (p = 0.0032). There were no significant differences in the nasal side of ACPs and approaches to surgery between the two groups. In both groups, the most common symptom was nasal obstruction, followed by nasal discharge. As for associated nasal diseases, there was a significant difference between the two groups in septal deviation (p = 0.0223). Adult patients showed significantly higher expression of IL-8 mRNA than children (p = 0.0424). The mRNA and protein levels of MMP-9 were also significantly higher in adult patients than in children (p = 0.0498 and 0.0009, respectively). CONCLUSION In the Chinese population, the comorbidities and immunopathological characteristics of adult ACP patients are different from those of children. The level of IL-8 and MMP-9 was significantly higher in ACPs of adults than in children, which may contribute to the more severe tissue remolding in adult ACP patients. LEVEL OF EVIDENCE 3 Laryngoscope, 134:2093-2099, 2024.
Collapse
Affiliation(s)
- Yanyi Tu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Tianjiao Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Yisha Wu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Jinfeng Luo
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Shujuan Sun
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Chuanping Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Peng Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Aiping Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Hongzhi Ji
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Yuzhu Wan
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Liang Yu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, China
| |
Collapse
|
8
|
Chalmers JD, Kettritz R, Korkmaz B. Dipeptidyl peptidase 1 inhibition as a potential therapeutic approach in neutrophil-mediated inflammatory disease. Front Immunol 2023; 14:1239151. [PMID: 38162644 PMCID: PMC10755895 DOI: 10.3389/fimmu.2023.1239151] [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: 06/12/2023] [Accepted: 11/14/2023] [Indexed: 01/03/2024] Open
Abstract
Neutrophils have a critical role in the innate immune response to infection and the control of inflammation. A key component of this process is the release of neutrophil serine proteases (NSPs), primarily neutrophil elastase, proteinase 3, cathepsin G, and NSP4, which have essential functions in immune modulation and tissue repair following injury. Normally, NSP activity is controlled and modulated by endogenous antiproteases. However, disruption of this homeostatic relationship can cause diseases in which neutrophilic inflammation is central to the pathology, such as chronic obstructive pulmonary disease (COPD), alpha-1 antitrypsin deficiency, bronchiectasis, and cystic fibrosis, as well as many non-pulmonary pathologies. Although the pathobiology of these diseases varies, evidence indicates that excessive NSP activity is common and a principal mediator of tissue damage and clinical decline. NSPs are synthesized as inactive zymogens and activated primarily by the ubiquitous enzyme dipeptidyl peptidase 1, also known as cathepsin C. Preclinical data confirm that inactivation of this protease reduces activation of NSPs. Thus, pharmacological inhibition of dipeptidyl peptidase 1 potentially reduces the contribution of aberrant NSP activity to the severity and/or progression of multiple inflammatory diseases. Initial clinical data support this view. Ongoing research continues to explore the role of NSP activation by dipeptidyl peptidase 1 in different disease states and the potential clinical benefits of dipeptidyl peptidase 1 inhibition.
Collapse
Affiliation(s)
- James D. Chalmers
- Department of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Ralph Kettritz
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation Between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité Universitätsmedizin, Berlin, Germany
| | - Brice Korkmaz
- INSERM UMR-1100, Research Center for Respiratory Diseases, University of Tours, Tours, France
| |
Collapse
|
9
|
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
|
10
|
Tromsdorf N, Ullrich FTH, Rethmeier M, Sommerhoff CP, Schaschke N. E-64c-Hydrazide Based Cathepsin C Inhibitors: Optimizing the Interactions with the S1'-S2' Area. ChemMedChem 2023; 18:e202300218. [PMID: 37424408 DOI: 10.1002/cmdc.202300218] [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: 04/23/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
The zymogens of the neutrophil serine proteases elastase, proteinase 3, and cathepsin G are converted proteolytically into their pro-inflammatory active forms by the action of cathepsin C. The inhibition of this cysteine protease therefore is an interesting therapeutic approach for the treatment of inflammatory disorders with a high neutrophil burden such as COPD. Based on E-64c-hydrazide as lead structure, we have recently developed a covalently acting cathepsin C inhibitor using a n-butyl residue attached at the amine nitrogen of the hydrazide moiety to efficiently address the deep hydrophobic S2 pocket. To further optimize the affinity and selectivity profile of this inhibitor, the S1'-S2' area was now investigated by a combinatorial approach, showing that Nle-tryptamide is a ligand superior to the initially used Leu-isoamylamide. Using the neutrophil precursor line U937 as a cell culture model, this optimized inhibitor blocks the intracellular cathepsin C activity and thereby suppresses the activation of neutrophil elastase.
Collapse
Affiliation(s)
- Nora Tromsdorf
- Fakultät für Chemie, Hochschule Aalen, Beethovenstraße 1, 73430, Aalen, Germany
| | - Fabian T H Ullrich
- Institut für Didaktik und Ausbildungsforschung in der Medizin und Institut für Laboratoriumsmedizin, LMU Klinikum, LMU München, Pettenkoferstraße 8a, 80336, München, Germany
| | - Markus Rethmeier
- Fakultät für Chemie, Universität Bielefeld, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Christian P Sommerhoff
- Institut für Didaktik und Ausbildungsforschung in der Medizin und Institut für Laboratoriumsmedizin, LMU Klinikum, LMU München, Pettenkoferstraße 8a, 80336, München, Germany
| | - Norbert Schaschke
- Fakultät für Chemie, Hochschule Aalen, Beethovenstraße 1, 73430, Aalen, Germany
| |
Collapse
|
11
|
Chen Y, Wu J, Li R, Kang W, Zhao A, Yin Y, Tong S, Yuan J, Li S. Individual and joint association of phenols, parabens, and phthalates with childhood lung function: Exploring the mediating role of peripheral immune responses. JOURNAL OF HAZARDOUS MATERIALS 2023; 454:131457. [PMID: 37099904 DOI: 10.1016/j.jhazmat.2023.131457] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
The functioning of the respiratory system can be interfered with by exposure to mixtures of environmental chemicals, however, the evidence is still ambiguous. We evaluated the association of exposure to mixtures of 14 chemicals, including 2 phenols, 2 parabens, and 10 phthalates, with four major lung function metrics. Based on data from the National Health and Nutrition Examination Survey 2007-2012, this analysis was conducted among 1462 children aged 6-19 years. Linear regression, Bayesian kernel machine regression, quantile-based g-computation regression, and a generalized additive model were performed to estimate the associations. Mediation analyses were performed to investigate plausible biological pathways mediated by immune cells. Our results indicated that the phenols, parabens, and phthalates mixture was negatively related to lung function parameters. And BPA and PP were identified as important contributors to negative associations with FEV1, FVC, and PEF, with non-linear relationships observed between BPA and those outcomes. The most influential factor for a probable FEF25-75 % decline was MCNP. BPA, and MCNP had an interaction effect on FEF25-75 %. The association of PP with FVC and FEV1 has been postulated to be mediated by neutrophils and monocytes. The findings offer insights into the associations of chemical mixtures with respiratory health and the possible driving mechanism, which would be of significance in adding novel evidence of the role of peripheral immune responses, as well as calling for remediation actions to be prioritized during childhood.
Collapse
Affiliation(s)
- Yiting Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhong Wu
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenhui Kang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anda Zhao
- Department of Nutrition, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shilu Tong
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Biostatistics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; School of Public Health, Institute of Environment and Population Health, Anhui Medical University, Hefei, China; School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - Jiajun Yuan
- Child Health Advocacy Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenghui Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Rojas-Quintero J, Laucho-Contreras ME, Wang X, Fucci QA, Burkett PR, Kim SJ, Zhang D, Tesfaigzi Y, Li Y, Bhashyam AR, Li Z, Khamas H, Celli B, Pilon AL, Polverino F, Owen CA. CC16 augmentation reduces exaggerated COPD-like disease in Cc16-deficient mice. JCI Insight 2023; 8:130771. [PMID: 36787195 PMCID: PMC10070105 DOI: 10.1172/jci.insight.130771] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Low Club Cell 16 kDa protein (CC16) plasma levels are linked to accelerated lung function decline in patients with chronic obstructive pulmonary disease (COPD). Cigarette smoke-exposed (CS-exposed) Cc16-/- mice have exaggerated COPD-like disease associated with increased NF-κB activation in their lungs. It is unclear whether CC16 augmentation can reverse exaggerated COPD in CS-exposed Cc16-/- mice and whether increased NF-κB activation contributes to the exaggerated COPD in CS-exposed Cc16-/- lungs. CS-exposed WT and Cc16-/- mice were treated with recombinant human CC16 (rhCC16) or an NF-κB inhibitor versus vehicle beginning at the midpoint of the exposures. COPD-like disease and NF-κB activation were measured in the lungs. RhCC16 limited the progression of emphysema, small airway fibrosis, and chronic bronchitis-like disease in WT and Cc16-/- mice partly by reducing pulmonary inflammation (reducing myeloid leukocytes and/or increasing regulatory T and/or B cells) and alveolar septal cell apoptosis, reducing NF-κB activation in CS-exposed Cc16-/- lungs, and rescuing the reduced Foxj1 expression in CS-exposed Cc16-/- lungs. IMD0354 treatment reduced exaggerated lung inflammation and rescued the reduced Foxj1 expression in CS-exposed Cc16-/- mice. RhCC16 treatment reduced NF-κB activation in luciferase reporter A549 cells. Thus, rhCC16 treatment limits COPD progression in CS-exposed Cc16-/- mice partly by inhibiting NF-κB activation and represents a potentially novel therapeutic approach for COPD.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Eugenia Laucho-Contreras
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Fundación Neumológica Colombiana, Bogotá, Colombia
| | - Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Clinical and Experimental Therapeutics program, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Quynh-Anh Fucci
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick R Burkett
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Se-Jin Kim
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics program, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yuhong Li
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Abhiram R Bhashyam
- Department of Orthopedic Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Zhang Li
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Haider Khamas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Bartolome Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Francesca Polverino
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
杨 红, 张 正, 王 左. [The Relationship Between Oral Microbiota and Chronic Obstructive Pulmonary Disease]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:54-60. [PMID: 36647643 PMCID: PMC10409024 DOI: 10.12182/20230160502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Indexed: 01/18/2023]
Abstract
Oral microbiota have a complex impact on the host's health and disease states. It has been found that the composition of lung flora bears a striking resemblance to the composition of oral flora. Moreover, oral pathogenic bacteria have been detected in the sputum and bronchoalveolar lavage fluid of patients with chronic obstructive pulmonary disease (COPD), suggesting that oral microbiota play an important role in the pathogenesis and development of COPD. Findings from lots of studies have shown that oral microbiota may participate in the pathogenesis and development of COPD through non-specific immune response, specific immune response, and the activities of protein hydrolase. Herein, we mainly summarized the available evidence on the relationship between oral microbiota and COPD. By examining the relationship between the two, we elaborated on the application of oral microbiota in the diagnosis and prevention of COPD, discussed possible directions for future research, and provided references for developing new therapeutic approaches.
Collapse
Affiliation(s)
- 红嘉 杨
- 首都医科大学附属北京朝阳医院 口腔科 (北京 100020)Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - 正 张
- 首都医科大学附属北京朝阳医院 口腔科 (北京 100020)Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
- 天津市口腔医院 南开大学医学院 (天津 300041)Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - 左敏 王
- 首都医科大学附属北京朝阳医院 口腔科 (北京 100020)Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| |
Collapse
|
14
|
Li D, Wang T, Ma Q, Zhou L, Le Y, Rao Y, Jin L, Pei Y, Cheng Y, Huang C, Gai X, Sun Y. IL-17A Promotes Epithelial ADAM9 Expression in Cigarette Smoke-Related COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:2589-2602. [PMID: 36267325 PMCID: PMC9578481 DOI: 10.2147/copd.s375006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Background It has been reported that a disintegrin and metalloproteinase 9 (ADAM9) is involved in the pathogenesis of cigarette smoke (CS)-associated chronic obstructive pulmonary disease (COPD). But how CS exposure leads to upregulation of ADAM9 remains unknown. Methods Patients who underwent lobectomy for a solitary pulmonary nodule were enrolled and divided into three groups: non-smokers with normal lung function, smokers without COPD and smoker patients with COPD. Immunoreactivity of interleukin (IL)-17A and ADAM9 in small airways and alveolar walls was measured by immunohistochemistry. Wild-type and Il17a−/− C57BL/6 mice were exposed to CS for six months, and ADAM9 expression in the airway epithelia was measured by immunoreactivity. In addition, the protein and mRNA expression levels of IL-17A and ADAM9 were assessed in CS extract (CSE) and/or IL-17A-treated human bronchial epithelial (HBE) cells. Results The immunoreactivity of ADAM9 was increased in the airway epithelia and alveolar walls of patients with COPD compared to that of the controls. The expression of IL-17A was also upregulated in airway epithelial cells of patients with COPD and correlated positively with the level of ADAM9. The results from the animal model showed that Il17a−/− mice were protected from emphysema induced by CS exposure, together with a reduced level of ADAM9 expression in the airway epithelia, suggesting a possible link between ADAM9 and IL-17A. Consistently, our in vitro cell model showed that CSE stimulated the expression of ADAM9 and IL-17A in HBE cells in a dose- and time-dependent manner. Recombinant IL-17A induced ADAM9 upregulation in HBE cells and had a synergistic effect with CSE, whereas blocking IL-17A inhibited CSE-induced ADAM9 expression. Further analysis revealed that IL-17A induced c-Jun N-terminal kinase (JNK) phosphorylation, thereby increasing ADAM9 expression. Conclusion Our results revealed a novel role of IL-17A in CS-related COPD, where IL-17A contributes to ADAM9 expression by activating JNK signaling.
Collapse
Affiliation(s)
- Danyang Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Tong Wang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Qianli Ma
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Liang Jin
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yuqiang Pei
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yaning Cheng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, People’s Republic of China
| | - Chen Huang
- Center of Basic Medical Research, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China,Correspondence: Xiaoyan Gai; Yongchang Sun, Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, People’s Republic of China, Email ;
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| |
Collapse
|
15
|
Asadi Jozani K, Kouthouridis S, Hirota JA, Zhang B. Next generation preclinical models of lung development, physiology and disease. CAN J CHEM ENG 2022. [DOI: 10.1002/cjce.24581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Kimia Asadi Jozani
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
| | - Sonya Kouthouridis
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| | - Jeremy Alexander Hirota
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Medicine, Division of Respirology McMaster University Hamilton Ontario Canada
- Firestone Institute for Respiratory Health St. Joseph’s Hospital, Hamilton Ontario Canada
| | - Boyang Zhang
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| |
Collapse
|
16
|
Sajjan U. Secretory Cells - New Players in Small Airway Mucosal Immunity? Am J Respir Cell Mol Biol 2022; 67:269-270. [PMID: 35704450 DOI: 10.1165/rcmb.2022-0210ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Umadevi Sajjan
- Temple University, 6558, Thoracic Medicine and Surgery, Philadelphia, Pennsylvania, United States;
| |
Collapse
|
17
|
Zhu L, Duerr J, Zhou-Suckow Z, Wagner WL, Weinheimer O, Salomon JJ, Leitz D, Konietzke P, Yu H, Ackermann M, Stiller W, Kauczor HU, Mall MA, Wielpütz MO. µCT to quantify muco-obstructive lung disease and effects of neutrophil elastase knockout in mice. Am J Physiol Lung Cell Mol Physiol 2022; 322:L401-L411. [PMID: 35080183 DOI: 10.1152/ajplung.00341.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Muco-obstructive lung diseases are characterized by airway obstruction and hyperinflation, which can be quantified by imaging. Our aim was to evaluate µCT for longitudinal quantification of muco-obstructive lung disease in β-epithelial Na+ channel overexpressing (Scnn1b-TG) mice and of the effects of neutrophil elastase (NE) knockout on its progression. Lungs from wild-type (WT), NE-/-, Scnn1b-TG, and Scnn1b-TG/NE-/- mice were scanned with 9 µm resolution at 0, 5, 14 and 60 days of age, and airway and parenchymal disease was quantified. Mucus adhesion lesions (MAL) were persistently increased in Scnn1b-TG compared to WT mice from 0 days (20.25±6.50 vs. 9.60±2.07, P<0.05), and this effect was attenuated in Scnn1b-TG/NE-/- mice (5.33±3.67, P<0.001). Airway wall area percentage (WA%) was increased in Scnn1b-TG mice compared to WT from 14 days onward (59.2±6.3% vs. 49.8±9.0%, P<0.001) but was similar in Scnn1b-TG/NE-/- compared to WT at 60 days (46.4±9.2% vs. 45.4±11.5%, P=0.97). Air proportion (Air%) and mean linear intercept (Lm) were persistently increased in Scnn1b-TG compared to WT from 5 days on (53.9±4.5% vs. 30.0±5.5% and 78.82±8.44µm vs. 65.66±4.15µm, respectively, P<0.001), whereas in Scnn1b-TG/NE-/- Air% and Lm were similar to WT from birth (27.7±5.5% vs.27.2±5.9%, P =0.92 and 61.48±9.20µm vs. 61.70±6.73µm, P=0.93, respectively). Our results suggest that µCT is sensitive to detect the onset and progression of muco-obstructive lung disease and effects of genetic deletion of NE on morphology of airways and lung parenchyma in Scnn1b-TG mice, and that it may serve as a sensitive endpoint for preclinical studies of novel therapeutic interventions for muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| | - Julia Duerr
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Lung Research (DZL), associated partner Berlin, Germany
| | - Zhe Zhou-Suckow
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Willi L Wagner
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| | - Oliver Weinheimer
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| | - Johanna Jessica Salomon
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany
| | - Dominik Leitz
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Lung Research (DZL), associated partner Berlin, Germany
| | - Philip Konietzke
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Institute of Pathology and Department of Molecular Pathology, Helios University Clinic Wuppertal, University of Witten-Herdecke, Wuppertal, Germany
| | - Wolfram Stiller
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Translational Pulmonology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Lung Research (DZL), associated partner Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Mark Oliver Wielpütz
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
18
|
Wu X, Wang C, Li H, Meng H, Jie J, Fu M, Bai Y, Li G, Wei W, Feng Y, Li M, Guan X, He M, Zhang X, Guo H. Circulating white blood cells and lung function impairment: the observational studies and Mendelian randomization analysis. Ann Med 2021; 53:1118-1128. [PMID: 34259107 PMCID: PMC8280897 DOI: 10.1080/07853890.2021.1948603] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Circulating white blood cell (WBC) counts have been related to lung function impairment, but causal relationship was not established. We aimed to evaluate independent effects and causal relationships of WBC subtypes with lung function. METHODS The 19,159 participants from NHANES 2011-2012 (n = 3570), coke-oven workers (COW, n = 1762) and Dongfeng-Tongji (DFTJ, n = 13,827) cohorts were included in the observational studies. The associations between circulating counts of WBC subtypes and prebronchodilator lung function were evaluated by linear regression models and LASSO regression was used to select effective WBC subtypes. Summary statistics for WBC-associated SNPs were extracted from literature, and Mendelian randomization (MR) analysis with inverse-variance weighted (IVW) method was applied to estimate the causal effects of total WBC and subtypes on lung function among 4012 subjects from COW (n = 1126) and DFTJ cohorts (n = 2886). RESULTS Total WBC counts were negatively associated with lung function among three populations and their pooled analysis indicated that per 1 × 109 cells/L increase in total WBC was associated with 36.13 (95% CI: 30.35, 41.91) mL and 25.23 (95% CI: 19.97, 30.50) mL decrease in FVC and FEV1, respectively. Independent associations with lung function were found for neutrophils, monocytes, eosinophils and basophils (all p < .05), except lymphocytes. Besides, IVW MR analysis showed that genetically predicted total WBC and neutrophil counts were associated with reduced FVC (p = .017 and .021, respectively) and FEV1 (p = .048 and .043, respectively). CONCLUSIONS WBC subtypes were independently associated with lower lung function except lymphocytes. Our findings suggest that circulating neutrophils may be causal factors in lung function impairment.KEY MESSAGESWhite blood cell (WBC) subtypes were negatively associated with lung function level except lymphocytes in the observational studies.Associations of WBC subtypes with lung function may be modified by sex and smoking.Mendelian randomization analysis shows that neutrophils may be causal factors in lung function impairment.
Collapse
Affiliation(s)
- Xiulong Wu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenming Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Meng
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiali Jie
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Fu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yansen Bai
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guyanan Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wei
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Feng
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengying Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Guan
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meian He
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Aloufi N, Alluli A, Eidelman DH, Baglole CJ. Aberrant Post-Transcriptional Regulation of Protein Expression in the Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:ijms222111963. [PMID: 34769392 PMCID: PMC8584689 DOI: 10.3390/ijms222111963] [Citation(s) in RCA: 4] [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: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable and prevalent respiratory disorder that is characterized by chronic inflammation and emphysema. COPD is primarily caused by cigarette smoke (CS). CS alters numerous cellular processes, including the post-transcriptional regulation of mRNAs. The identification of RNA-binding proteins (RBPs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as main factors engaged in the regulation of RNA biology opens the door to understanding their role in coordinating physiological cellular processes. Dysregulation of post-transcriptional regulation by foreign particles in CS may lead to the development of diseases such as COPD. Here we review current knowledge about post-transcriptional events that may be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Noof Aloufi
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medical Laboratory Technology, Applied Medical Science, Taibah University, Universities Road, Medina P.O. Box 344, Saudi Arabia
| | - Aeshah Alluli
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Carolyn J. Baglole
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
20
|
Lessieur EM, Liu H, Saadane A, Du Y, Tang J, Kiser J, Kern TS. Neutrophil-Derived Proteases Contribute to the Pathogenesis of Early Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2021; 62:7. [PMID: 34643662 PMCID: PMC8525836 DOI: 10.1167/iovs.62.13.7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Purpose Previous studies indicate that leukocytes, notably neutrophils, play a causal role in the capillary degeneration observed in diabetic retinopathy (DR), however, the mechanism by which they cause such degeneration is unknown. Neutrophil elastase (NE) is a protease released by neutrophils which participates in a variety of inflammatory diseases. In the present work, we investigated the potential involvement of NE in the development of early DR. Methods Experimental diabetes was induced in NE-deficient mice (Elane-/-), in mice treated daily with the NE inhibitor, sivelestat, and in mice overexpressing human alpha-1 antitrypsin (hAAT+). Mice were assessed for diabetes-induced retinal superoxide generation, inflammation, leukostasis, and capillary degeneration. Results In mice diabetic for 2 months, deletion of NE or selective inhibition of NE inhibited diabetes-induced retinal superoxide levels and inflammation, and inhibited leukocyte-mediated cytotoxicity of retinal endothelial cells. In mice diabetic for 8 months, genetic deletion of NE significantly inhibited diabetes-induced retinal capillary degeneration. Conclusions These results suggest that a protease released from neutrophils contributes to the development of DR, and that blocking NE activity could be a novel therapy to inhibit DR.
Collapse
Affiliation(s)
- Emma M Lessieur
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California - Irvine, Irvine, California, United States
| | - Haitao Liu
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States
| | - Aicha Saadane
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California - Irvine, Irvine, California, United States
| | - Yunpeng Du
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California - Irvine, Irvine, California, United States
| | - Jie Tang
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States
| | - Jianying Kiser
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California - Irvine, Irvine, California, United States
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California - Irvine, Irvine, California, United States.,Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States.,Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States.,Veterans Administration Medical Center Research Service, Long Beach, California, United States
| |
Collapse
|
21
|
Saidi A, Wartenberg M, Madinier JB, Ilango G, Seren S, Korkmaz B, Lecaille F, Aucagne V, Lalmanach G. Monitoring Human Neutrophil Activation by a Proteinase 3 Near-Infrared Fluorescence Substrate-Based Probe. Bioconjug Chem 2021; 32:1782-1790. [PMID: 34269060 DOI: 10.1021/acs.bioconjchem.1c00267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A near-infrared fluorescent (NIRF) substrate-based probe (SBP) was conceived to monitor secreted human proteinase 3 (hPR3) activity. This probe, called pro3-SBP, is shaped by a fused peptide hairpin loop structure, which associates a hPR3 recognition domain (Val-Ala-Asp-Nva-Ala-Asp-Tyr-Gln, where Nva is norvaline) and an electrostatic zipper (consisting of complementary polyanionic (d-Glu)5 and polycationic (d-Arg)5 sequences) in close vicinity of the N- and C-terminal FRET couple (fluorescent donor, sulfoCy5.5; dark quencher, QSY21). Besides its subsequent stability, no intermolecular fluorescence quenching was detected following its complete hydrolysis by hPR3, advocating that pro3-SBP could further afford unbiased imaging. Pro3-SBP was specifically hydrolyzed by hPR3 (kcat/Km= 440 000 ± 5500 M-1·s-1) and displayed a sensitive detection threshold for hPR3 (subnanomolar concentration range), while neutrophil elastase showed a weaker potency. Conversely, pro3-SBP was not cleaved by cathepsin G. Pro3-SBP was successfully hydrolyzed by conditioned media of activated human neutrophils but not by quiescent neutrophils. Moreover, unlike unstimulated neutrophils, a strong NIRF signal was specifically detected by confocal microscopy following neutrophil ionomycin-induced degranulation. Fluorescence release was abolished in the presence of a selective hPR3 inhibitor, indicating that pro3-SBP is selectively cleaved by extracellular hPR3. Taken together, the present data support that pro3-SBP could be a convenient tool, allowing straightforward monitoring of human neutrophil activation.
Collapse
Affiliation(s)
- Ahlame Saidi
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Mylène Wartenberg
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Jean-Baptiste Madinier
- Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology″, CNRS UPR 4301, Orléans 45071, France
| | - Guy Ilango
- IBiSA Electron Microscopy Platform, Université de Tours, Tours 37032, France
| | - Seda Seren
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Brice Korkmaz
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Fabien Lecaille
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Vincent Aucagne
- Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology″, CNRS UPR 4301, Orléans 45071, France
| | - Gilles Lalmanach
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| |
Collapse
|
22
|
Zhang G, Ma L, Bai L, Li M, Guo T, Tian B, He Z, Fu Q. Inflammatory microenvironment-targeted nanotherapies. J Control Release 2021; 334:114-126. [PMID: 33887284 DOI: 10.1016/j.jconrel.2021.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory microenvironments (IMEs) are common pathological characteristics and drive the development of multiple chronic diseases. Thus, IME-targeted therapies exhibit potential for the treatment of inflammatory diseases. Nanoplatforms have significant advantages in improving the efficiency of anti-inflammatory treatments. Owing to their improved therapeutic effects and reduced side effects, IME-targeted nanotherapies have recently drawn interest from the research community. This review introduces IMEs and discusses the application of IME-targeted nanotherapies for inflammatory diseases. The development of rational targeting strategies tailored to IMEs in damaged tissues can help promote therapies for chronic diseases.
Collapse
Affiliation(s)
- Guangshuai Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Lixue Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Lijun Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Tiange Guo
- Laboratory Animal Department, General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
23
|
Xue Y, Bai H, Peng B, Fang B, Baell J, Li L, Huang W, Voelcker NH. Stimulus-cleavable chemistry in the field of controlled drug delivery. Chem Soc Rev 2021; 50:4872-4931. [PMID: 33734247 DOI: 10.1039/d0cs01061h] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Stimulus-cleavable nanoscale drug delivery systems are receiving significant attention owing to their capability of achieving exquisite control over drug release via the exposure to specific stimuli. Central to the construction of such systems is the integration of cleavable linkers showing susceptibility to one stimulus or several stimuli with drugs, prodrugs or fluorogenic probes on the one hand, and nanocarriers on the other hand. This review summarises recent advances in stimulus-cleavable linkers from various research areas and the corresponding mechanisms of linker cleavage and biological applications. The feasibility of extending their applications to the majority of nanoscale drug carriers including nanomaterials, polymers and antibodies are further highlighted and discussed. This review also provides general design guidelines to incorporate stimulus-cleavable linkers into nanocarrier-based drug delivery systems, which will hopefully spark new ideas and applications.
Collapse
Affiliation(s)
- Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Jonathan Baell
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing, 211816, P. R. China
| | - Nicolas Hans Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. and Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia and Department of Materials Science & Engineering, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
24
|
Roe K. Potential New Treatments for Kawasaki Disease, Its Variations, and Multisystem Inflammatory Syndrome. ACTA ACUST UNITED AC 2021; 3:1076-1080. [PMID: 33786417 PMCID: PMC7993892 DOI: 10.1007/s42399-021-00872-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
The causation of Kawasaki disease has been a medical mystery for over 54 years. However, the causations of Kawasaki disease, its variations, and COVID-19-associated Multisystem Inflammatory Syndrome have been recently explained to involve high replication rate viral infections. In a subset of patients, the extensive antigen-antibody immune complexes that are not quickly cleared by phagocytosis will create a type III hypersensitivity immune reaction. The subsequent release of proteases and other enzymes and the expression or exposure of new immunogenic antigens due to protease attacks on basement membranes of epithelial cells or endothelial cells in blood vessels will induce new autoantibodies and cause Kawasaki disease, its variations, and COVID-19-related Multisystem Inflammatory Syndrome. There is now increasing evidence that a viral infection of a large surface area of tissue, such as the respiratory tract, gastrointestinal tract or blood vessels, and a resultant type III hypersensitivity immune reaction is the most plausible explanation for the causations of Kawasaki disease, its variations, and COVID-19-related Multisystem Inflammatory Syndrome. Furthermore, an improved understanding of these causations also suggests several potential new treatments which can be more effective.
Collapse
|
25
|
Wang X, Zhang D, Fucci QA, Dollery CM, Owen CA. Surface-bound matrix metalloproteinase-8 on macrophages: Contributions to macrophage pericellular proteolysis and migration through tissue barriers. Physiol Rep 2021; 9:e14778. [PMID: 33656791 PMCID: PMC7927794 DOI: 10.14814/phy2.14778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/30/2021] [Accepted: 02/05/2021] [Indexed: 01/21/2023] Open
Abstract
Objective MMP‐8 binds to surface‐bound tissue inhibitor of metalloproteinase‐1 (TIMP‐1) on PMNs to promote pericellular proteolysis during the development of inflammatory diseases associated with tissue destruction. Little is known about the biology of MMP‐8 in macrophages. We tested the hypotheses that: (1) MMP‐8 and TIMP‐1 are also expressed on the surface of activated macrophages, (2) surface‐bound MMP‐8 on macrophages promotes TIMP‐resistant pericellular proteolysis and macrophage migration through tissue barriers, and (3) MMP‐8 binds to surface‐bound TIMP‐1 on macrophages. Methods Surface MMP‐8 and TIMP‐1 levels were measured on human monocyte‐derived macrophages (MDM) and/or murine macrophages using immunostaining, biotin‐labeling, and substrate cleavage methods. The susceptibility of membrane‐bound Mmp‐8 on activated macrophages from wild‐type (WT) mice to TIMPs was measured. Migration of WT and Mmp‐8−/− macrophages through models of tissue barriers in vitro and the accumulation of peritoneal macrophages in WT versus Mmp‐8−/− mice with sterile peritonitis was compared. Surface levels of Mmp‐8 were compared on activated macrophages from WT and Timp‐1−/− mice. Results Lipopolysaccharides and a cluster of differentiation 40 ligand increased surface MMP‐8 and/or TIMP‐1 staining and surface type I collagenase activity on MDM and/or murine macrophages. Activated Mmp‐8−/− macrophages degraded less type I collagen than activated WT macrophages. The surface type‐I collagenase activity on WT macrophages was resistant to inhibition by Timp‐1. Peritoneal macrophage accumulation was similar in WT and Mmp‐8−/− mice with sterile acute peritonitis. However, Mmp‐8−/− macrophages migrated less efficiently through models of tissue barriers (especially those containing type I collagen) than WT cells. Activated WT and Timp‐1−/− macrophages had similar surface‐bound Mmp‐8 levels. Conclusions MMP‐8 and TIMP‐1 are expressed on the surface of activated human MDM and murine macrophages, but Mmp‐8 is unlikely to bind to surface‐bound Timp‐1 on these cells. Surface‐bound MMP‐8 contributes to TIMP‐resistant monocyte/macrophage pericellular proteolysis and macrophage migration through collagen‐containing tissue barriers.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA.,Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Quynh-Anh Fucci
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clare M Dollery
- Whittington Hospital, Wittington Health NHS Trust, London, UK
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Wang X, Rojas-Quintero J, Zhang D, Nakajima T, Walker KH, Peh HY, Li Y, Fucci QA, Tesfaigzi Y, Owen CA. A disintegrin and metalloproteinase domain-15 deficiency leads to exaggerated cigarette smoke-induced chronic obstructive pulmonary disease (COPD)-like disease in mice. Mucosal Immunol 2021; 14:342-356. [PMID: 32690871 PMCID: PMC8422911 DOI: 10.1038/s41385-020-0325-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/21/2020] [Accepted: 07/06/2020] [Indexed: 02/04/2023]
Abstract
A disintegrin and metalloproteinase domain-15 (ADAM15) is expressed by cells implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD), but its contributions to COPD are unknown. To address this gap, ADAM15 levels were measured in samples from cigarette smoke (CS)-versus air-exposed wild-type (WT) mice. CS-induced COPD-like disease was compared in CS-exposed WT, Adam15-/-, and Adam15 bone marrow chimeric mice. CS exposure increased Adam15 expression in lung macrophages and CD8+ T cells and to a lesser extent in airway epithelial cells in WT mice. CS-exposed Adam15-/- mice had greater emphysema, small airway fibrosis, and lung inflammation (macrophages and CD8+ T cells) than WT mice. Adam15 bone marrow chimera studies revealed that Adam15 deficiency in leukocytes led to exaggerated pulmonary inflammation and COPD-like disease in mice. Adam15 deficiency in CD8+ T cells was required for the exaggerated pulmonary inflammation and COPD-like disease in CS-exposed Adam15-/- mice (as assessed by genetically deleting CD8+ T cells in Adam15-/- mice). Adam15 deficiency increased pulmonary inflammation by rendering CD8+ T cells and macrophages resistant to CS-induced activation of the mitochondrial apoptosis pathway by preserving mTOR signaling and intracellular Mcl-1 levels in these cells. These results strongly link ADAM15 deficiency to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Duo Zhang
- Program in Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, 30901, USA,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Takahiro Nakajima
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Katherine H. Walker
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA,Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Yuhong Li
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Quynh-Anh Fucci
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
27
|
Thomas C, Nothaft H, Yadav R, Fodor C, Alemka A, Oni O, Bell M, Rada B, Szymanski CM. Characterization of ecotin homologs from Campylobacter rectus and Campylobacter showae. PLoS One 2020; 15:e0244031. [PMID: 33378351 PMCID: PMC7773321 DOI: 10.1371/journal.pone.0244031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022] Open
Abstract
Ecotin, first described in Escherichia coli, is a potent
inhibitor of a broad range of serine proteases including those typically
released by the innate immune system such as neutrophil elastase (NE). Here we
describe the identification of ecotin orthologs in various
Campylobacter species, including Campylobacter
rectus and Campylobacter showae residing in the
oral cavity and implicated in the development and progression of periodontal
disease in humans. To investigate the function of these ecotins in
vitro, the orthologs from C.
rectus and C. showae were
recombinantly expressed and purified from E.
coli. Using CmeA degradation/protection assays,
fluorescence resonance energy transfer and NE activity assays, we found that
ecotins from C. rectus and C.
showae inhibit NE, factor Xa and trypsin, but not the
Campylobacter jejuni serine protease HtrA or its ortholog
in E. coli, DegP. To further evaluate ecotin
function in vivo, an E. coli
ecotin-deficient mutant was complemented with the C.
rectus and C. showae
homologs. Using a neutrophil killing assay, we demonstrate that the low survival
rate of the E. coli ecotin-deficient mutant
can be rescued upon expression of ecotins from C.
rectus and C. showae. In
addition, the C. rectus and
C. showae ecotins partially compensate for
loss of N-glycosylation and increased protease susceptibility in the related
pathogen, Campylobacter jejuni, thus implicating a similar role
for these proteins in the native host to cope with the protease-rich environment
of the oral cavity.
Collapse
Affiliation(s)
- Cody Thomas
- Department of Microbiology and Complex Carbohydrate Research Center,
University of Georgia, Athens, Georgia, United States of
America
| | - Harald Nothaft
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Ruchi Yadav
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Christopher Fodor
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Abofu Alemka
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Oluwadamilola Oni
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Michael Bell
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Christine M. Szymanski
- Department of Microbiology and Complex Carbohydrate Research Center,
University of Georgia, Athens, Georgia, United States of
America
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
- * E-mail:
| |
Collapse
|
28
|
Roe K. An explanation of the pathogenesis of several autoimmune diseases in immuno-compromised individuals. Scand J Immunol 2020; 93:e12994. [PMID: 33151588 DOI: 10.1111/sji.12994] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Some pathogen infections and immune system deficiencies have been linked to a few autoimmune diseases. However, the pathogenesis of most autoimmune diseases is unknown. An explanatory hypothesis for the pathogenesis of infection-initiated autoimmune diseases is provided. Virulent pathogen infections create extensive pathogen antigens that frequently require antibodies. These antibodies create extensive antigen-antibody immune complexes, which some immuno-compromised individuals will not adequately eliminate. This will cause inflammatory type III hypersensitivity symptoms, including protease releases that destroy epithelium, mesothelium and endothelium basement membranes, express new immunogenic antigens from previously sequestered basement membrane constituents, and ultimately induce new autoantibodies. This can continue after the infection ends, if the first wave of protease attacks on basement membranes induces new autoantibodies that cause new uncleared antigen-antibody immune complexes and type III hypersensitivity reactions. The secreted proteases and other enzymes will have preferred substrates and these proteases or other enzymes by themselves, or by their processed protein substrates, can express immunogenic antigens that induce new autoantibodies and initiate various autoimmune diseases. In summary, several autoimmune diseases can be initiated in immuno-compromised individuals during extensive pathogen infections, if these individuals have two immune problems: (a) slow or weak initial immune responses that result in a reliance on antibodies and (b) an inability to eliminate the resulting antigen-antibody immune complexes by phagocytosis. These two immune problems and the resulting immune system type III hypersensitivity reaction can explain the causation of several autoimmune diseases, including the most common and the rarest autoimmune diseases, both their differences and their similarities.
Collapse
|
29
|
The role of neutrophils in innate immunity-driven nonalcoholic steatohepatitis: lessons learned and future promise. Hepatol Int 2020; 14:652-666. [PMID: 32880077 DOI: 10.1007/s12072-020-10081-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
The enrichment of innate immune cells and the enhanced inflammation represent the hallmark of non-alcoholic steatohepatitis (NASH), the advanced subtype with a significantly increased risk of progression to end-stage liver diseases within the spectrum of non-alcoholic fatty liver disease. Neutrophils are traditionally recognized as key components in the innate immune system to defend against pathogens. Recently, a growing body of evidence supports neutrophils as emerging key player in mediating the transition from steatosis to NASH, which is largely inspired by the histological findings in human liver biopsy indicating the enhanced infiltration of neutrophils as one of the key histological features of NASH. In this review, we discuss data regarding histological perspectives of hepatic infiltration of neutrophils in NASH. We also highlight the pathophysiological role of neutrophils in promoting metabolic inflammation in the liver through the release of a vast array of granule proteins, the interaction with other pro-inflammatory immune cells, and the formation of neutrophil extracellular traps. Neutrophil granule proteins possess pleiotropic effects on regulating neutrophil biology and functions. A variety of granule proteins (including lipocalin-2, myeloperoxidase, proteinase 3, neutrophil elastase, etc.) produced by neutrophils enhance liver metabolic inflammation, thereby promoting NASH progression by mediating neutrophil-macrophage interaction. Therapeutically, pharmacological inhibitors targeting neutrophil granule proteins hold promise to combat NASH. In addition, this article also summarizes potentials of neutrophils and its derived various granule proteins for the accurate, even non-invasive diagnosis of NASH.
Collapse
|
30
|
Wang X, Zhang D, Higham A, Wolosianka S, Gai X, Zhou L, Petersen H, Pinto-Plata V, Divo M, Silverman EK, Celli B, Singh D, Sun Y, Owen CA. ADAM15 expression is increased in lung CD8 + T cells, macrophages, and bronchial epithelial cells in patients with COPD and is inversely related to airflow obstruction. Respir Res 2020; 21:188. [PMID: 32677970 PMCID: PMC7364636 DOI: 10.1186/s12931-020-01446-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A disintegrin and metalloproteinase domain-15 (ADAM15) is expressed by activated leukocytes, and fibroblasts in vitro. Whether ADAM15 expression is increased in the lungs of COPD patients is not known. METHODS ADAM15 gene expression and/or protein levels were measured in whole lung and bronchoalveolar lavage (BAL) macrophage samples obtained from COPD patients, smokers, and non-smokers. Soluble ADAM15 protein levels were measured in BAL fluid (BALF) and plasma samples from COPD patients and controls. Cells expressing ADAM15 in the lungs were identified using immunostaining. Staining for ADAM15 in different cells in the lungs was related to forced expiratory volume in 1 s (FEV1), ratio of FEV1 to forced vital capacity (FEV1/FVC), and pack-years of smoking history. RESULTS ADAM15 gene expression and/or protein levels were increased in alveolar macrophages and whole lung samples from COPD patients versus smokers and non-smokers. Soluble ADAM15 protein levels were similar in BALF and plasma samples from COPD patients and controls. ADAM15 immunostaining was increased in macrophages, CD8+ T cells, epithelial cells, and airway α-smooth muscle (α-SMA)-positive cells in the lungs of COPD patients. ADAM15 immunostaining in macrophages, CD8+ T cells and bronchial (but not alveolar) epithelial cells was related inversely to FEV1 and FEV1/FVC, but not to pack-years of smoking history. ADAM15 staining levels in airway α-SMA-positive cells was directly related to FEV1/FVC. Over-expressing ADAM15 in THP-1 cells reduced their release of matrix metalloproteinases and CCL2. CONCLUSIONS These results link increased ADAM15 expression especially in lung leukocytes and bronchial epithelial cells to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Duo Zhang
- Program in Clinical and Experimental Therapeutics, Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, 30901, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Andrew Higham
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Sophie Wolosianka
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Xiaoyan Gai
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Lu Zhou
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Hans Petersen
- The Lovelace Respiratory Research Institute, Albuquerque, NM, 87108, USA
| | - Victor Pinto-Plata
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Edwin K Silverman
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Bartolome Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dave Singh
- Medicines Evaluation Unit, University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Yongchang Sun
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Caroline A Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- The Lovelace Respiratory Research Institute, Albuquerque, NM, 87108, USA.
| |
Collapse
|
31
|
Cao T, Teng Z, Zheng L, Qian J, Ma H, Wang J, Qin W, Guo H. Activity-based ratiometric fluorescent small-molecule probe for endogenously monitoring neutrophil elastase in living cells. Anal Chim Acta 2020; 1127:295-302. [PMID: 32800135 DOI: 10.1016/j.aca.2020.06.070] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/06/2020] [Accepted: 06/28/2020] [Indexed: 11/15/2022]
Abstract
Neutrophil elastase (NE), a representative protease which is closely related to many diseases, acts an indispensable role in inflammatory diseases and clinical medicine. In this work, one activity-based non-peptide ratiometric fluorescent probe DCDF was designed with pentafluoropropionic anhydride as identification group. To our knowledge, this is the first probe capable of detecting NE in ratio. After the addition of the NE, the emission spectrum of DCDF has obvious bathochromic-shift phenomenon, and there is large Stokes shifts of ∼60 nm. Compared to only a few reported NE probes, DCDF is sensitive and selective and has very low detection limit (0-14 μg/mL, DL = 30.8 ng/mL). A possible response mechanism was proposed and verified by HPLC and HRMS spectra. What's more, DCDF is capable of endogenous recognition imaging in biological cells without interference from other enzymes under the ratio signal. A549 and HeLa cells were used for endogenous cell imaging experiments of NE and the feasibility of DCDF for the specific detection of NE in cells was proved. This experimental result makes probe DCDF a very promising tool for the clinical diagnosis and treatment of NE related diseases.
Collapse
Affiliation(s)
- Ting Cao
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Zhidong Teng
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province, 730046, PR China
| | - Lei Zheng
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Jing Qian
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Hong Ma
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Jiemin Wang
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Wenwu Qin
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Special Fund Project of Guiding Scientific and Technological Innovation Development of Gansu Province (2019ZX-04), Key Laboratory of Special Function Materials and Structure Design and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China.
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province, 730046, PR China.
| |
Collapse
|
32
|
Guo-Parke H, Linden D, Weldon S, Kidney JC, Taggart CC. Mechanisms of Virus-Induced Airway Immunity Dysfunction in the Pathogenesis of COPD Disease, Progression, and Exacerbation. Front Immunol 2020; 11:1205. [PMID: 32655557 PMCID: PMC7325903 DOI: 10.3389/fimmu.2020.01205] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/14/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the integrated form of chronic obstructive bronchitis and pulmonary emphysema, characterized by persistent small airway inflammation and progressive irreversible airflow limitation. COPD is characterized by acute pulmonary exacerbations and associated accelerated lung function decline, hospitalization, readmission and an increased risk of mortality, leading to huge social-economic burdens. Recent evidence suggests ~50% of COPD acute exacerbations are connected with a range of respiratory viral infections. Nevertheless, respiratory viral infections have been linked to the severity and frequency of exacerbations and virus-induced secondary bacterial infections often result in a synergistic decline of lung function and longer hospitalization. Here, we review current advances in understanding the cellular and molecular mechanisms underlying the pathogenesis of COPD and the increased susceptibility to virus-induced exacerbations and associated immune dysfunction in patients with COPD. The multiple immune regulators and inflammatory signaling pathways known to be involved in host-virus responses are discussed. As respiratory viruses primarily target airway epithelial cells, virus-induced inflammatory responses in airway epithelium are of particular focus. Targeting virus-induced inflammatory pathways in airway epithelial cells such as Toll like receptors (TLRs), interferons, inflammasomes, or direct blockade of virus entry and replication may represent attractive future therapeutic targets with improved efficacy. Elucidation of the cellular and molecular mechanisms of virus infections in COPD pathogenesis will undoubtedly facilitate the development of these potential novel therapies that may attenuate the relentless progression of this heterogeneous and complex disease and reduce morbidity and mortality.
Collapse
Affiliation(s)
- Hong Guo-Parke
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Dermot Linden
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Sinéad Weldon
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Joseph C Kidney
- Department of Respiratory Medicine Mater Hospital Belfast, Belfast, United Kingdom
| | - Clifford C Taggart
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Sciences, Queens University Belfast, Belfast, United Kingdom
| |
Collapse
|
33
|
De Cunto G, Cavarra E, Bartalesi B, Lucattelli M, Lungarella G. Innate Immunity and Cell Surface Receptors in the Pathogenesis of COPD: Insights from Mouse Smoking Models. Int J Chron Obstruct Pulmon Dis 2020; 15:1143-1154. [PMID: 32547002 PMCID: PMC7246326 DOI: 10.2147/copd.s246219] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is mainly associated with smoking habit. Inflammation is the major initiating process whereby neutrophils and monocytes are attracted into the lung microenvironment by external stimuli present in tobacco leaves and in cigarette smoke, which promote chemotaxis, adhesion, phagocytosis, release of superoxide anions and enzyme granule contents. A minority of smokers develops COPD and different molecular factors, which contribute to the onset of the disease, have been put forward. After many years of research, the pathogenesis of COPD is still an object of debate. In vivo models of cigarette smoke-induced COPD may help to unravel cellular and molecular mechanisms underlying the pathogenesis of COPD. The mouse represents the most favored animal choice with regard to the study of immune mechanisms due to its genetic and physiological similarities to humans, the availability of a large variability of inbred strains, the presence in the species of several genetic disorders analogous to those in man, and finally on the possibility to create models “made-to-measure” by genetic manipulation. The review outlines the different response of mouse strains to cigarette smoke used in COPD studies while retaining a strong focus on their relatability to human patients. These studies reveal the importance of innate immunity and cell surface receptors in the pathogenesis of pulmonary injury induced by cigarette smoking. They further advance the way in which we use wild type or genetically manipulated strains to improve our overall understanding of a multifaceted disease such as COPD. The structural and functional features, which have been found in the different strains of mice after chronic exposure to cigarette smoke, can be used in preclinical studies to develop effective new therapeutic agents for the different phenotypes in human COPD.
Collapse
Affiliation(s)
- Giovanna De Cunto
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Eleonora Cavarra
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Barbara Bartalesi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Monica Lucattelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giuseppe Lungarella
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
34
|
Liu SY, Yan AM, Guo WYZ, Fang YY, Dong QJ, Li RR, Ni SN, Sun Y, Yang WC, Yang GF. Human Neutrophil Elastase Activated Fluorescent Probe for Pulmonary Diseases Based on Fluorescence Resonance Energy Transfer Using CdSe/ZnS Quantum Dots. ACS NANO 2020; 14:4244-4254. [PMID: 32208668 DOI: 10.1021/acsnano.9b09493] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
There is an increasing demand for effective noninvasive diagnosis against common pulmonary diseases, which are rising sharply due to the serious air pollution. Human neutrophil elastase (HNE), a typical protease highly involved in pulmonary inflammatory diseases and lung cancer, is a potential predictor for disease progression. Currently, few of the HNE-targeting probes are applicable in vivo due to the limitation in sensitivity and biocompatibility. Herein, we reported the achievement of in vitro detection and in vivo imaging of HNE by incorporating the HNE-specific peptide substrate, quantum dots (QDs), and organic dyes into the fluorescence resonance energy transfer (FRET) system. The refined nanoprobe, termed QDP, could specifically measure the HNE with excellent sensitivity of 7.15 pM in aqueous solution and successfully image the endogenous and exogenous HNE in living cells. In addition, this nanoprobe enabled HNE imaging in mouse models of lung cancer and acute lung injury, and the HNE activity at high temporal and spatial resolution was continuously monitored. Most importantly, QDP successfully discriminated the serums of patients with lung diseases from those of the healthy controls based on the HNE activity determination. Overall, this study demonstrates the advantages of a FRET-system-based nanoprobe in imaging performance and provides an applicable tool for in vivo HNE detection and pulmonary disease diagnosis.
Collapse
Affiliation(s)
- Shi-Yu Liu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Ai-Min Yan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Wu Ying-Zheng Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yuan-Yuan Fang
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, P.R. China
| | - Qing-Jian Dong
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, P.R. China
| | - Rong-Rong Li
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Sheng-Nan Ni
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yao Sun
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Wen-Chao Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 30071, P.R. China
| |
Collapse
|
35
|
Rojas-Quintero J, Wang X, Owen CA. Endothelial Cell Death in Emphysema: More Sugarcoating Needed? Am J Respir Crit Care Med 2020; 200:1078-1080. [PMID: 31298928 PMCID: PMC6888664 DOI: 10.1164/rccm.201906-1254ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Joselyn Rojas-Quintero
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital/Harvard Medical SchoolBoston, Massachusetts
| | - Xiaoyun Wang
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital/Harvard Medical SchoolBoston, Massachusetts
| | - Caroline A Owen
- Division of Pulmonary and Critical Care MedicineBrigham and Women's Hospital/Harvard Medical SchoolBoston, Massachusetts
| |
Collapse
|
36
|
Stockley RA. Alpha-1 Antitrypsin Deficiency: Have We Got the Right Proteinase? CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:163-171. [PMID: 32396717 DOI: 10.15326/jcopdf.7.3.2019.0151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alpha-1 antitrypsin deficiency (AATD) has traditionally been associated with the development of early onset panlobular emphysema thought to reflect the direct interstitial damage caused by neutrophil elastase. Since this enzyme is highly sensitive to irreversible inhibition by alpha-1 antitrypsin (AAT), the logic of intravenous augmentation therapy has remained unquestioned and efficacy is supported by both observational studies and formal clinical trials. However, evidence suggests that although AAT augmentation modulates the progression of emphysema, it only slows it down. This raises the issue of whether our long-held beliefs of the cause of the susceptibility to develop emphysema in deficient individuals are correct. There are several aspects of our understanding of the disease that might benefit from a radical departure from traditional thought. This review addresses these concepts and alternative pathways that may be central to progression of emphysema.
Collapse
Affiliation(s)
- Robert A Stockley
- Lung Investigation Unit, University Hospitals, Birmingham National Health Service Foundation Trust, Queen Elizabeth Hospital, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
37
|
Liu H, Lessieur EM, Saadane A, Lindstrom SI, Taylor PR, Kern TS. Neutrophil elastase contributes to the pathological vascular permeability characteristic of diabetic retinopathy. Diabetologia 2019; 62:2365-2374. [PMID: 31612267 PMCID: PMC6866660 DOI: 10.1007/s00125-019-04998-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Levels of neutrophil elastase, a serine protease secreted by neutrophils, are elevated in diabetes. The purpose of this study was to determine whether neutrophil elastase (NE) contributes to the diabetes-induced increase in retinal vascular permeability in mice with streptozotocin-induced diabetes, and, if so, to investigate the potential role of IL-17 in this process. METHODS In vivo, diabetes was induced in neutrophil elastase-deficient (Elane-/-), Il-17a-/- and wild-type mice. After 8 months of diabetes, Elane-/- mice and wild-type age-matched control mice were injected with FITC-BSA. Fluorescence microscopy was used to assess leakage of FITC-BSA from the retinal vasculature into the neural retina. The level of NE in Il-17a-/- diabetic retina and sera were determined by ELISA. In vitro, the effect of NE on the permeability and viability of human retinal endothelial cells and the expression of junction proteins and adhesion molecules were studied. RESULTS Eight months of diabetes resulted in increased retinal vascular permeability and levels of NE in retina and plasma of wild-type animals. All of these abnormalities were significantly inhibited in mice lacking the elastase. The diabetes-induced increase in NE was inhibited in mice lacking IL-17. In vitro, NE increased retinal endothelial cell permeability, which was partially inhibited by a myeloid differentiation primary response 88 (MyD88) inhibitor, NF-κB inhibitor, and protease-activated receptor (PAR)2 inhibitor. NE degraded vascular endothelial-cadherin (VE-cadherin) in a concentration-dependent manner. CONCLUSIONS/INTERPRETATION IL-17 regulates NE expression in diabetes. NE contributes to vascular leakage in diabetic retinopathy, partially through activation of MyD88, NF-κB and PAR2 and degradation of VE-cadherin.
Collapse
Affiliation(s)
- Haitao Liu
- Department of Biology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People's Republic of China
| | - Emma M Lessieur
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, School of Medicine, University of California-Irvine, 829 Health Sciences Rd. Gillespie Neuroscience Research Facility, Room 2107, Irvine, CA, 92697, USA
| | - Aicha Saadane
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, School of Medicine, University of California-Irvine, 829 Health Sciences Rd. Gillespie Neuroscience Research Facility, Room 2107, Irvine, CA, 92697, USA
| | - Sarah I Lindstrom
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Patricia R Taylor
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
- Veterans Administration Medical Center Research Service 151, Cleveland, OH, USA
| | - Timothy S Kern
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, School of Medicine, University of California-Irvine, 829 Health Sciences Rd. Gillespie Neuroscience Research Facility, Room 2107, Irvine, CA, 92697, USA.
- Veterans Administration Medical Center Research Service 151, Cleveland, OH, USA.
- Veterans Administration Medical Center Research Service, Long Beach, CA, USA.
| |
Collapse
|
38
|
Castellani S, D'Oria S, Diana A, Polizzi AM, Di Gioia S, Mariggiò MA, Guerra L, Favia M, Vinella A, Leonetti G, De Venuto D, Gallo C, Montemurro P, Conese M. G-CSF and GM-CSF Modify Neutrophil Functions at Concentrations found in Cystic Fibrosis. Sci Rep 2019; 9:12937. [PMID: 31506515 PMCID: PMC6736848 DOI: 10.1038/s41598-019-49419-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023] Open
Abstract
The role of colony stimulating factors (CSFs) in cystic fibrosis (CF) circulating neutrophils has not been thoroughly evaluated, considering that the neutrophil burden of lung inflammation in these subjects is very high. The aim of this study was to assess granulocyte-CSF (G-CSF) and granulocyte-macrophage-CSF (GM-CSF) levels in CF patients in various clinical conditions and how these cytokines impact on activation and priming of neutrophils. G-CSF and GM-CSF levels were measured in sputum and serum samples of stable CF patients (n = 21) and in CF patients with acute exacerbation before and after a course of antibiotic therapy (n = 19). CSFs were tested on non CF neutrophils to investigate their effects on reactive oxygen species (ROS) production, degranulation (CD66b, elastase, lactoferrin, MMP-9), and chemotaxis. At very low concentrations found in CF patients (0.005-0.1 ng/ml), both cytokines inhibited ROS production, while higher concentrations (1-5 ng/ml) exerted a stimulatory effect. While either CSF induced elastase and MMP-9 secretion, lactoferrin levels were increased only by G-CSF. Chemotaxis was inhibited by GM-CSF, but was increased by G-CSF. However, when present together at low concentrations, CSFs increased basal and fMLP-stimulated ROS production and chemotaxis. These results suggest the CSF levels that circulating neutrophils face before extravasating into the lungs of CF patients may enhance their function contributing to the airway damage.
Collapse
Affiliation(s)
- Stefano Castellani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Susanna D'Oria
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Anna Diana
- Department of Biomedical Sciences and Human Oncology, Section of Medical Genetics, University of Bari, Bari, Italy
| | - Angela Maria Polizzi
- Department of Biomedical Sciences and Human Oncology, Section of Medical Genetics, University of Bari, Bari, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Maria Addolorata Mariggiò
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Maria Favia
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Angela Vinella
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy
| | - Giuseppina Leonetti
- Cystic Fibrosis Regional Center, Department of Biomedical and Human Oncology, Pediatrics Section, U.O. "B. Trambusti", Policlinico, University of Bari, Bari, Italy
| | - Domenica De Venuto
- Cystic Fibrosis Regional Center, Department of Biomedical and Human Oncology, Pediatrics Section, U.O. "B. Trambusti", Policlinico, University of Bari, Bari, Italy
| | - Crescenzio Gallo
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Pasqualina Montemurro
- Department of Biomedical Sciences and Human Oncology, Section of General Pathology, University of Bari, Bari, Italy.
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
39
|
Cho JL, Medoff BD. Chronic Obstructive Pulmonary Disease: Back to the Basics. Am J Respir Crit Care Med 2019; 198:1241-1242. [PMID: 30011221 DOI: 10.1164/rccm.201806-1148ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Josalyn L Cho
- 1 Division of Pulmonary, Critical Care and Occupational Medicine University of Iowa Iowa City, Iowa and
| | - Benjamin D Medoff
- 2 Division of Pulmonary and Critical Care Medicine Massachusetts General Hospital Boston, Massachusetts
| |
Collapse
|
40
|
van Eeden SF, Hogg JC. Immune-Modulation in Chronic Obstructive Pulmonary Disease: Current Concepts and Future Strategies. Respiration 2019; 99:550-565. [PMID: 31480060 DOI: 10.1159/000502261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by the chronic inhalation of toxic particles and gases that are primarily but not exclusively derived from cigarette smoke that may be either actively or passively inhaled, which initiates a persistent innate and adaptive immune response in the lung. This immune response is associated with an aberrant tissue repair and remodeling process that results in varying degrees of chronic inflammation with excess production of mucus in the central airways and permanent destruction of the smaller conducting airways and gas exchanging surface in the peripheral lung. Currently, the primary aims of treatment in COPD are bronchodilation (inhaled short- and long-acting β-agonist and antimuscarinic therapies), to control symptoms and nonspecific broad-acting anti-inflammatory agents (inhaled and oral corticosteroids, phosphor-di-esterase inhibitors, and macrolides). That provide symptomatic relief but have little or no impact on either disease progression or mortality. As our understanding of the immune pathogenesis of the COPD improves, available immune modulation therapies have the potential to alter or interfere with damaging immune pathways, thereby slowing relentless progression of lung tissue destruction. The purpose of this brief review is to discuss our current understanding of the immune pathogenesis of both the airways and parenchymal injury as well as the dysfunctional tissue repair process to propose immune modulating interventions in an attempt to stabilize or return these pathological changes to their normal state. The ultimate goal of the immune modulation therapy is to improve both morbidity and mortality associated with COPD.
Collapse
Affiliation(s)
- Stephan F van Eeden
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada, .,Pacific Lung Health Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada,
| | - James C Hogg
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
41
|
Chronic Obstructive Pulmonary Disease and Lung Cancer: Underlying Pathophysiology and New Therapeutic Modalities. Drugs 2019; 78:1717-1740. [PMID: 30392114 DOI: 10.1007/s40265-018-1001-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are major lung diseases affecting millions worldwide. Both diseases have links to cigarette smoking and exert a considerable societal burden. People suffering from COPD are at higher risk of developing lung cancer than those without, and are more susceptible to poor outcomes after diagnosis and treatment. Lung cancer and COPD are closely associated, possibly sharing common traits such as an underlying genetic predisposition, epithelial and endothelial cell plasticity, dysfunctional inflammatory mechanisms including the deposition of excessive extracellular matrix, angiogenesis, susceptibility to DNA damage and cellular mutagenesis. In fact, COPD could be the driving factor for lung cancer, providing a conducive environment that propagates its evolution. In the early stages of smoking, body defences provide a combative immune/oxidative response and DNA repair mechanisms are likely to subdue these changes to a certain extent; however, in patients with COPD with lung cancer the consequences could be devastating, potentially contributing to slower postoperative recovery after lung resection and increased resistance to radiotherapy and chemotherapy. Vital to the development of new-targeted therapies is an in-depth understanding of various molecular mechanisms that are associated with both pathologies. In this comprehensive review, we provide a detailed overview of possible underlying factors that link COPD and lung cancer, and current therapeutic advances from both human and preclinical animal models that can effectively mitigate this unholy relationship.
Collapse
|
42
|
Ling Z, Xu F, Edwards JV, Prevost NT, Nam S, Condon BD, French AD. Nanocellulose as a colorimetric biosensor for effective and facile detection of human neutrophil elastase. Carbohydr Polym 2019; 216:360-368. [DOI: 10.1016/j.carbpol.2019.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/18/2019] [Accepted: 04/05/2019] [Indexed: 12/30/2022]
|
43
|
Guerra M, Frey D, Hagner M, Dittrich S, Paulsen M, Mall MA, Schultz C. Cathepsin G Activity as a New Marker for Detecting Airway Inflammation by Microscopy and Flow Cytometry. ACS CENTRAL SCIENCE 2019; 5:539-548. [PMID: 30937381 PMCID: PMC6439450 DOI: 10.1021/acscentsci.8b00933] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Indexed: 06/01/2023]
Abstract
Muco-obstructive lung diseases feature extensive bronchiectasis due to the uncontrolled release of neutrophil serine proteases into the airways. To assess if cathepsin G (CG) is a novel key player in chronic lung inflammation, we developed membrane-bound (mSAM) and soluble (sSAM) FRET reporters. The probes quantitatively revealed elevated CG activity in samples from 46 patients. For future basic science and personalized clinical applications, we developed a rapid, highly informative, and easily translatable small-molecule FRET flow cytometry assay for monitoring protease activity including cathepsin G. We demonstrated that mSAM distinguished healthy from patient cells by FRET-based flow cytometry with excellent correlation to confocal microscopy data.
Collapse
Affiliation(s)
- Matteo Guerra
- Molecular
Medicine Partnership Unit (MMPU), European
Molecular Biology Laboratory (EMBL) and University of Heidelberg, 69117 Heidelberg, Germany
- Faculty
of Biosciences, Collaboration for Joint
Ph.D. Degree between EMBL and Heidelberg University, 69117 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Dario Frey
- Department
of Translational Pulmonology, University
of Heidelberg, 69120 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Matthias Hagner
- Department
of Translational Pulmonology, University
of Heidelberg, 69120 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Susanne Dittrich
- Department
of Translational Pulmonology, University
of Heidelberg, 69120 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Michelle Paulsen
- Department
of Translational Pulmonology, University
of Heidelberg, 69120 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Marcus A. Mall
- Department
of Translational Pulmonology, University
of Heidelberg, 69120 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department
of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Berlin Institute
of Health (BIH), 10178 Berlin, Germany
| | - Carsten Schultz
- Molecular
Medicine Partnership Unit (MMPU), European
Molecular Biology Laboratory (EMBL) and University of Heidelberg, 69117 Heidelberg, Germany
- Translational
Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department
of Physiology and Pharmacology, Oregon Health
& Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239-3098, United States
| |
Collapse
|
44
|
Linden D, Guo-Parke H, Coyle PV, Fairley D, McAuley DF, Taggart CC, Kidney J. Respiratory viral infection: a potential "missing link" in the pathogenesis of COPD. Eur Respir Rev 2019; 28:28/151/180063. [PMID: 30872396 DOI: 10.1183/16000617.0063-2018] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the third most common cause of global mortality. Acute exacerbations of COPD frequently necessitate hospital admission to enable more intensive therapy, incurring significant healthcare costs. COPD exacerbations are also associated with accelerated lung function decline and increased risk of mortality. Until recently, bacterial pathogens were believed to be responsible for the majority of disease exacerbations. However, with the advent of culture-independent molecular diagnostic techniques it is now estimated that viruses are detected during half of all COPD exacerbations and are associated with poorer clinical outcomes. Human rhinovirus, respiratory syncytial virus and influenza are the most commonly detected viruses during exacerbation. The role of persistent viral infection (adenovirus) has also been postulated as a potential pathogenic mechanism in COPD. Viral pathogens may play an important role in driving COPD progression by acting as triggers for exacerbation and subsequent lung function decline whilst the role of chronic viral infection remains a plausible hypothesis that requires further evaluation. There are currently no effective antiviral strategies for patients with COPD. Herein, we focus on the current understanding of the cellular and molecular mechanisms of respiratory viral infection in COPD.
Collapse
Affiliation(s)
- Dermot Linden
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Hong Guo-Parke
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Peter V Coyle
- The Regional Virus Laboratory, Belfast Trust, Belfast, UK
| | - Derek Fairley
- The Regional Virus Laboratory, Belfast Trust, Belfast, UK
| | - Danny F McAuley
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Clifford C Taggart
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Joe Kidney
- Dept of Respiratory Medicine, Mater Hospital Belfast, Belfast, UK
| |
Collapse
|
45
|
Rønnow SR, Langholm LL, Sand JMB, Thorlacius-Ussing J, Leeming DJ, Manon-Jensen T, Tal-Singer R, Miller BE, Karsdal MA, Vestbo J. Specific elastin degradation products are associated with poor outcome in the ECLIPSE COPD cohort. Sci Rep 2019; 9:4064. [PMID: 30858579 PMCID: PMC6412140 DOI: 10.1038/s41598-019-40785-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a slow heterogeneous progression. Therefore, improved biomarkers that can accurately identify patients with the highest likelihood of progression and therefore the ability to benefit from a given treatment, are needed. Elastin is an essential structural protein of the lungs. In this study, we investigated whether elastin degradation products generated by the enzymes proteinase 3, cathepsin G, neutrophil elastase, MMP7 or MMP9/12 were prognostic biomarkers for COPD-related outcomes. The elastin degradome was assessed in a subpopulation (n = 1307) of the Evaluation of COPD Longitudinally to Identify Predictive Surrogate End-points (ECLIPSE) cohort with 3 years of clinical follow-up. Elastin degraded by proteinase 3 could distinguish between COPD participants and non-smoking controls (p = 0.0006). A total of 30 participants (3%) died over the 3 years of observation. After adjusting for confounders, plasma levels of elastin degraded by proteinase 3 and cathepsin G were independently associated with mortality outcome with a hazard ratio per 1 SD of 1.49 (95%CI 1.24-1.80, p < 0.0001) and 1.31 (95%CI 1.10-1.57, p = 0.0029), respectively. Assessing the elastin degradome demonstrated that specific elastin degradation fragments have potential utility as biomarkers identifying subtypes of COPD patients at risk of poor prognosis and supports further exploration in confirmatory studies.
Collapse
Affiliation(s)
- Sarah Rank Rønnow
- Nordic Bioscience A/S, Herlev, Denmark. .,University of Southern Denmark, The Faculty of Health Science, Odense, Denmark.
| | - Lasse Løcke Langholm
- Nordic Bioscience A/S, Herlev, Denmark.,University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | - Jørgen Vestbo
- Division of Infection Immunity and Respiratory Medicine, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, England
| |
Collapse
|
46
|
Higham A, Quinn AM, Cançado JED, Singh D. The pathology of small airways disease in COPD: historical aspects and future directions. Respir Res 2019; 20:49. [PMID: 30832670 PMCID: PMC6399904 DOI: 10.1186/s12931-019-1017-y] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 02/25/2019] [Indexed: 12/16/2022] Open
Abstract
Small airways disease (SAD) is a cardinal feature of chronic obstructive pulmonary disease (COPD) first recognized in the nineteenth century. The diverse histopathological features associated with SAD underpin the heterogeneous nature of COPD. Our understanding of the key molecular mechanisms which drive the pathological changes are not complete. In this article we will provide a historical overview of key histopathological studies which have helped shape our understanding of SAD and discuss the hallmark features of airway remodelling, mucous plugging and inflammation. We focus on the relationship between SAD and emphysema, SAD in the early stages of COPD, and the mechanisms which cause SAD progression, including bacterial colonization and exacerbations. We discuss the need to specifically target SAD to attenuate the progression of COPD.
Collapse
Affiliation(s)
- Andrew Higham
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK.
| | - Anne Marie Quinn
- Department of Histopathology, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Dave Singh
- The University of Manchester Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK.,Medicines Evaluation Unit, The Langley Building, Southmoor Road, Manchester, UK
| |
Collapse
|
47
|
Liu SY, Xiong H, Li RR, Yang WC, Yang GF. Activity-Based Near-Infrared Fluorogenic Probe for Enabling in Vitro and in Vivo Profiling of Neutrophil Elastase. Anal Chem 2019; 91:3877-3884. [DOI: 10.1021/acs.analchem.8b04455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Shi-Yu Liu
- Key Laboratory
of Pesticide and Chemical Biology of Ministry of Education, International
Joint Research Center for Intelligent Biosensor Technology and Health,
and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, People’s Republic of China
| | - Hao Xiong
- Key Laboratory
of Pesticide and Chemical Biology of Ministry of Education, International
Joint Research Center for Intelligent Biosensor Technology and Health,
and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, People’s Republic of China
| | - Rong-Rong Li
- Key Laboratory
of Pesticide and Chemical Biology of Ministry of Education, International
Joint Research Center for Intelligent Biosensor Technology and Health,
and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, People’s Republic of China
| | - Wen-Chao Yang
- Key Laboratory
of Pesticide and Chemical Biology of Ministry of Education, International
Joint Research Center for Intelligent Biosensor Technology and Health,
and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, People’s Republic of China
| | - Guang-Fu Yang
- Key Laboratory
of Pesticide and Chemical Biology of Ministry of Education, International
Joint Research Center for Intelligent Biosensor Technology and Health,
and Chemical Biology Center, College of Chemistry, Central China Normal University, Wuhan 430079, People’s Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 30071, People’s Republic of China
| |
Collapse
|
48
|
Kim DK, Eun KM, Kim MK, Cho D, Han SA, Han SY, Seo Y, Lee DH, Cho SH, Kim DW. Comparison Between Signature Cytokines of Nasal Tissues in Subtypes of Chronic Rhinosinusitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2019; 11:201-211. [PMID: 30661312 PMCID: PMC6340796 DOI: 10.4168/aair.2019.11.2.201] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Endotype in chronic rhinosinusitis (CRS) has been established in the last decade. However, the exact immunologic profile of CRS still has controversy because it has a considerable immunologic heterogeneity. Therefore, we investigated various inflammatory mediators according to different nasal tissues in chronic rhinosinusitis and compared them within the same subject. METHODS We collected uncinate process mucosa (UP) and nasal polyp (NP) tissues from controls, CRS without NP (CRSsNP) and CRS with NP (CRSwNP). Expression levels of 28 inflammatory mediators including T helper (Th) 1, Th2, Th17, proinflammatory cytokines and remodeling markers were determined by multiplex immunoassay and were analyzed using paired tests as well as principal component analysis (PCA) to investigate endotype in each subtype of CRS. RESULTS Signature inflammatory mediators are interleukin (IL)-5, C-C motif chemokine ligand (CCL)-24, monocyte chemoattractant protein (MCP)-4, and vascular cell adhesion molecule (VCAM)-1 in eosinophilic NP, whereas IL-17A, IL-1β, and matrix metallopeptidase (MMP)-9 were detected as signature inflammatory markers in non-eosinophilic NP. Despite differences in inflammatory cytokine profile between eosinophilic and non-eosinophilic NP, the common upregulation of IL-5, CCL-11, IL-23, IL-2Rα, VCAM-1, MMP-3 and MMP-9 were shown in NP compared to UP within the same subject. In the PCA, we observed that Th2 immune response was helpful in discriminating between nasal tissues in subtypes of CRS and that there was a partial overlap between non-eosinophilic NP and eosinophilic NP in terms of Th2 mediators. CONCLUSIONS Commonly upregulated mediators in NP were Th2-associated, compared with UP regardless of CRS subtypes, whereas signature markers were distinct in each NP subtype. These findings imply that Th2 inflammatory responses may play a role in the development of NP regardless of CRSwNP subtypes.
Collapse
Affiliation(s)
- Dong Kyu Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital and Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea.,Clinical Mucosal Immunology Study Group, Seoul, Korea
| | - Kyoung Mi Eun
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Min Kyung Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Deuktae Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Sun A Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Yoon Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Yuju Seo
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Han Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Ho Cho
- Division of Allergy-Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Dae Woo Kim
- Clinical Mucosal Immunology Study Group, Seoul, Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
49
|
Dong J, Liao W, Tan LH, Yong A, Peh WY, Wong WSF. Gene silencing of receptor-interacting protein 2 protects against cigarette smoke-induced acute lung injury. Pharmacol Res 2019; 139:560-568. [PMID: 30394320 DOI: 10.1016/j.phrs.2018.10.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Chronic obstructive pulmonary disease (COPD) is characterized by progressive alveolar damage and generally irreversible airflow limitation. Nuclear factor-κB (NF-κB) plays a critical role in COPD pathogenesis. Receptor-interacting protein 2 (Rip2), a 60 kDa adaptor protein, is a positive regulator of NF-κB pathway and also an inducible transcriptional product of NF-κB activation. We sought to investigate if Rip2 gene silencing could protect against cigarette smoke (CS)-induced acute lung injury. EXPERIMENTAL APPROACH Gene silencing efficacy of Rip2 siRNA was characterized in mouse macrophage and mouse lung epithelial cell lines, and in a CS-induced acute lung injury mouse model. Bronchoalveolar lavage (BAL) fluid cell counts, levels of pro-inflammatory and oxidative damage markers, lung section inflammatory and epithelium thickness scorings, and nuclear NF-κB translocation were measured. KEY RESULTS CS was found to upregulate Rip2 level in mouse lungs. Rip2 siRNA was able to suppress Rip2 levels in both macrophage and lung epithelial cell lines and in mouse lungs, block CS extract (CSE)-induced mediator release by the cultured cells, and abate neutrophil counts in BAL fluid from CS-challenged mice. Rip2 siRNA suppressed CS-induced inflammatory and oxidative damage markers, and nuclear p65 accumulation and transcriptional activation in lung tissues. Besides, Rip2 siRNA was able to disrupt CSE-induced NF-κB activation in a NF-κB reporter gene assay. CONCLUSIONS AND IMPLICATIONS Taken together, we report for the first time that Rip2 gene silencing ameliorated CS-induced acute lung injury probably via disruption of the NF-κB activity, postulating that Rip2 may be a novel therapeutic target for COPD.
Collapse
Affiliation(s)
- Jinrui Dong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Lay Hong Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Amy Yong
- Department of Pharmacology and Therapeutics, Faculty of Life Sciences and Medicine, King's College London, UK
| | - Wen Yan Peh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Immunology Program, Life Science Institute, National University of Singapore, Singapore; Singapore-HUJ Alliance for Research and Enterprise, Molecular Mechanisms of Inflammatory Diseases Interdisciplinary Research Group, Singapore.
| |
Collapse
|
50
|
Zhou L, Le Y, Tian J, Yang X, Jin R, Gai X, Sun Y. Cigarette smoke-induced RANKL expression enhances MMP-9 production by alveolar macrophages. Int J Chron Obstruct Pulmon Dis 2018; 14:81-91. [PMID: 30587964 PMCID: PMC6304243 DOI: 10.2147/copd.s190023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background and purpose Cigarette smoke (CS) induces alveolar destruction through overproduction of proteinases including matrix metalloproteinase (MMP)-9 by alveolar macrophages (AMs). Receptor activator of nuclear factor-κB ligand (RANKL) functions in immune regulation and cytokine secretion; whether it is involved in CS-induced MMP-9 expression is unknown. The purpose of our study was to investigate the expression and functional role of RANKL pathway in MMP-9 production pertaining to the pathogenesis of COPD. Materials and methods We first localized RANKL and its receptor RANK in the lungs of mice exposed to long-term CS exposure. Next, we studied RANKL and RANK expression under CS extract (CSE) stimulation in vitro. Lastly, we studied the in vitro biological function of RANKL in CS-induced production of MMP-9. Results Both RANKL and RANK were highly expressed in AMs in CS-exposed mice, but not in the control mice. In vitro, CSE increased the expressions of RANKL and RANK in macrophages. AMs responded to CSE and RANKL stimulation by overexpressing MMP-9, and CSE-induced MMP-9 expression was partly blocked by using monoclonal anti-RANKL antibody. Conclusion RANKL/RANK pathway mediates CS-induced MMP-9 expression in AMs, suggesting a novel mechanism for CS-associated emphysema.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Yanqing Le
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Jieyu Tian
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoyan Gai
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| | - Yongchang Sun
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China,
| |
Collapse
|