1
|
Rani A, Stadler JT, Marsche G. HDL-based therapeutics: A promising frontier in combating viral and bacterial infections. Pharmacol Ther 2024; 260:108684. [PMID: 38964560 DOI: 10.1016/j.pharmthera.2024.108684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Low levels of high-density lipoprotein (HDL) and impaired HDL functionality have been consistently associated with increased susceptibility to infection and its serious consequences. This has been attributed to the critical role of HDL in maintaining cellular lipid homeostasis, which is essential for the proper functioning of immune and structural cells. HDL, a multifunctional particle, exerts pleiotropic effects in host defense against pathogens. It functions as a natural nanoparticle, capable of sequestering and neutralizing potentially harmful substances like bacterial lipopolysaccharides. HDL possesses antiviral activity, preventing viruses from entering or fusing with host cells, thereby halting their replication cycle. Understanding the complex relationship between HDL and the immune system may reveal innovative targets for developing new treatments to combat infectious diseases and improve patient outcomes. This review aims to emphasize the role of HDL in influencing the course of bacterial and viral infections and its and its therapeutic potential.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Styria, Austria.
| |
Collapse
|
2
|
Zhang Y, Jiang Z, Chen L, Lei T, Zheng X. Repurposing lipid-lowering drugs on asthma and lung function: evidence from a genetic association analysis. J Transl Med 2024; 22:615. [PMID: 38961500 PMCID: PMC11223406 DOI: 10.1186/s12967-024-05359-5] [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: 12/09/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE To explore the correlation between asthma risk and genetic variants affecting the expression or function of lipid-lowering drug targets. METHODS We conducted Mendelian randomization (MR) analyses using variants in several genes associated with lipid-lowering medication targets: HMGCR (statin target), PCSK9 (alirocumab target), NPC1L1 (ezetimibe target), APOB (mipomersen target), ANGPTL3 (evinacumab target), PPARA (fenofibrate target), and APOC3 (volanesorsen target), as well as LDLR and LPL. Our objective was to investigate the relationship between lipid-lowering drugs and asthma through MR. Finally, we assessed the efficacy and stability of the MR analysis using the MR Egger and inverse variance weighted (IVW) methods. RESULTS The elevated triglyceride (TG) levels associated with the APOC3, and LPL targets were found to increase asthma risk. Conversely, higher LDL-C levels driven by LDLR were found to decrease asthma risk. Additionally, LDL-C levels (driven by APOB, NPC1L1 and HMGCR targets) and TG levels (driven by the LPL target) were associated with improved lung function (FEV1/FVC). LDL-C levels driven by PCSK9 were associated with decreased lung function (FEV1/FVC). CONCLUSION In conclusion, our findings suggest a likely causal relationship between asthma and lipid-lowering drugs. Moreover, there is compelling evidence indicating that lipid-lowering therapies could play a crucial role in the future management of asthma.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Zichao Jiang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Hunan, 410008, China
| | - Lingli Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Hunan, 410008, China.
| | - Ting Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Hunan, 410008, China.
| | - Xiangrong Zheng
- Department of Pediatrics, Xiangya Hospital, Central South University, Hunan, 410008, China.
| |
Collapse
|
3
|
Wen J, Zhuang R, He Q, Wei C, Giri M, Chi J. Association between serum lipid and all-cause mortality in asthmatic populations: a cohort study. Lipids Health Dis 2024; 23:189. [PMID: 38907251 PMCID: PMC11191228 DOI: 10.1186/s12944-024-02179-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/05/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Presently, the majority of investigations primarily evaluate the association between lipid profiles and asthma. However, few investigations explore the connection between lipids and mortality related to the disease. This study aims to explore the association of serum lipids with all-cause mortality within asthmatic adults. METHODS The investigation included 3233 eligible patients with asthma from the NHANES (2011-2018). The potential associations were explored using three Cox proportional hazards models, restricted cubic splines (RCS), threshold effect models, and CoxBoost models. In addition, subgroup analyses were conducted to investigate these associations within distinct populations. RESULTS After controlling all covariables, the Cox proportional hazards model proved a 17% decrease in the probability of death for each increased unit of low-density lipoprotein-cholesterol (LDL-C) (mmol/L). Yet, there was no association seen between blood high-density lipoprotein cholesterol (HDL-C), total cholesterol, or triglyceride and all-cause mortality in asthmatics. The application of RCS and threshold effect models verified an inverse and linear association of LDL-C with all-cause mortality. According to the results from the CoxBoost model, LDL-C exhibited the most substantial impact on the follow-up status of asthmatics among the serum lipids. CONCLUSION Our investigation concluded that in American asthmatic populations, LDL-C levels were inversely and linearly correlated with mortality. However, no independent relationship was found between triglycerides, total cholesterol, or HDL-C and mortality.
Collapse
Affiliation(s)
- Jun Wen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Rongjuan Zhuang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Qingliu He
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Urology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengcheng Wei
- Department of Urology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mohan Giri
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jing Chi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Guo Y, Liu Q, Zheng Z, Qing M, Yao T, Wang B, Zhou M, Wang D, Ke Q, Ma J, Shan Z, Chen W. Genetic association of inflammatory marker GlycA with lung function and respiratory diseases. Nat Commun 2024; 15:3751. [PMID: 38704398 PMCID: PMC11069551 DOI: 10.1038/s41467-024-47845-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
Association of circulating glycoprotein acetyls (GlycA), a systemic inflammation biomarker, with lung function and respiratory diseases remain to be investigated. We examined the genetic correlation, shared genetics, and potential causality of GlycA (N = 115,078) with lung function and respiratory diseases (N = 497,000). GlycA showed significant genetic correlation with FEV1 (rg = -0.14), FVC (rg = -0.18), asthma (rg = 0.21) and COPD (rg = 0.31). We consistently identified ten shared loci (including chr3p21.31 and chr8p23.1) at both SNP and gene level revealing potential shared biological mechanisms involving ubiquitination, immune response, Wnt/β-catenin signaling, cell growth and differentiation in tissues or cells including blood, epithelium, fibroblast, fetal thymus, and fetal intestine. Genetically elevated GlycA was significantly correlated with lung function and asthma susceptibility (354.13 ml decrement of FEV1, 442.28 ml decrement of FVC, and 144% increased risk of asthma per SD increment of GlycA) from MR analyses. Our findings provide insights into biological mechanisms of GlycA in relating to lung function, asthma, and COPD.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Department of Epidemiology, Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA.
| | - Quanhong Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhilin Zheng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Mengxia Qing
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Tianci Yao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Min Zhou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Dongming Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Qinmei Ke
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jixuan Ma
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhilei Shan
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
5
|
Yao X, Kaler M, Qu X, Kalidhindi RSR, Sviridov D, Dasseux A, Barr E, Keeran K, Jeffries KR, Yu ZX, Gao M, Gordon S, Barochia AV, Mills J, Shahid S, Weir NA, Kalchiem-Dekel O, Theard P, Playford MP, Stylianou M, Fitzgerald W, Remaley AT, Levine SJ. Asthmatic patients with high serum amyloid A have proinflammatory HDL: Implications for augmented systemic and airway inflammation. J Allergy Clin Immunol 2024; 153:1010-1024.e14. [PMID: 38092139 PMCID: PMC10999351 DOI: 10.1016/j.jaci.2023.11.917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/26/2023] [Accepted: 11/10/2023] [Indexed: 01/15/2024]
Abstract
RATIONALE Serum amyloid A (SAA) is bound to high-density lipoproteins (HDL) in blood. Although SAA is increased in the blood of patients with asthma, it is not known whether this modifies asthma severity. OBJECTIVE We sought to define the clinical characteristics of patients with asthma who have high SAA levels and assess whether HDL from SAA-high patients with asthma is proinflammatory. METHODS SAA levels in serum from subjects with and without asthma were quantified by ELISA. HDLs isolated from subjects with asthma and high SAA levels were used to stimulate human monocytes and were intravenously administered to BALB/c mice. RESULTS An SAA level greater than or equal to 108.8 μg/mL was defined as the threshold to identify 11% of an asthmatic cohort (n = 146) as being SAA-high. SAA-high patients with asthma were characterized by increased serum C-reactive protein, IL-6, and TNF-α; older age; and an increased prevalence of obesity and severe asthma. HDL isolated from SAA-high patients with asthma (SAA-high HDL) had an increased content of SAA as compared with HDL from SAA-low patients with asthma and induced the secretion of IL-6, IL-1β, and TNF-α from human monocytes via a formyl peptide receptor 2/ATP/P2X purinoceptor 7 axis. Intravenous administration to mice of SAA-high HDL, but not normal HDL, induced systemic inflammation and amplified allergen-induced neutrophilic airway inflammation and goblet cell metaplasia. CONCLUSIONS SAA-high patients with asthma are characterized by systemic inflammation, older age, and an increased prevalence of obesity and severe asthma. HDL from SAA-high patients with asthma is proinflammatory and, when intravenously administered to mice, induces systemic inflammation, and amplifies allergen-induced neutrophilic airway inflammation. This suggests that systemic inflammation induced by SAA-high HDL may augment disease severity in asthma.
Collapse
Affiliation(s)
- Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Xuan Qu
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | | | - Denis Sviridov
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Amaury Dasseux
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Eric Barr
- Animal Surgery and Resources Core Facility, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Karen Keeran
- Animal Surgery and Resources Core Facility, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Kenneth R Jeffries
- Animal Surgery and Resources Core Facility, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Zu-Xi Yu
- Pathology Core Facility, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Meixia Gao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Scott Gordon
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Joni Mills
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Shahid Shahid
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Nargues A Weir
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Or Kalchiem-Dekel
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Patricia Theard
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Martin P Playford
- Section on Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Mario Stylianou
- Office of Biostatistics Research, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Wendy Fitzgerald
- Section on Intercellular Interactions, National Institute of Child Health and Development, National Institutes of Health, Bethesda, Md
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, National Heart, Lung, and Blood Institute, Bethesda, Md
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, Md.
| |
Collapse
|
6
|
Feng J, Huang Y, Huang M, Li X, Amoah K, Huang Y, Jian J. Apolipoprotein Eb (On-ApoEb) protects Oreochromis niloticus against Streptococcus agalactiae infection. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109069. [PMID: 37696347 DOI: 10.1016/j.fsi.2023.109069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Apolipoprotein E (ApoE), a critical targeting protein, has been found to play an essential role in the protection against infection and inflammation. However, the immune functions of ApoE against bacterial infection in fish have not been investigated. In this study, a full-length cDNA for ApoE, named On-ApoEb was cloned from Oreochromis niloticus. The predicted cDNA sequence was 831bp in length and coded for a protein of 276 amino acid residues, which shared 63.87%-98.55% identity with ApoEb from other fishes, and about 22% identity with ApoEb from mammals. On-ApoEb from O. niloticus was highly expressed in the liver and could be activated in the tissues (liver, spleen, brain, and intestine) after infection with Streptococcus agalactiae. Moreover, the results revealed that On-ApoEb could decrease the expression levels of pro-inflammatory factors, immune-related pathways, and apoptosis, while increasing the expression levels of anti-inflammatory factors. Furthermore, On-ApoEb was noted to improve the survival rate and reduce the bacterial load in the liver and spleen. These results suggested that On-ApoEb was connected with immune response and had anti-inflammation and anti-apoptosis activities.
Collapse
Affiliation(s)
- Jiamin Feng
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Yongxiong Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Meiling Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Xing Li
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Kwaku Amoah
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China
| | - Yu Huang
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| | - Jichang Jian
- Fisheries College of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture & Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institutes, Zhanjiang, 524088, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, China.
| |
Collapse
|
7
|
Shen Y, Li L, Chen W, Li Q, Xu Y, He F, Wang C, Tian Z, Chen Y, Yang Y. Apolipoprotein E negatively regulates allergic airway inflammation and remodeling in mice with OVA-induced chronic asthma. Int Immunopharmacol 2023; 116:109776. [PMID: 36731155 DOI: 10.1016/j.intimp.2023.109776] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023]
Abstract
Apolipoprotein E (ApoE) is a corticosteroid-unresponsive gene that negatively regulates ovalbumin (OVA) -induced allergic airway inflammation in mice with acute asthma. However, whether ApoE negatively regulates airway remodeling in mice with OVA-induced chronic asthma remains unknown. This study aimed to investigate the effects of ApoE on OVA-induced chronic asthma in a murine model. ApoE knockout (ApoE-/-) and wild-type (WT) mice were sensitized and challenged with OVA for 10 weeks to establish the chronic asthma model. Compared with WT mice, the results demonstrated that ApoE deficiency exacerbated OVA-induced airway inflammation, including elevated numbers of inflammatory cells in the blood and bronchoalveolar lavage fluid (BALF), as well as increased T helper type 2 (Th2) cells in lung tissue, Th2 cytokines in BALF, and total IgE levels in plasma. Importantly, ApoE deficiency aggravated OVA-induced airway remodeling, as evidenced by higher plasma transforming growth factor (TGF)-β1 levels, airway goblet cell hyperplasia, and collagen deposition compared with WT mice. These results revealed that ApoE deficiency aggravates airway remodeling and inflammation in mice with OVA-induced chronic allergic asthma.
Collapse
Affiliation(s)
- Yunqin Shen
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Lingjie Li
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Wushi Chen
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Qin Li
- Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Yixuan Xu
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Fang He
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Caixia Wang
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Zezhong Tian
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China
| | - Yanqiu Chen
- Department of Otolaryngology, Guangzhou Women and Children Medical Centre, Guangzhou, China
| | - Yan Yang
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China; Guangdong Engineering Technology Research Center of Nutrition Translation, Guangzhou, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China.
| |
Collapse
|
8
|
Matz J, Farra YM, Cotto HM, Bellini C, Oakes JM. Respiratory mechanics following chronic cigarette smoke exposure in the Apoe
−
/
−
mouse model. Biomech Model Mechanobiol 2023; 22:233-252. [PMID: 36335185 DOI: 10.1007/s10237-022-01644-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022]
Abstract
Even though cigarette smoking (CS) has been on the decline over the past 50 years, it is still the leading cause of preventable premature death in the United States. Preclinical models have investigated the cardiopulmonary effects of CS exposure (CSE), but the structure-function relationship in the respiratory system has not yet been fully explored. To evaluate these relationships, we exposed female apolipoprotein E-deficient (Apoe− / − ) mice to mainstream CS (n = 8 ) for 5 days/week over 24 weeks with room air as a control (AE,n = 8 ). To contextualize the impact of CSE, we also assessed the natural aging effects over 24 weeks of air exposure (baseline,n = 8 ). Functional assessments were performed on a small animal mechanical ventilator (flexiVent, SCIREQ), where pressure-volume curves and impedance data at four levels of positive end-expiratory pressure (P peep and with increasing doses of methacholine were collected. Constant phase model parameters (R N : Newtonian resistance, H: coefficient of tissue elastance, and G: coefficient of tissue resistance) were calculated from the impedance data. Perfusion fixed-left lung tissue was utilized for quantification of parenchyma airspace size and tissue thickness, airway wall thickness, and measurements of elastin, cytoplasm + nucleus, fibrin, and collagen content for the parenchyma and airways. Aging caused the lung to become more compliant, with an upward-leftward shift of the pressure-volume curve and a reduction in all constant phase model parameters. This was supported by larger parenchyma airspace sizes, with a reduction in cell cytoplasm + nucleus area. Airway walls became thinner, even though low-density collagen content increased. In contrast, CSE caused a downward-rightward shift of the pressure-volume curve along with an increase in H, G, and hysteresivity (η = G / H ). Organ stiffening was accompanied by enhanced airway hyper-responsiveness following methacholine challenge. Structurally, parenchyma airspaces enlarged, as indicated by an increase in equivalent airspace diameter (D 0 ), and the septum thickened with significant deposition of low-density collagen along with an influx of cells. Airway walls thickened due to deposition of both high and low-density collagen, infiltration of cells, and epithelial cell elongation. In all, our data suggest that CSE in female Apoe− / − mice reduces respiratory functionality and causes morphological alterations in both central and peripheral airways that results in lung stiffening, compared to AE controls.
Collapse
Affiliation(s)
- Jacqueline Matz
- Department of Bioengineering, Northeastern University, Boston, USA
| | - Yasmeen M Farra
- Department of Bioengineering, Northeastern University, Boston, USA
| | | | - Chiara Bellini
- Department of Bioengineering, Northeastern University, Boston, USA
| | - Jessica M Oakes
- Department of Bioengineering, Northeastern University, Boston, USA.
| |
Collapse
|
9
|
Liang S, Zhou Z, Zhou Z, Liang J, Lin W, Zhang C, Zhou C, Zhao H, Meng X, Zou F, Yu C, Cai S. Blockade of CBX4-mediated β-catenin SUMOylation attenuates airway epithelial barrier dysfunction in asthma. Int Immunopharmacol 2022; 113:109333. [DOI: 10.1016/j.intimp.2022.109333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
|
10
|
Xu MM, Kang JY, Ji S, Wei YY, Wei SL, Ye JJ, Wang YG, Shen JL, Wu HM, Fei GH. Melatonin Suppresses Macrophage M1 Polarization and ROS-Mediated Pyroptosis via Activating ApoE/LDLR Pathway in Influenza A-Induced Acute Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2520348. [PMID: 36425057 PMCID: PMC9681554 DOI: 10.1155/2022/2520348] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2023]
Abstract
Influenza virus infection is one of the strongest pathogenic factors for the development of acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS). However, the underlying cellular and molecular mechanisms have not been clarified. In this study, we aim to investigate whether melatonin modulates macrophage polarization, oxidative stress, and pyroptosis via activating Apolipoprotein E/low-density lipoprotein receptor (ApoE/LDLR) pathway in influenza A-induced ALI. Here, wild-type (WT) and ApoE-/- mice were instilled intratracheally with influenza A (H3N2) and injected intraperitoneally with melatonin for 7 consecutive days. In vitro, WT and ApoE-/- murine bone marrow-derived macrophages (BMDMs) were pretreated with melatonin before H3N2 stimulation. The results showed that melatonin administration significantly attenuated H3N2-induced pulmonary damage, leukocyte infiltration, and edema; decreased the expression of proinflammatory M1 markers; enhanced anti-inflammatory M2 markers; and switched the polarization of alveolar macrophages (AMs) from M1 to M2 phenotype. Additionally, melatonin inhibited reactive oxygen species- (ROS-) mediated pyroptosis shown by downregulation of malonaldehyde (MDA) and ROS levels as well as inhibition of the NLRP3/GSDMD pathway and lactate dehydrogenase (LDH) release. Strikingly, the ApoE/LDLR pathway was activated when melatonin was applied in H3N2-infected macrophages and mice. ApoE knockout mostly abrogated the protective impacts of melatonin on H3N2-induced ALI and its regulatory ability on macrophage polarization, oxidative stress, and pyroptosis. Furthermore, recombinant ApoE3 (re-ApoE3) inhibited H3N2-induced M1 polarization of BMDMs with upregulation of MT1 and MT2 expression, but re-ApoE2 and re-ApoE4 failed to do this. Melatonin combined with re-ApoE3 played more beneficial protective effects on modulating macrophage polarization, oxidative stress, and pyroptosis in H3N2-infected ApoE-/- BMDMs. Our study indicated that melatonin attenuated influenza A- (H3N2-) induced ALI by inhibiting the M1 polarization of pulmonary macrophages and ROS-mediated pyroptosis via activating the ApoE/LDLR pathway. This study suggested that melatonin-ApoE/LDLR axis may serve as a novel therapeutic strategy for influenza virus-induced ALI.
Collapse
Affiliation(s)
- Meng-Meng Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Jia-Ying Kang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Shuang Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Yuan-Yuan Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Si-Liang Wei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Jing-Jing Ye
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Yue-Guo Wang
- Department of Emergency Critical Care Medicine, First Affiliated Hospital of Anhui Provincial Hospital, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, 230001 Anhui, China
| | - Ji-Long Shen
- Provincial Laboratory of Microbiology and Parasitology of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Hui-Mei Wu
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Anhui Geriatric Institute, Department of Geriatric Respiratory Critical and Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| | - Guang-He Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
- Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui, China
| |
Collapse
|
11
|
Ghosh S, Rihan M, Ahmed S, Pande AH, Sharma SS. Immunomodulatory potential of apolipoproteins and their mimetic peptides in asthma: Current perspective. Respir Med 2022; 204:107007. [DOI: 10.1016/j.rmed.2022.107007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/03/2022] [Indexed: 10/31/2022]
|
12
|
Zhu Y, Esnault S, Ge Y, Jarjour NN, Brasier AR. Airway fibrin formation cascade in allergic asthma exacerbation: implications for inflammation and remodeling. Clin Proteomics 2022; 19:15. [PMID: 35590254 PMCID: PMC9117591 DOI: 10.1186/s12014-022-09351-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Airway remodeling in patients with asthma, which leads to a decline in pulmonary function, is likely the result of repeated exacerbations often provoked by aeroallergen exposures. Aeroallegen exposure triggers a stereotypic response orchestrated by growth factor cytokines and other protein mediators. This results in a late-phase allergic reaction characterized by vascular permeability, recruitment of activated leukocytes, and activation of structural cells of the airway. The spectrum of protein mediators and their functions are incompletely understood. METHODS Bronchoalveolar lavage fluid (BALF) samples were obtained from 12 volunteers who exhibited robust eosinophilic recruitment following segmental bronchial provocation with allergen (SBP-Ag). We systematically identified and quantified proteins in BALF using high-performance liquid chromatography-high-resolution mass spectrometry (LC-MS/MS) followed by pathway analysis and correlations with airway physiology. RESULTS Pairwise analysis of protein abundance in BALF pre- vs post-SBP-Ag revealed that 55 proteins were upregulated and 103 proteins were downregulated. We observed enrichment of groups of proteins mapping to hemostasis/fibrin clot, platelet activation, lipoprotein assembly, neutrophil degranulation proteins, and acute-phase inflammation-airway remodeling pathways. The abundances of F2 and Fibrinogen γ (FGG) correlated with eosinophil numbers, whereas SERPINA3 negatively correlated with change in FeNO. The coagulation proteins F2 and KNG negatively correlated with FN1 an index of airway remodeling. Interestingly, patients with lower FEV1 showed distinct allergen-induced patterns of 8 BALF proteins, including MUC1, alarmins (HSPB1), and actin polymerization factors. CONCLUSIONS Protein abundance of the fibrin formation cascade, platelet activation and remodeling are associated with late-phase leukocyte numbers and markers of remodeling. Patients with lower FEV1 have distinct dynamic responses to allergen.
Collapse
Affiliation(s)
- Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Stephane Esnault
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Nizar N Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, 53705, USA
| | - Allan R Brasier
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, 715 Highland Ave, Madison, WI, 53705, USA.
| |
Collapse
|
13
|
ADAMTS7 Attenuates House Dust Mite-Induced Airway Inflammation and Th2 Immune Responses. Lung 2022; 200:305-313. [PMID: 35503474 PMCID: PMC9205806 DOI: 10.1007/s00408-022-00538-x] [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: 03/02/2022] [Accepted: 04/15/2022] [Indexed: 10/30/2022]
Abstract
PURPOSE ADAMTS7 is a secreted metalloproteinase enzyme and proteoglycan associated with the early progression of coronary artery disease. However, there is limited information regarding the role of ADAMTS7 in lung adaptive immunity and inflammation. Thus, we sought to assess whether ADAMTS7 expression in the lung modulates house dust mite (HDM)-induced airway inflammation and Th2 immune response. METHODS The role of ADAMTS7 in HDM-induced airway disease was assessed in ADAMTS7-deficient (ADAMTS7-/-) mice and compared with the wild-type control mice by flow cytometry, ELISA, and histopathology. Furthermore, the antigen priming capability of dendritic cells (DC) was determined ex vivo by employing coculture with CD4+ OT-II cells. RESULTS ADAMTS7-/- mice develop an augmented eosinophilic airway inflammation, mucous cell metaplasia, and increased Th2 immune response to inhaled HDM. In addition, allergen uptake by lung DC and migration to draining mediastinal lymph node were significantly increased in ADAMTS7-/- mice, which shows an enhanced capacity to mount allergen-specific T-cell proliferation and effector Th2 cytokine productions. We propose that the mechanism by which ADAMTS7 negatively regulates DC function involves attenuated antigen uptake and presentation capabilities, which reduces allergic sensitization and Th2 immune responses in the lung. CONCLUSION In aggregate, we provide compelling evidence that ADAMTS7 plays a pivotal role in allergic airway disease and Th2 immunity and would be an attractive target for asthma.
Collapse
|
14
|
Bangert C, Villazala-Merino S, Fahrenberger M, Krausgruber T, Bauer WM, Stanek V, Campion NJ, Bartosik T, Quint T, Regelsberger G, Niederberger-Leppin V, Bock C, Schneider S, Eckl-Dorna J. Comprehensive Analysis of Nasal Polyps Reveals a More Pronounced Type 2 Transcriptomic Profile of Epithelial Cells and Mast Cells in Aspirin-Exacerbated Respiratory Disease. Front Immunol 2022; 13:850494. [PMID: 35418991 PMCID: PMC8996080 DOI: 10.3389/fimmu.2022.850494] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
Chronic rhinosinusitis with nasal polyps is affecting up to 3% of Western populations. About 10% of patients with nasal polyps also suffer from asthma and intolerance to aspirin, a syndrome called aspirin-exacerbated respiratory disease. Although eosinophilic inflammation is predominant in polyps of both diseases, phenotypic differences in the tissue-derived microenvironment, elucidating disease-specific characteristics, have not yet been identified. We sought to obtain detailed information about phenotypic and transcriptional differences in epithelial and immune cells in polyps of aspirin-tolerant and intolerant patients. Cytokine profiles in nasal secretions and serum of patients suffering from aspirin-exacerbated respiratory disease (n = 10) or chronic rhinosinusitis with nasal polyps (n = 9) were assessed using a multiplex mesoscale discovery assay. After enrichment for immune cell subsets by flow cytometry, we performed transcriptomic profiling by employing single-cell RNA sequencing. Aspirin-intolerant patients displayed significantly elevated IL-5 and CCL17 levels in nasal secretions corresponding to a more pronounced eosinophilic type 2 inflammation. Transcriptomic profiling revealed that epithelial and mast cells not only complement one another in terms of gene expression associated with the 15-lipoxygenase pathway but also show a clear type 2-associated inflammatory phenotype as identified by the upregulation of POSTN, CCL26, and IL13 in patients with aspirin-exacerbated respiratory disease. Interestingly, we also observed cellular stress responses indicated by an increase of MTRNR2L12, MTRNR2L8, and NEAT1 across all immune cell subsets in this disease entity. In conclusion, our findings support the hypothesis that epithelial and mast cells act in concert as potential drivers of the pathogenesis of the aspirin-exacerbated respiratory disease.
Collapse
Affiliation(s)
- Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Martin Fahrenberger
- Center for Integrative Bioinformatics Vienna (CIBIV), Max Perutz Labs, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Victoria Stanek
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | | | - Tina Bartosik
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Tamara Quint
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Guenther Regelsberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | | | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Institute of Artificial Intelligence and Decision Support, Center for Medical Statistics, Informatics, and Intelligence Systems, Medical University of Vienna, Vienna, Austria
| | - Sven Schneider
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Lewis BW, Ford ML, Khan AQ, Walum J, Britt RD. Chronic Allergen Challenge Induces Corticosteroid Insensitivity With Persistent Airway Remodeling and Type 2 Inflammation. Front Pharmacol 2022; 13:855247. [PMID: 35479312 PMCID: PMC9035517 DOI: 10.3389/fphar.2022.855247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2-high severe asthma is described as a distinct endotype with Th2 inflammation, high eosinophil lung infiltration, impaired lung function, and reduced corticosteroid sensitivity. While the inflammatory milieu is similar to mild asthma, patients with type 2-high severe asthma likely have underlying mechanisms that sustain asthma pathophysiology despite corticosteroid treatments. Acute and chronic allergen models induce robust type 2 inflammatory responses, however differences in corticosteroid sensitivity remains poorly understood. In the present study, we sensitized and challenged mice with ovalbumin (OVA; acute model) or mixed allergens (MA; chronic model). Corticosteroid sensitivity was assessed by administering vehicle, 1, or 3 mg/kg fluticasone propionate (FP) and examining key asthmatic features such as airway inflammation, remodeling, hyperresponsiveness, and antioxidant capacity. Both acute and chronic allergen exposure exhibited enhanced AHR, immune cell infiltration, airway inflammation, and remodeling, but corticosteroids were unable to fully alleviate inflammation, AHR, and airway smooth muscle mass in MA-challenged mice. While there were no differences in antioxidant capacity, persistent IL-4+ Th2 cell population suggests the MA model induces type 2 inflammation that is insensitive to corticosteroids. Our data indicate that chronic allergen exposure is associated with more persistent type 2 immune responses and corticosteroid insensitivity. Understanding differences between acute and chronic allergen models could unlock underlying mechanisms related to type 2-high severe asthma.
Collapse
Affiliation(s)
- Brandon W. Lewis
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Maria L. Ford
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Aiman Q. Khan
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Joshua Walum
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Rodney D. Britt
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- *Correspondence: Rodney D. Britt Jr,
| |
Collapse
|
16
|
Lewis BW, Amici SA, Kim HY, Shalosky EM, Khan AQ, Walum J, Gowdy KM, Englert JA, Porter NA, Grayson MH, Britt RD, Guerau-de-Arellano M. PRMT5 in T Cells Drives Th17 Responses, Mixed Granulocytic Inflammation, and Severe Allergic Airway Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1525-1533. [PMID: 35288471 PMCID: PMC9055570 DOI: 10.4049/jimmunol.2100994] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 01/13/2023]
Abstract
Severe asthma is characterized by steroid insensitivity and poor symptom control and is responsible for most asthma-related hospital costs. Therapeutic options remain limited, in part due to limited understanding of mechanisms driving severe asthma. Increased arginine methylation, catalyzed by protein arginine methyltransferases (PRMTs), is increased in human asthmatic lungs. In this study, we show that PRMT5 drives allergic airway inflammation in a mouse model reproducing multiple aspects of human severe asthma. We find that PRMT5 is required in CD4+ T cells for chronic steroid-insensitive severe lung inflammation, with selective T cell deletion of PRMT5 robustly suppressing eosinophilic and neutrophilic lung inflammation, pathology, airway remodeling, and hyperresponsiveness. Mechanistically, we observed high pulmonary sterol metabolic activity, retinoic acid-related orphan receptor γt (RORγt), and Th17 responses, with PRMT5-dependent increases in RORγt's agonist desmosterol. Our work demonstrates that T cell PRMT5 drives severe allergic lung inflammation and has potential implications for the pathogenesis and therapeutic targeting of severe asthma.
Collapse
Affiliation(s)
- Brandon W Lewis
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Stephanie A Amici
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, Columbus, OH
| | - Hye-Young Kim
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | - Emily M Shalosky
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH
| | - Aiman Q Khan
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Joshua Walum
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Kymberly M Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH
| | - Joshua A Englert
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN
| | - Mitchell H Grayson
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH.,Division of Allergy and Immunology, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Rodney D Britt
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH; .,Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Mireia Guerau-de-Arellano
- Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, Columbus, OH; .,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH; and.,Department of Neuroscience, The Ohio State University, Columbus, OH
| |
Collapse
|
17
|
Halayko AJ, Pascoe CD, Gereige JD, Peters MC, Cohen RT, Woodruff PG. Update in Adult Asthma 2020. Am J Respir Crit Care Med 2021; 204:395-402. [PMID: 34181860 DOI: 10.1164/rccm.202103-0552up] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Andrew J Halayko
- University of Manitoba, 8664, SECTION OF RESPIRATORY DISEASES, Winnipeg, Manitoba, Canada.,University of Manitoba, 8664, Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Christopher D Pascoe
- University of Manitoba, 8664, Physiology and Pathophysiology, Winnipeg, Manitoba, Canada.,University of Manitoba Children's Hospital Research Institute of Manitoba, 423136, Winnipeg, Manitoba, Canada
| | - Jessica D Gereige
- Boston University School of Medicine, 12259, Division of Pulmonary, Allergy, Sleep, and Critical Care Medicine, Department of Medicine, Boston, Massachusetts, United States
| | - Michael C Peters
- University of California San Francisco, 8785, Pulmonary and Critical Care, San Francisco, California, United States
| | - Robyn T Cohen
- Boston University School of Medicine, 12259, Pediatrics, Boston, Massachusetts, United States
| | - Prescott G Woodruff
- UCSF, 8785, Division of Pulmonary and Critical Care Medicine, Department of Medicine and CVRI, San Francisco, California, United States;
| |
Collapse
|
18
|
Reece SW, Varikuti S, Kilburg-Basnyat B, Dunigan-Russell K, Hodge MX, Luo B, Madenspacher JH, Thomas SY, Tokarz DA, Tighe RM, Cook DN, Fessler MB, Gowdy KM. Scavenger Receptor BI Attenuates IL-17A-Dependent Neutrophilic Inflammation in Asthma. Am J Respir Cell Mol Biol 2021; 64:698-708. [PMID: 33647226 PMCID: PMC8456883 DOI: 10.1165/rcmb.2020-0007oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/26/2021] [Indexed: 12/20/2022] Open
Abstract
Asthma is a common respiratory disease currently affecting more than 300 million worldwide and is characterized by airway inflammation, hyperreactivity, and remodeling. It is a heterogeneous disease consisting of corticosteroid-sensitive T-helper cell type 2-driven eosinophilic and corticosteroid-resistant, T-helper cell type 17-driven neutrophilic phenotypes. One pathway recently described to regulate asthma pathogenesis is cholesterol trafficking. Scavenger receptors, in particular SR-BI (scavenger receptor class B type I), are known to direct cellular cholesterol uptake and efflux. We recently defined SR-BI functions in pulmonary host defense; however, the function of SR-BI in asthma pathogenesis is unknown. To elucidate the role of SR-BI in allergic asthma, SR-BI-sufficient (SR-BI+/+) and SR-BI-deficient (SR-BI-/-) mice were sensitized (Days 0 and 7) and then challenged (Days 14, 15, and 16) with a house dust mite (HDM) preparation administered through oropharyngeal aspiration. Airway inflammation and cytokine production were quantified on Day 17. When compared with SR-BI+/+ mice, the HDM-challenged SR-BI-/- mice had increased neutrophils and pulmonary IL-17A production in BAL fluid. This augmented IL-17A production in SR-BI-/- mice originated from a non-T-cell source that included neutrophils and alveolar macrophages. Given that SR-BI regulates adrenal steroid hormone production, we tested whether the changes in SR-BI-/- mice were glucocorticoid dependent. Indeed, SR-BI-/- mice were adrenally insufficient during the HDM challenge, and corticosterone replacement decreased pulmonary neutrophilia and IL-17A production in SR-BI-/- mice. Taken together, these data indicate that SR-BI dampens pulmonary neutrophilic inflammation and IL-17A production in allergic asthma at least in part by maintaining adrenal function.
Collapse
Affiliation(s)
- Sky W. Reece
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Sanjay Varikuti
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Brita Kilburg-Basnyat
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Katelyn Dunigan-Russell
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Myles X. Hodge
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Bin Luo
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Jennifer H. Madenspacher
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Seddon Y. Thomas
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Debra A. Tokarz
- Center for Human Health and the Environment, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina; and
| | - Robert M. Tighe
- Department of Medicine, Duke University, Durham, North Carolina
| | - Donald N. Cook
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Michael B. Fessler
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Kymberly M. Gowdy
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
19
|
Busse PJ, McDonald VM, Wisnivesky JP, Gibson PG. Asthma Across the Ages: Adults. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 8:1828-1838. [PMID: 32499032 DOI: 10.1016/j.jaip.2020.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023]
Abstract
Asthma is a common disease affecting approximately 300 million people worldwide, across all age ranges. Despite advances in asthma outcomes of the last few decades, there remains room for improvement in asthma management and for patient outcomes, particularly in older patients. The heterogeneity of asthma is now well recognized, and is known to complicate response to treatment and patient behavior and impact health outcomes. Asthma and its heterogeneity change according to age. Asthma affects people differently across the life span. In adults, prevalence is highest among those in middle age; however, mortality is greater in the older age group. In this clinical commentary, we describe how age impacts asthma prevalence and incidence, outcomes, disease expression, and approach to management in adulthood and in older patients.
Collapse
Affiliation(s)
- Paula J Busse
- Division of Allergy and Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Vanessa M McDonald
- National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia; Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; Viruses, Immunology, Vaccines, Asthma (VIVA) Programme, Hunter Medical Research Institute, Newcastle, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Juan P Wisnivesky
- Division of General Internal Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peter G Gibson
- National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia; Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia; Viruses, Immunology, Vaccines, Asthma (VIVA) Programme, Hunter Medical Research Institute, Newcastle, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
20
|
Yao X, Levine SJ. The Long and Winding Road from GWAS to Obstructive Lung Disease: Is There a Role for LRP1? Am J Respir Cell Mol Biol 2021; 64:279-280. [PMID: 33352084 PMCID: PMC7909341 DOI: 10.1165/rcmb.2020-0567ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Xianglan Yao
- Laboratory of Asthma and Lung Inflammation National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda, Maryland
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda, Maryland
| |
Collapse
|
21
|
Kalchiem-Dekel O, Yao X, Barochia AV, Kaler M, Figueroa DM, Karkowsky WB, Gordon EM, Gao M, Fergusson MM, Qu X, Liu P, Li Y, Seifuddin F, Pirooznia M, Levine SJ. Apolipoprotein E Signals via TLR4 to Induce CXCL5 Secretion by Asthmatic Airway Epithelial Cells. Am J Respir Cell Mol Biol 2020; 63:185-197. [PMID: 32338995 DOI: 10.1165/rcmb.2019-0209oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The primary function of APOE (apolipoprotein E) is to mediate the transport of cholesterol- and lipid-containing lipoprotein particles into cells by receptor-mediated endocytosis. APOE also has pro- and antiinflammatory effects, which are both context and concentration dependent. For example, Apoe-/- mice exhibit enhanced airway remodeling and hyperreactivity in experimental asthma, whereas increased APOE levels in lung epithelial lining fluid induce IL-1β secretion from human asthmatic alveolar macrophages. However, APOE-mediated airway epithelial cell inflammatory responses and signaling pathways have not been defined. Here, RNA sequencing of human asthmatic bronchial brushing cells stimulated with APOE identified increased expression of mRNA transcripts encoding multiple proinflammatory genes, including CXCL5 (C-X-C motif chemokine ligand 5), an epithelial-derived chemokine that promotes neutrophil activation and chemotaxis. We subsequently characterized the APOE signaling pathway that induces CXCL5 secretion by human asthmatic small airway epithelial cells (SAECs). Neutralizing antibodies directed against TLR4 (Toll-like receptor 4), but not TLR2, attenuated APOE-mediated CXCL5 secretion by human asthmatic SAECs. Inhibition of TAK1 (transforming growth factor-β-activated kinase 1), IκKβ (inhibitor of nuclear factor κ B kinase subunit β), TPL2 (tumor progression locus 2), and JNK (c-Jun N-terminal kinase), but not p38 MAPK (mitogen-activated protein kinase) or MEK1/2 (MAPK kinase 1/2), attenuated APOE-mediated CXCL5 secretion. The roles of TAK1, IκKβ, TPL2, and JNK in APOE-mediated CXCL5 secretion were verified by RNA interference. Furthermore, RNA interference showed that after APOE stimulation, both NF-κB p65 and TPL2 were downstream of TAK1 and IκKβ, whereas JNK was downstream of TPL2. In summary, elevated levels of APOE in the airway may activate a TLR4/TAK1/IκKβ/NF-κB/TPL2/JNK signaling pathway that induces CXCL5 secretion by human asthmatic SAECs. These findings identify new roles for TLR4 and TPL2 in APOE-mediated proinflammatory responses in asthma.
Collapse
Affiliation(s)
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | | | | | - Meixia Gao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | | | - Xuan Qu
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch
| | - Poching Liu
- DNA Sequencing and Genomics Core Facility, and
| | - Yuesheng Li
- DNA Sequencing and Genomics Core Facility, and
| | - Fayaz Seifuddin
- Bioinformatics and Computational Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Mehdi Pirooznia
- Bioinformatics and Computational Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | |
Collapse
|
22
|
Zhao CC, Xu J, Xie QM, Fan XY, Fei GH, Wu HM. Apolipoprotein E negatively regulates murine allergic airway inflammation via suppressing the activation of NLRP3 inflammasome and oxidative stress. Int Immunopharmacol 2020; 81:106301. [PMID: 32062073 DOI: 10.1016/j.intimp.2020.106301] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/17/2020] [Accepted: 02/09/2020] [Indexed: 12/16/2022]
Abstract
Apolipoprotein E (ApoE) has been reported as a steroid unresponsive gene and functions as a negative regulator of airway hyperreactivity (AHR) and goblet cell hyperplasia in house dust mite (HDM)-challenged mice. However, the role of ApoE in Ovalbumin (OVA)-induced allergic airway inflammation disease and the underlying mechanism are still unknown. In the present study, murine allergic airway inflammation was induced by inhaled OVA for consecutive 7 days in wild type (WT) and ApoE-/- mice. In the OVA-induced model, the ApoE level in the bronchoalveolar lavage fluid (BALF) and lung tissues was significantly higher than that of control mice. And ApoE deficiency aggravated airway inflammation including leukocytes infiltration, goblet cell hyperplasia and IgE production as compared to those of WT mice after OVA- challenged, suggesting ApoE servers as an endogenous negative regulator of airway inflammation. Furthermore, OVA challenge elevated the activation of NLRP3 inflammasome with higher protein expression of NLRP3, caspase1 and IL-1β, enhanced oxidative stress with higher expression of 8-OHdG, nitrotyrosine and SOD2, increased the expression of mitochondrial fusion/fission markers including Optic Atrophy 1 (OPA1), Mitofusion 2 (Mfn2), dynamin-related protein 1 (DRP1) and Fission 1 (Fis1). However, these OVA-induced changes were augmented in ApoE-/- mice. Collectively, our results demonstrated that the OVA-induced airway inflammation was aggravated in ApoE-/- mice, and suggested that the underlying mechanism may be associated with the augmented activation of NLRP3 inflammasome and oxidative stress in ApoE-/- mice, therefore targeting ApoE pathway might be a novel therapy approach for allergic airway diseases such as asthma.
Collapse
Affiliation(s)
- Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Juan Xu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Xiao-Yun Fan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Guang-He Fei
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, PR China.
| |
Collapse
|
23
|
Roula D, Theiler A, Luschnig P, Sturm GJ, Tomazic PV, Marsche G, Heinemann A, Sturm EM. Apolipoprotein A-IV acts as an endogenous anti-inflammatory protein and is reduced in treatment-naïve allergic patients and allergen-challenged mice. Allergy 2020; 75:392-402. [PMID: 31408538 PMCID: PMC7065107 DOI: 10.1111/all.14022] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/07/2019] [Accepted: 06/26/2019] [Indexed: 12/15/2022]
Abstract
Background Recent studies pointed to a crucial role for apolipoproteins in the pathogenesis of inflammatory diseases. However, the role of apolipoprotein‐IV (ApoA‐IV) in allergic inflammation has not been addressed thoroughly thus far. Objective Here, we explored the anti‐inflammatory effects and underlying signaling pathways of ApoA‐IV on eosinophil effector function in vitro and in vivo. Methods Migratory responsiveness, Ca2+‐flux and apoptosis of human peripheral blood eosinophils were assessed in vitro. Allergen‐driven airway inflammation was assessed in a mouse model of acute house dust mite‐induced asthma. ApoA‐IV serum levels were determined by ELISA. Results Recombinant ApoA‐IV potently inhibited eosinophil responsiveness in vitro as measured by Ca2+‐flux, shape change, integrin (CD11b) expression, and chemotaxis. The underlying molecular mechanism involved the activation of Rev‐ErbA‐α and induced a PI3K/PDK1/PKA‐dependent signaling cascade. Systemic application of ApoA‐IV prevented airway hyperresponsiveness (AHR) and airway eosinophilia in mice following allergen challenge. ApoA‐IV levels were decreased in serum from allergic patients compared to healthy controls. Conclusion Our data suggest that ApoA‐IV is an endogenous anti‐inflammatory protein that potently suppresses effector cell functions in eosinophils. Thus, exogenously applied ApoA‐IV may represent a novel pharmacological approach for the treatment of allergic inflammation and other eosinophil‐driven disorders.
Collapse
Affiliation(s)
- David Roula
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| | - Anna Theiler
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| | - Petra Luschnig
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| | - Gunter J. Sturm
- Department of Dermatology and Venerology Medical University of Graz Graz Austria
- Allergy Outpatient Clinic Reumannplatz Vienna Austria
| | | | - Gunther Marsche
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| | - Eva M. Sturm
- Division of Pharmacology, Otto‐Loewi Research Center for Vascular Biology, Immunology and Inflammation Medical University of Graz Graz Austria
| |
Collapse
|
24
|
Banskar S, Detzner AA, Juarez-Rodriguez MD, Hozo I, Gupta D, Dziarski R. The Pglyrp1-Regulated Microbiome Enhances Experimental Allergic Asthma. THE JOURNAL OF IMMUNOLOGY 2019; 203:3113-3125. [PMID: 31704882 DOI: 10.4049/jimmunol.1900711] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022]
Abstract
Changes in intestinal or respiratory microbiomes in infants correlate with increased incidence of asthma, but the causative role of microbiome in the susceptibility to asthma and the host genes that regulate these changes in microbiome are mostly unknown. In this study, we show that decreased responsiveness to allergic asthma in Pglyrp1 -/- mice (lacking bactericidal peptidoglycan recognition protein 1) could be transferred to germ-free wild-type mice by colonization of mothers and newborns with microbiota from Pglyrp1 -/- mice. These colonized mice had decreased airway resistance and fewer inflammatory cells, less severe histopathology, and lower levels of IgE and proallergic cytokines and chemokines in the lungs. This microbiome-dependent decreased responsiveness to asthma was most pronounced in colonized germ-free BALB/c mice (genetically predisposed to asthma), only partially evident in outbred germ-free Swiss Webster mice, and marginal in conventional BALB/c mice following depletion of microbiome with antibiotics. Mice with a low asthmatic response colonized with microbiota from Pglyrp1 -/- mice had increased abundance of Bacteroidetes and decreased abundance of Firmicutes, Tenericutes, Deferribacteres, and Spirochaetes in the feces and increased abundance of Pasteurella in the oropharynx. These changes in bacterial abundance in the feces and oropharynx correlated with lower asthmatic responses in the lungs. Thus, our results show that Pglyrp1 enhances allergic asthmatic responses primarily through its effect on the host intestinal microbiome and identify several bacteria that may increase or decrease sensitivity to asthma. This effect of microbiome is strong in asthma-prone BALB/c mice and weak in asthma-resistant outbred mice and requires germ-free conditions before colonization with microbiota from Pglyrp1 -/- mice.
Collapse
Affiliation(s)
- Sunil Banskar
- Indiana University School of Medicine-Northwest, Gary, IN 46408; and
| | - Ashley A Detzner
- Indiana University School of Medicine-Northwest, Gary, IN 46408; and
| | | | - Iztok Hozo
- Department of Mathematics, Indiana University-Northwest, Gary, IN 46408
| | - Dipika Gupta
- Indiana University School of Medicine-Northwest, Gary, IN 46408; and
| | - Roman Dziarski
- Indiana University School of Medicine-Northwest, Gary, IN 46408; and
| |
Collapse
|
25
|
Bhowmik M, Majumdar S, Dasgupta A, Gupta Bhattacharya S, Saha S. Pilot-Scale Study Of Human Plasma Proteomics Identifies ApoE And IL33 As Markers In Atopic Asthma. J Asthma Allergy 2019; 12:273-283. [PMID: 31571934 PMCID: PMC6759800 DOI: 10.2147/jaa.s211569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Background The pathobiology of atopic asthma is complex and the symptoms similar to other respiratory diseases. As such, identification of biomarkers of atopic asthma is of prime importance for better diagnosis and control of the disease. Objectives We sought to study the changes in plasma proteome and cytokine-expression profile across healthy and atopic asthmatics for identifying biomarkers and exploring aberrant pathways for atopic asthma. Methods A pilot-scale study in humans was performed to identify differentially expressed proteins in blood plasma of healthy controls (n=5) and treatment-naïve atopic asthma patients (n=5) using quantitative label-free liquid chromatography-tandem mass spectrometry proteomics and ELISA. Results Mass spectrometry-based proteomic analysis revealed ApoE to be significantly downregulated in atopic asthmatics compared to healthy volunteers. Decreased expression of ApoE in atopic asthmatics was validated by immunoblotting (50.74% decrease). Comparison with atopic asthmatics and COPD patients showed that ApoE was decreased (36.33%) in atopic asthma compared to COPD. IL33 was significantly upregulated in atopic asthmatics compared to healthy subjects (3.84-fold). Conclusion ApoE was downregulated and IL33 upregulated in atopic asthma patients compared to healthy volunteers. These two proteins' profiles were distinct in atopic asthma from healthy and COPD plasma samples. Differential expression of these proteins could serve as a probable candidate for a two-protein classifier-based prognostic biomarker of atopic asthma.
Collapse
Affiliation(s)
- Moumita Bhowmik
- Division of Plant Biology, Bose Institute, Kolkata, West Bengal, India
| | - Sreyashi Majumdar
- Division of Bioinformatics, Bose Institute, Kolkata, West Bengal, India
| | - Angira Dasgupta
- BR Singh Hospital and Centre for Medical Education and Research, Kolkata, West Bengal, India
| | | | - Sudipto Saha
- Division of Bioinformatics, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
26
|
Nicholas TP, Kavanagh TJ, Faustman EM, Altemeier WA. The Effects of Gene × Environment Interactions on Silver Nanoparticle Toxicity in the Respiratory System. Chem Res Toxicol 2019; 32:952-968. [PMID: 31124663 DOI: 10.1021/acs.chemrestox.8b00234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Silver nanoparticles (AgNP) are used in multiple applications but primarily in the manufacturing of antimicrobial products. AgNP toxicity in the respiratory system is well characterized, but few in vitro or in vivo studies have evaluated the effects of interactions between host genetic and acquired factors or gene × environment interactions (G × E) on AgNP toxicity in the respiratory system. The primary goal of this article is to review host genetic and acquired factors identified across in vitro and in vivo studies and prioritize those necessary for defining exposure limits to protect all populations. The impact of these exposures and the work being done to address the current limited protections are also discussed. Future research on G × E effects on AgNP toxicity is warranted and will assist with informing regulatory or recommended exposure limits that enforce special protections for all populations to AgNP exposures in occupational settings.
Collapse
Affiliation(s)
- Tyler P Nicholas
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98109 , United States
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98109 , United States
| | - Elaine M Faustman
- Department of Environmental and Occupational Health Sciences , University of Washington , Seattle , Washington 98109 , United States
| | - William A Altemeier
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine , University of Washington , Seattle , Washington 98109 , United States
| |
Collapse
|
27
|
Gordon EM, Yao X, Xu H, Karkowsky W, Kaler M, Kalchiem-Dekel O, Barochia AV, Gao M, Keeran KJ, Jeffries KR, Levine SJ. Apolipoprotein E is a concentration-dependent pulmonary danger signal that activates the NLRP3 inflammasome and IL-1β secretion by bronchoalveolar fluid macrophages from asthmatic subjects. J Allergy Clin Immunol 2019; 144:426-441.e3. [PMID: 30872118 DOI: 10.1016/j.jaci.2019.02.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 02/15/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND House dust mite (HDM)-challenged Apoe-/- mice display enhanced airway hyperreactivity and mucous cell metaplasia. OBJECTIVE We sought to characterize the pathways that induce apolipoprotein E (APOE) expression by bronchoalveolar lavage fluid (BALF) macrophages from asthmatic subjects and identify how APOE regulates IL-1β secretion. METHODS Macrophages were isolated from asthmatic BALF and derived from THP-1 cells and human monocytes. RESULTS HDM-derived cysteine and serine proteases induced APOE secretion from BALF macrophages through protease-activated receptor 2. APOE at concentrations of less than 2.5 nmol/L, which are similar to levels found in epithelial lining fluid from healthy adults, did not induce IL-1β release from BALF macrophages. In contrast, APOE at concentrations of 25 nmol/L or greater induced nucleotide-binding oligomerization domain, leucine-rich repeat-containing protein (NLRP) 3 and pro-IL-1β expression by BALF macrophages, as well as the caspase-1-mediated generation of mature IL-1β secreted from cells. HDM acted synergistically with APOE to both prime and activate the NLRP3 inflammasome. In a murine model of neutrophilic airway inflammation induced by HDM and polyinosinic-polycytidylic acid, APOE reached a concentration of 32 nmol/L in epithelial lining fluid, with associated increases in BALF IL-1β levels. APOE-dependent NLRP3 inflammasome activation in macrophages was primarily mediated through a potassium efflux-dependent mechanism. CONCLUSION APOE can function as an endogenous, concentration-dependent pulmonary danger signal that primes and activates the NLPR3 inflammasome in BALF macrophages from asthmatic subjects to secrete IL-1β. This might represent a mechanism through which APOE amplifies pulmonary inflammatory responses when concentrations in the lung are increased to greater than normal levels, which can occur during viral exacerbations of HDM-induced asthma characterized by neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Haitao Xu
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - William Karkowsky
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Or Kalchiem-Dekel
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Meixia Gao
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Karen J Keeran
- Animal Surgery and Resources Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Kenneth R Jeffries
- Animal Surgery and Resources Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
28
|
Trieb M, Wolf P, Knuplez E, Weger W, Schuster C, Peinhaupt M, Holzer M, Trakaki A, Eichmann T, Lass A, Wadsack C, Schuligoi R, Heinemann A, Marsche G. Abnormal composition and function of high-density lipoproteins in atopic dermatitis patients. Allergy 2019; 74:398-402. [PMID: 30267572 PMCID: PMC6491993 DOI: 10.1111/all.13620] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Markus Trieb
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
- BioTechMed-Graz; Graz Austria
| | - Peter Wolf
- Department of Dermatology; Medical University of Graz; Graz Austria
| | - Eva Knuplez
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
| | - Wolfgang Weger
- Department of Dermatology; Medical University of Graz; Graz Austria
| | | | - Miriam Peinhaupt
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
| | - Michael Holzer
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
| | - Athina Trakaki
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
| | - Thomas Eichmann
- BioTechMed-Graz; Graz Austria
- Center of Explorative Lipidomics; Graz Austria
| | - Achim Lass
- BioTechMed-Graz; Graz Austria
- Institute of Molecular Biosciences; University of Graz; Graz Austria
| | - Christian Wadsack
- BioTechMed-Graz; Graz Austria
- Department of Obstetrics and Gynaecology; Medical University of Graz; Graz Austria
| | - Rufina Schuligoi
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
| | - Akos Heinemann
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
- BioTechMed-Graz; Graz Austria
| | - Gunther Marsche
- Division of Pharmacology; Otto Loewi Research Center; Medical University of Graz; Graz Austria
- BioTechMed-Graz; Graz Austria
| |
Collapse
|
29
|
Kulminski AM, Barochia AV, Loika Y, Raghavachari N, Arbeev KG, Wojczynski MK, Thyagarajan B, Vardarajan BN, Christensen K, Yashin AI, Levine SJ. The APOE ε4 allele is associated with a reduction in FEV1/FVC in women: A cross-sectional analysis of the Long Life Family Study. PLoS One 2018; 13:e0206873. [PMID: 30412599 PMCID: PMC6226172 DOI: 10.1371/journal.pone.0206873] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Murine studies have shown that apolipoprotein E modulates pulmonary function during development, aging, and allergen-induced airway disease. It is not known whether the polymorphic human APOE gene influences pulmonary function. OBJECTIVES We assessed whether an association exists between the polymorphic human APOE ε2, ε3, and ε4 alleles and pulmonary function among participants in the Long Life Family Study. METHODS Data from 4,468 Caucasian subjects who had genotyping performed for the APOE ε2, ε3, and ε4 alleles were analyzed, with and without stratification by sex. Statistical models were fitted considering the effects of the ε2 allele, defined as ε2/2 or ε2/3 genotypes, and the ε4 allele, defined as ε3/4 or ε4/4 genotypes, which were compared to the ε3/3 genotype. RESULTS The mean FEV1/FVC ratio (the forced expiratory volume in one second divided by the forced vital capacity) was lower among women with the ε4 allele as compared to women with the ε3/3 genotype or the ε2 allele. Carriage of the APOE ε4 allele was associated with FEV1/FVC, which implied lower values. Further analysis showed that the association primarily reflected women without lung disease who were older than 70 years. The association was not mediated by lipid levels, smoking status, body mass index, or cardiovascular disease. CONCLUSIONS This study for the first time identifies that the APOE gene is associated with modified lung physiology in women. This suggests that a link may exist between the APOE ε4 allele, female sex, and a reduction in the FEV1/FVC ratio in older individuals.
Collapse
Affiliation(s)
- Alexander M. Kulminski
- Biodemography of Aging Research Unit, Social Sciences Research Institute, Duke University, Durham, NC, United States of America
| | - Amisha V. Barochia
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, United States of America
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Sciences Research Institute, Duke University, Durham, NC, United States of America
| | - Nalini Raghavachari
- National Institute on Aging, Gateway Building, Suite, Bethesda, MD, United States of America
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Sciences Research Institute, Duke University, Durham, NC, United States of America
| | - Mary K. Wojczynski
- Division of Statistical Genomics, Department of Genetics, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, United States of America
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, United States of America
| | - Kaare Christensen
- The Danish Aging Research Center, University of Southern Denmark, Odense C, Denmark
- Department of Clinical Genetics and Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense C, Denmark
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Sciences Research Institute, Duke University, Durham, NC, United States of America
| | - Stewart J. Levine
- Laboratory of Asthma and Lung Inflammation, Pulmonary Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, United States of America
| |
Collapse
|
30
|
Su M, Lin W, Tsai C, Chiang B, Yang Y, Lin Y, Wang L, Lee J, Chou C, Wu Y, Yeh Y, Lee YL. Childhood asthma clusters reveal neutrophil-predominant phenotype with distinct gene expression. Allergy 2018; 73:2024-2032. [PMID: 29574758 DOI: 10.1111/all.13439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND Childhood asthma comprises different phenotypes with complex pathophysiology. Different asthma phenotypes evoke various clinical symptoms and vary in their responses to treatments. METHODS We applied k-means clustering algorithm of twelve objective laboratory tests among 351 asthmatic children enrolled in the Taiwanese Consortium of Childhood Asthma Study (TCCAS). We constructed gene expression profiles of peripheral blood mononuclear cells (PBMC) from children with different asthma phenotypes. RESULTS Five distinct phenotypes of childhood asthma were identified and can be characterized by either eosinophil-predominant or neutrophil-predominant inflammatory characteristics. In the gene expression profile analysis, significant differences were noted for neutrophil-predominant asthma, compared with samples from all the other asthma phenotypes. The vast majority of the differentially expressed genes in neutrophil-predominant asthma was associated with corticosteroid response. From an independent inhaled corticosteroid (ICS) response cohort, we also found neutrophils could be activated in this severe asthma phenotype and neutrophil-predominant asthma may be associated with corticosteroid nonresponsiveness. CONCLUSION Phenotype clustering of childhood asthma can be helpful to identify clinically relevant patients and reveal different inflammatory characteristics in asthmatic children. Neutrophil-predominant asthma is the most severe asthma phenotype with poor corticosteroid response. Gene expression profile of different asthma phenotypes not only improve our knowledge of childhood asthma, but also can guide asthma precision medicine.
Collapse
Affiliation(s)
- M.‐W. Su
- Institute of Epidemiology and Preventive Medicine College of Public Health National Taiwan University Taipei Taiwan
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - W.‐C. Lin
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - C.‐H. Tsai
- Institute of Epidemiology and Preventive Medicine College of Public Health National Taiwan University Taipei Taiwan
| | - B.‐L. Chiang
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - Y.‐H. Yang
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - Y.‐T. Lin
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - L.‐C. Wang
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - J.‐H. Lee
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
| | - C.‐C. Chou
- Department of Pediatrics National Taiwan University Hospital Taipei Taiwan
- Doctor Chou's Pediatric Clinic Taipei Taiwan
| | - Y.‐F. Wu
- Institute of Epidemiology and Preventive Medicine College of Public Health National Taiwan University Taipei Taiwan
| | - Y.‐L. Yeh
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
- School of Medicine College of Medicine Taipei Medical University Taipei Taiwan
| | - Y. L. Lee
- Institute of Epidemiology and Preventive Medicine College of Public Health National Taiwan University Taipei Taiwan
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| |
Collapse
|
31
|
Abstract
The lung has a unique relationship to cholesterol that is shaped by its singular physiology. On the one hand, the lungs receive the full cardiac output and have a predominant dependence on plasma lipoprotein uptake for their cholesterol supply. On the other hand, surfactant lipids, including cholesterol, are continually susceptible to oxidation owing to direct environmental exposure and must be cleared or recycled because of the very narrow biophysical mandates placed upon surfactant lipid composition. Interestingly, increased lipid-laden macrophage "foam cells" have been noted in a wide range of human lung pathologies. This suggests that lipid dysregulation may be a unifying and perhaps contributory event in chronic lung disease pathogenesis. Recent studies have shown that perturbations in intracellular cholesterol trafficking critically modify the immune response of macrophages and other cells. This minireview discusses literature that has begun to demonstrate the importance of regulated cholesterol traffic through the lung to pulmonary immunity, inflammation, and fibrosis. This emerging recognition of coupling between immunity and lipid homeostasis in the lung presents potentially transformative concepts for understanding lung disease and may also offer novel and exciting avenues for therapeutic development.
Collapse
|
32
|
Scaduto F, Giglio RV, Benfante A, Nikolic D, Montalto G, Rizzo M, Scichilone N. Serum lipoproteins are not associated with the severity of asthma. Pulm Pharmacol Ther 2018; 50:57-61. [PMID: 29626633 DOI: 10.1016/j.pupt.2018.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/18/2018] [Accepted: 04/03/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Asthma is a chronic inflammatory disorder of the bronchi with a complicated and largely unknown pathogenesis. In this context, an emerging role is attributed to the apolipoproteins which serve as structural components of plasma lipoproteins. Low density lipoproteins (LDL) may be involved in the inflammatory pathways of the asthmatic airways; in particular, small dense LDL (sdLDL) particles were associated with increased oxidative susceptibility compared to medium and large sized LDL. In our previous study, we found a positive correlation between forced expiratory volume 1 s (FEV1) % predicted and larger LDL particles (LDL-1), and an inverse correlation between FEV1% predicted and sdLDL (LDL-3) in mild, untreated asthmatics. Although LDL appear to be important modulators of inflammation, data on their clinical implications are still lacking. OBJECTIVE The aim of the study is to investigate whether LDL subclasses correlate with the severity of asthma, assuming that the atherogenic and most pro-inflammatory LDL contribute to ignite and perpetuate the airway inflammatory processes. METHODS The study was conducted in one visit, and included clinical and lung functional assessments, as well as measurements of serum concentrations of the LDL subclasses. Non-denaturing, linear polyacrylamide gel electrophoresis was used to separate and measure LDL subclasses, with the LipoPrint© System (Quantimetrix Corporation, Redondo Beach, CA, USA). LDL subclasses were distributed as seven bands (LDL-1 to LDL-7), LDL-1 and -2 being defined as large LDL (least pro-inflammatory), and LDL-3 to 7 defined as sdLDL (most pro-inflammatory). RESULTS 70 asthmatics under inhaled treatment (M/F: 35/35) were enrolled; 10 healthy subjects (M/F: 3/7) served as controls. In the asthmatic group, FEV1% predicted was 81 ± 22% (mean ± SD), vital capacity (VC) % predicted was 97 ± 18%, and FEV1/FVC was 0.68 ± 0.1. The mean asthma control test (ACT) score was 18 ± 5. LDL-1 were significantly lower in asthmatics as compared to controls (18 ± 4% vs. 22 ± 4%, p = 0.008). On the contrary, LDL-2 (12 ± 4% vs. 12 ± 5%) and LDL-3 (3 ± 3% vs. 2 ± 2%) were not statistically different between the two groups; smaller subclasses were undetectable. To comply with the design of the study, subjects were classified according to their degree of severity into the 5 Global Initiative for Asthma (GINA) steps: Step 1 (M/F: 4/3, 44 ± 12 yrs), Step 2 (M/F: 1/2, 37 ± 11 yrs), Step 3 (M/F: 12/7, 47 ± 12 yrs), Step 4 (M/F: 8/15, 54 ± 12 yrs), and Step 5 (M/F: 7/9, 56 ± 9 yrs). None of the LDL subclasses showed significant differences between classes of severity: LDL-1 were 16.1 ± 5.6% in Step 1, 18 ± 2.8% in Step 2, 16.7 ± 3.7% in Step 3, 18 ± 3.3% in Step 4, and 19.5 ± 3.2% in Step 5 (p = NS); LDL2 were 14 ± 3.6%, 15 ± 3.4%, 12.4 ± 5.3%, 12.7 ± 4.4% and 11.3 ± 4.2%, respectively (p = NS); LDL3 were 5 ± 5.2%, 4.4 ± 2.6%, 3.3 ± 3.6%, 3.2 ± 2.6% and 2.4 ± 1.8%, p = NS. Finally, no relationship was detected between LDL subclasses and lung function parameters as well as the ACT scores. CONCLUSIONS The current findings confirm a role of LDL as a potential biomarker in the diagnostic process for asthma, and suggest that LDL cannot be used as marker of severity of the disease.
Collapse
Affiliation(s)
- Federica Scaduto
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Rosaria Vincenza Giglio
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Alida Benfante
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Dragana Nikolic
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Giuseppe Montalto
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Manfredi Rizzo
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy
| | - Nicola Scichilone
- BioMedical Department of Internal Medicine and Medical Specialties (DiBiMIS), University of Palermo, Italy.
| |
Collapse
|
33
|
Mishra A, Yao X, Saxena A, Gordon EM, Kaler M, Cuento RA, Barochia AV, Dagur PK, McCoy JP, Keeran KJ, Jeffries KR, Qu X, Yu ZX, Levine SJ. Low-density lipoprotein receptor-related protein 1 attenuates house dust mite-induced eosinophilic airway inflammation by suppressing dendritic cell-mediated adaptive immune responses. J Allergy Clin Immunol 2017; 142:1066-1079.e6. [PMID: 29274414 DOI: 10.1016/j.jaci.2017.10.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/20/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 1 (LRP-1) is a scavenger receptor that regulates adaptive immunity and inflammation. LRP-1 is not known to modulate the pathogenesis of allergic asthma. OBJECTIVE We sought to assess whether LRP-1 expression by dendritic cells (DCs) modulates adaptive immune responses in patients with house dust mite (HDM)-induced airways disease. METHODS LRP-1 expression on peripheral blood DCs was quantified by using flow cytometry. The role of LRP-1 in modulating HDM-induced airways disease was assessed in mice with deletion of LRP-1 in CD11c+ cells (Lrp1fl/fl; CD11c-Cre) and by adoptive transfer of HDM-pulsed CD11b+ DCs from Lrp1fl/fl; CD11c-Cre mice to wild-type (WT) mice. RESULTS Human peripheral blood myeloid DC subsets from patients with eosinophilic asthma have lower LRP-1 expression than cells from healthy nonasthmatic subjects. Similarly, LRP-1 expression by CD11b+ lung DCs was significantly reduced in HDM-challenged WT mice. HDM-challenged Lrp1fl/fl; CD11c-Cre mice have a phenotype of increased eosinophilic airway inflammation, allergic sensitization, TH2 cytokine production, and mucous cell metaplasia. The adoptive transfer of HDM-pulsed LRP-1-deficient CD11b+ DCs into WT mice generated a similar phenotype of enhanced eosinophilic inflammation and allergic sensitization. Furthermore, CD11b+ DCs in the lungs of Lrp1fl/fl; CD11c-Cre mice have an increased ability to take up HDM antigen, whereas bone marrow-derived DCs display enhanced antigen presentation capabilities. CONCLUSION This identifies a novel role for LRP-1 as a negative regulator of DC-mediated adaptive immune responses in the setting of HDM-induced eosinophilic airway inflammation. Furthermore, the reduced LRP-1 expression by circulating myeloid DCs in patients with eosinophilic asthma suggests a possible role for LRP-1 in modulating type 2-high asthma.
Collapse
Affiliation(s)
- Amarjit Mishra
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Ankit Saxena
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Rosemarie A Cuento
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Pradeep K Dagur
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - J Philip McCoy
- Flow Cytometry Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Karen J Keeran
- Animal Surgery and Resources Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Kenneth R Jeffries
- Animal Surgery and Resources Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Xuan Qu
- Pathology Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Zu-Xi Yu
- Pathology Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
34
|
Fawzy El-Sayed KM, Dörfer CE. Animal Models for Periodontal Tissue Engineering: A Knowledge-Generating Process. Tissue Eng Part C Methods 2017; 23:900-925. [DOI: 10.1089/ten.tec.2017.0130] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Karim M. Fawzy El-Sayed
- Department of Oral Medicine and Periodontology, Faculty of Oral and Dental Medicine, Cairo University, Giza, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
35
|
Zuo J, Lei M, Wen M, Chen Y, Liu Z. Overexpression of ATP5b promotes cell proliferation in asthma. Mol Med Rep 2017; 16:6946-6952. [PMID: 28901394 DOI: 10.3892/mmr.2017.7413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 11/18/2016] [Indexed: 11/06/2022] Open
Abstract
Asthma is a complicated systemic disease of the airways, which is characterized by variable symptoms, including bronchial hyper‑responsive-ness, inflammation and airflow obstruction. The prevalence of asthma has increased 2‑3‑fold over recent decades in developed countries; however, the molecular mechanism of asthma remains unclear. In the current study, the expression of recombinant protein Dermatophagoides farinaeI (Derf I) was induced by isopropyl β‑D‑1‑thiogalactoside (IPTG) and purified using Ni‑NTA. Derf I, an important antigen of asthma, was used to establish the animal model of asthma. Airway hyper‑responsiveness was mea-sured using unrestrained whole‑body plethysmography with a four‑chamber system. Immunoglobulin (Ig)E, IgG and IgG2a were analyzed using indirect enzyme‑linked immunosorbent assay (ELISA). Proteomic technology was applied to detect the difference between the normal lung tissue and asthma lung tissue samples of the asthma model. Cytokines in bronchoalveolar lavage fluid and the splenocyte culture medium were measured by ELISA and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was performed to detect the mRNA expression of ATP synthase, H+ transporting, mitochondrial F1 complex, β polypeptide (ATP5b). In addition, cell growth of arterial smooth muscle cells (ASMCs) was evaluated by MTT assay. In the current study, Derf I was successfully used to construct the animal model of asthma. Out of 23 proteins that exhibit 3‑fold upregulation or downregulation, ATP5b was chosen for further investigation. The data indicated that ATP5b was overexpressed in the asthma lung tissue when compared with the normal lung tissue. However, when ATP5b was knocked down, cell growth decreased. Therefore, overexpressed ATP5b leads to airway smooth muscle cell (ASMC) proliferation and finally to ASM thickening. Thus, to the best of our knowledge, this is the first study to report that the expression level of ATP5b was markedly increased in lung tissue samples of an asthma model compared with the tissue samples from normal lungs, which promoted ASMC proliferation and contributed to airway remodeling.
Collapse
Affiliation(s)
- Jianhong Zuo
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Institute, School of Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mingsheng Lei
- Department of Respiratory and Critical Care Medicine, Zhangjiajie City Hospital, Zhangjiajie, Hunan 427000, P.R. China
| | - Meilin Wen
- Department of Oncology, Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yikun Chen
- State Key Laboratory of Respiratory Disease for Allergy, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Zhigang Liu
- State Key Laboratory of Respiratory Disease for Allergy, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
36
|
Yao X, Gordon EM, Figueroa DM, Barochia AV, Levine SJ. Emerging Roles of Apolipoprotein E and Apolipoprotein A-I in the Pathogenesis and Treatment of Lung Disease. Am J Respir Cell Mol Biol 2017; 55:159-69. [PMID: 27073971 DOI: 10.1165/rcmb.2016-0060tr] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Emerging roles are being recognized increasingly for apolipoproteins in the pathogenesis and treatment of lung diseases on the basis of their ability to suppress inflammation, oxidative stress, and tissue remodeling, and to promote adaptive immunity and host defense. Apolipoproteins, such as apolipoprotein E (apoE) and apolipoprotein A-I (apoA-I), are important components of lipoprotein particles that facilitate the transport of cholesterol, triglycerides, and phospholipids between plasma and cells. ApoE-containing lipoprotein particles are internalized into cells by low-density lipoprotein receptors (LDLRs), whereas apoA-I can interact with the ATP-binding cassette subfamily A member 1 (ABCA1) transporter to efflux cholesterol and phospholipids out of cells. ApoE and apoA-I also mediate receptor-independent effects, such as binding to and neutralizing LPS. Both apoE and apoA-I are expressed by lung cells, which allows apoE/LDLR- and apoA-I/ABCA1-dependent pathways to modulate normal lung health and the pathogenesis of respiratory diseases, including asthma, acute lung injury, cancer, emphysema, pulmonary fibrosis, and pulmonary hypertension. Data from human studies and research using experimental murine model systems have shown that both apoE and apoA-I pathways play primarily protective roles in lung biology and respiratory disease. Furthermore, apolipoprotein mimetic peptides, corresponding to the LDLR-binding domain of apoE or the class A amphipathic α-helical structure of apoA-I, have antiinflammatory and antioxidant effects that attenuate the severity of lung disease in murine models. Thus, the development of inhaled apolipoprotein mimetic peptides as a novel treatment paradigm could represent a significant advance for patients with respiratory disease who do not respond to current therapies.
Collapse
Affiliation(s)
- Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Debbie M Figueroa
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Cardiovascular and Pulmonary Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Barochia AV, Gordon EM, Kaler M, Cuento RA, Theard P, Figueroa DM, Yao X, Weir NA, Sampson ML, Stylianou M, Choy DF, Holweg CTJ, Remaley AT, Levine SJ. High density lipoproteins and type 2 inflammatory biomarkers are negatively correlated in atopic asthmatics. J Lipid Res 2017; 58:1713-1721. [PMID: 28655726 DOI: 10.1194/jlr.p077776] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Blood eosinophil counts and serum periostin levels are biomarkers of type 2 inflammation. Although serum levels of HDL and apoA-I have been associated with less severe airflow obstruction in asthma, it is not known whether serum lipids or lipoprotein particles are correlated with type 2 inflammation in asthmatics. Here, we assessed whether serum lipids and lipoproteins correlated with blood eosinophil counts or serum periostin levels in 165 atopic asthmatics and 163 nonasthmatic subjects with and without atopy. Serum lipids and lipoproteins were quantified using standard laboratory assays and NMR spectroscopy. Absolute blood eosinophils were quantified by complete blood counts. Periostin levels were measured using the Elecsys® periostin assay. In atopic asthmatics, blood eosinophils negatively correlated with serum HDL cholesterol and total HDL particles measured by NMR spectroscopy (HDLNMR). Serum periostin levels negatively correlated with total HDLNMR In contrast, blood eosinophil counts positively correlated with serum triglyceride levels. This study demonstrates for the first time that HDL particles were negatively correlated, whereas serum triglycerides were positively correlated, with blood eosinophils in atopic asthmatics. This supports the concept that serum levels of HDL and triglycerides may be linked to systemic type 2 inflammation in atopic asthma.
Collapse
Affiliation(s)
- Amisha V Barochia
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892.
| | - Elizabeth M Gordon
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Maryann Kaler
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Rosemarie A Cuento
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Patricia Theard
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Debbie M Figueroa
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Xianglan Yao
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Nargues A Weir
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| | - Maureen L Sampson
- National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Mario Stylianou
- Cardiovascular and Pulmonary Branch, and Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | - Alan T Remaley
- Lipoprotein Metabolism Section,National Institutes of Health, Bethesda, MD 20892
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
38
|
Kantarci A, Hasturk H, Van Dyke TE. Animal models for periodontal regeneration and peri-implant responses. Periodontol 2000 2017; 68:66-82. [PMID: 25867980 DOI: 10.1111/prd.12052] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2013] [Indexed: 11/28/2022]
Abstract
Translation of experimental data to the clinical setting requires the safety and efficacy of such data to be confirmed in animal systems before application in humans. In dental research, the animal species used is dependent largely on the research question or on the disease model. Periodontal disease and, by analogy, peri-implant disease, are complex infections that result in a tissue-degrading inflammatory response. It is impossible to explore the complex pathogenesis of periodontitis or peri-implantitis using only reductionist in-vitro methods. Both the disease process and healing of the periodontal and peri-implant tissues can be studied in animals. Regeneration (after periodontal surgery), in response to various biologic materials with potential for tissue engineering, is a continuous process involving various types of tissue, including epithelia, connective tissues and alveolar bone. The same principles apply to peri-implant healing. Given the complexity of the biology, animal models are necessary and serve as the standard for successful translation of regenerative materials and dental implants to the clinical setting. Smaller species of animal are more convenient for disease-associated research, whereas larger animals are more appropriate for studies that target tissue healing as the anatomy of larger animals more closely resembles human dento-alveolar architecture. This review focuses on the animal models available for the study of regeneration in periodontal research and implantology; the advantages and disadvantages of each animal model; the interpretation of data acquired; and future perspectives of animal research, with a discussion of possible nonanimal alternatives. Power calculations in such studies are crucial in order to use a sample size that is large enough to generate statistically useful data, whilst, at the same time, small enough to prevent the unnecessary use of animals.
Collapse
|
39
|
Patel S, Meher B. A review on emerging frontiers of house dust mite and cockroach allergy research. Allergol Immunopathol (Madr) 2016; 44:580-593. [PMID: 26994963 DOI: 10.1016/j.aller.2015.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 11/16/2015] [Indexed: 12/28/2022]
Abstract
Currently, mankind is afflicted with diversified health issues, allergies being a common, yet little understood malady. Allergies, the outcome of a baffled immune system encompasses myriad allergens and causes an array of health consequences, ranging from transient to recurrent and mild to fatal. Indoor allergy is a serious hypersensitivity in genetically-predisposed people, triggered by ingestion, inhalation or mere contact of allergens, of which mite and cockroaches are one of the most-represented constituents. Arduous to eliminate, these aeroallergens pose constant health challenges, mostly manifested as respiratory and dermatological inflammations, leading to further aggravations if unrestrained. Recent times have seen an unprecedented endeavour to understand the conformation of these allergens, their immune manipulative ploys and other underlying causes of pathogenesis, most importantly therapies. Yet a large section of vulnerable people is ignorant of these innocuous-looking immune irritants, prevailing around them, and continues to suffer. This review aims to expedite this field by a concise, informative account of seminal findings in the past few years, with particular emphasis on leading frontiers like genome-wide association studies (GWAS), epitope mapping, metabolomics etc. Drawbacks linked to current approaches and solutions to overcome them have been proposed.
Collapse
|
40
|
Lung surfactant metabolism: early in life, early in disease and target in cell therapy. Cell Tissue Res 2016; 367:721-735. [PMID: 27783217 DOI: 10.1007/s00441-016-2520-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/27/2016] [Indexed: 01/07/2023]
Abstract
Lung surfactant is a complex mixture of lipids and proteins lining the alveolar epithelium. At the air-liquid interface, surfactant lowers surface tension, avoiding alveolar collapse and reducing the work of breathing. The essential role of lung surfactant in breathing and therefore in life, is highlighted by surfactant deficiency in premature neonates, which causes neonatal respiratory distress syndrome and results in early death after birth. In addition, defects in surfactant metabolism alter lung homeostasis and lead to disease. Special attention should be paid to two important key cells responsible for surfactant metabolism: alveolar epithelial type II cells (AE2C) and alveolar macrophages (AM). On the one hand, surfactant deficiency coming from abnormal AE2C function results in high surface tension, promoting alveolar collapse and mechanical stress in the epithelium. This epithelial injury contributes to tissue remodeling and lung fibrosis. On the other hand, impaired surfactant catabolism by AM leads to accumulation of surfactant in air spaces and the associated altered lung function in pulmonary alveolar proteinosis (PAP). We review here two recent cell therapies that aim to recover the activity of AE2C or AM, respectively, therefore targeting the restoring of surfactant metabolism and lung homeostasis. Applied therapies successfully show either transplantation of healthy AE2C in fibrotic lungs, to replace injured AE2C cells and surfactant, or transplantation of bone marrow-derived macrophages to counteract accumulation of surfactant lipid and proteinaceous material in the alveolar spaces leading to PAP. These therapies introduce an alternative treatment with great potential for patients suffering from lung diseases.
Collapse
|
41
|
Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA, Pirottin D, Janss T, Starkl P, Ramery E, Henket M, Schleich FN, Radermecker M, Thielemans K, Gillet L, Thiry M, Belvisi MG, Louis R, Desmet C, Marichal T, Bureau F. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 2016; 126:3279-95. [PMID: 27548519 DOI: 10.1172/jci85664] [Citation(s) in RCA: 402] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Increases in eosinophil numbers are associated with infection and allergic diseases, including asthma, but there is also evidence that eosinophils contribute to homeostatic immune processes. In mice, the normal lung contains resident eosinophils (rEos), but their function has not been characterized. Here, we have reported that steady-state pulmonary rEos are IL-5-independent parenchymal Siglec-FintCD62L+CD101lo cells with a ring-shaped nucleus. During house dust mite-induced airway allergy, rEos features remained unchanged, and rEos were accompanied by recruited inflammatory eosinophils (iEos), which were defined as IL-5-dependent peribronchial Siglec-FhiCD62L-CD101hi cells with a segmented nucleus. Gene expression analyses revealed a more regulatory profile for rEos than for iEos, and correspondingly, mice lacking lung rEos showed an increase in Th2 cell responses to inhaled allergens. Such elevation of Th2 responses was linked to the ability of rEos, but not iEos, to inhibit the maturation, and therefore the pro-Th2 function, of allergen-loaded DCs. Finally, we determined that the parenchymal rEos found in nonasthmatic human lungs (Siglec-8+CD62L+IL-3Rlo cells) were phenotypically distinct from the iEos isolated from the sputa of eosinophilic asthmatic patients (Siglec-8+CD62LloIL-3Rhi cells), suggesting that our findings in mice are relevant to humans. In conclusion, our data define lung rEos as a distinct eosinophil subset with key homeostatic functions.
Collapse
|
42
|
ApoE deficiency promotes colon inflammation and enhances inflammatory potential oxidized-LDL and TNF-α in colon epithelial cells. Biosci Rep 2016; 36:BSR20160195. [PMID: 27538678 PMCID: PMC5052706 DOI: 10.1042/bsr20160195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/18/2016] [Indexed: 12/12/2022] Open
Abstract
Although deficiency in Apolipoprotein E (ApoE) is linked to many diseases, its effect on colon homoeostasis remains unknown. ApoE appears to control inflammation by regulating nuclear factor-κB (NF-κB). The present study was designed to examine whether ApoE deficiency affects factors of colon integrity in vivo and given the likelihood that ApoE deficiency increases oxidized lipids and TNF-α, the present study also examined whether such deficiency enhances the inflammatory potential of oxidized-LDL (oxLDL) and TNF-α in colon epithelial cells (CECs), in vitro. Here we show that ApoE deficiency is associated with chronic inflammation systemically and in colonic tissues as assessed by TNF-α levels. Increased colon TNF-α mRNA coincided with a substantial increase in cyclooxygenase (COX)-2. ApoE deficiency enhanced the potential of oxLDL and TNF-α to induce COX-2 expression as well as several other inflammatory factors in primary CECs. Interestingly, oxLDL enhanced TGF-β expression only in ApoE−/−, but not in wild-type, epithelial cells. ApoE deficiency appears to promote COX-2 expression enhancement through a mechanism that involves persistent NF-κB nuclear localization and PI3 and p38 MAP kinases but independently of Src. In mice, ApoE deficiency promoted a moderate increase in crypt length, which was associated with opposing effects of an increase in cell proliferation and apoptosis at the bottom and top of the crypt respectively. Our results support the notion that ApoE plays a central role in colon homoeostasis and that ApoE deficiency may constitute a risk factor for colon pathologies.
Collapse
|
43
|
Smet M, Van Hoecke L, De Beuckelaer A, Vander Beken S, Naessens T, Vergote K, Willart M, Lambrecht BN, Gustafsson JÅ, Steffensen KR, Grooten J. Cholesterol-sensing liver X receptors stimulate Th2-driven allergic eosinophilic asthma in mice. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:350-61. [PMID: 27621817 PMCID: PMC5004289 DOI: 10.1002/iid3.118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/02/2016] [Accepted: 06/29/2016] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Liver X receptors (LXRs) are nuclear receptors that function as cholesterol sensors and regulate cholesterol homeostasis. High cholesterol has been recognized as a risk factor in asthma; however, the mechanism of this linkage is not known. METHODS To explore the importance of cholesterol homeostasis for asthma, we investigated the contribution of LXR activity in an ovalbumin- and a house dust mite-driven eosinophilic asthma mouse model. RESULTS In both models, airway inflammation, airway hyper-reactivity, and goblet cell hyperplasia were reduced in mice deficient for both LXRα and LXRβ isoforms (LXRα(-/-)β(-/-)) as compared to wild-type mice. Inversely, treatment with the LXR agonist GW3965 showed increased eosinophilic airway inflammation. LXR activity contributed to airway inflammation through promotion of type 2 cytokine production as LXRα(-/-)β(-/-) mice showed strongly reduced protein levels of IL-5 and IL-13 in the lungs as well as reduced expression of these cytokines by CD4(+) lung cells and lung-draining lymph node cells. In line herewith, LXR activation resulted in increased type 2 cytokine production by the lung-draining lymph node cells. CONCLUSIONS In conclusion, our study demonstrates that the cholesterol regulator LXR acts as a positive regulator of eosinophilic asthma in mice, contributing to airway inflammation through regulation of type 2 cytokine production.
Collapse
Affiliation(s)
- Muriel Smet
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Lien Van Hoecke
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium; Medical Biotechnology CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Ans De Beuckelaer
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Seppe Vander Beken
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Thomas Naessens
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Karl Vergote
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Monique Willart
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Bart N Lambrecht
- Department of Respiratory MedicineGhent University HospitalGhentBelgium; Inflammation Research CenterFlanders Institute for BiotechnologyGhentBelgium
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition at NovumKarolinska InstitutetStockholmSweden; Department of Biology and BiochemistryUniversity of HoustonHoustonTexas
| | - Knut R Steffensen
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Johan Grooten
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| |
Collapse
|
44
|
Liu CL, Zhang JY, Shi GP. Interaction between allergic asthma and atherosclerosis. Transl Res 2016; 174:5-22. [PMID: 26608212 PMCID: PMC4826642 DOI: 10.1016/j.trsl.2015.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 09/25/2015] [Accepted: 09/29/2015] [Indexed: 12/15/2022]
Abstract
Prior studies have established an essential role of mast cells in allergic asthma and atherosclerosis. Mast cell deficiency or inactivation protects mice from allergen-induced airway hyper-responsiveness and diet-induced atherosclerosis, suggesting that mast cells share pathologic activities in both diseases. Allergic asthma and atherosclerosis are inflammatory diseases that contain similar sets of elevated numbers of inflammatory cells in addition to mast cells in the airway and arterial wall, such as macrophages, monocytes, T cells, eosinophils, and smooth muscle cells. Emerging evidence from experimental models and human studies points to a potential interaction between the 2 seemingly unrelated diseases. Patients or mice with allergic asthma have a high risk of developing atherosclerosis or vice versa, despite the fact that asthma is a T-helper (Th)2-oriented disease, whereas Th1 immunity promotes atherosclerosis. In addition to the preferred Th1/Th2 responses that may differentiate the 2 diseases, mast cells and many other inflammatory cells also contribute to their pathogenesis by more than just T cell immunity. Here, we summarize the different roles of airway and arterial wall inflammatory cells and vascular cells in asthma and atherosclerosis and propose an interaction between the 2 diseases, although limited investigations are available to delineate the molecular and cellular mechanisms by which 1 disease increases the risk of the other. Results from mouse allergic asthma and atherosclerosis models and from human population studies lead to the hypothesis that patients with atherosclerosis may benefit from antiasthmatic medications or that the therapeutic regimens targeting atherosclerosis may also alleviate allergic asthma.
Collapse
Affiliation(s)
- Cong-Lin Liu
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Jin-Ying Zhang
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo-Ping Shi
- Department of Cardiology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
45
|
Regulation of Adaptive Immunity in Health and Disease by Cholesterol Metabolism. Curr Allergy Asthma Rep 2015; 15:48. [PMID: 26149587 DOI: 10.1007/s11882-015-0548-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Four decades ago, it was observed that stimulation of T cells induces rapid changes in cellular cholesterol that are required before proliferation can commence. Investigators returning to this phenomenon have finally revealed its molecular underpinnings. Cholesterol trafficking and its dysregulation are now also recognized to strongly influence dendritic cell function, T cell polarization, and antibody responses. In this review, the state of the literature is reviewed on how cholesterol and its trafficking regulate the cells of the adaptive immune response and in vivo disease phenotypes of dysregulated adaptive immunity, including allergy, asthma, and autoimmune disease. Emerging evidence supporting a potential role for statins and other lipid-targeted therapies in the treatment of these diseases is presented. Just as vascular biologists have embraced immunity in the pathogenesis and treatment of atherosclerosis, so should basic and clinical immunologists in allergy, pulmonology, and other disciplines seek to encompass a basic understanding of lipid science.
Collapse
|
46
|
Himes BE, Koziol-White C, Johnson M, Nikolos C, Jester W, Klanderman B, Litonjua AA, Tantisira KG, Truskowski K, MacDonald K, Panettieri RA, Weiss ST. Vitamin D Modulates Expression of the Airway Smooth Muscle Transcriptome in Fatal Asthma. PLoS One 2015; 10:e0134057. [PMID: 26207385 PMCID: PMC4514847 DOI: 10.1371/journal.pone.0134057] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/03/2015] [Indexed: 01/20/2023] Open
Abstract
Globally, asthma is a chronic inflammatory respiratory disease affecting over 300 million people. Some asthma patients remain poorly controlled by conventional therapies and experience more life-threatening exacerbations. Vitamin D, as an adjunct therapy, may improve disease control in severe asthma patients since vitamin D enhances glucocorticoid responsiveness and mitigates airway smooth muscle (ASM) hyperplasia. We sought to characterize differences in transcriptome responsiveness to vitamin D between fatal asthma- and non-asthma-derived ASM by using RNA-Seq to measure ASM transcript expression in five donors with fatal asthma and ten non-asthma-derived donors at baseline and with vitamin D treatment. Based on a Benjamini-Hochberg corrected p-value <0.05, 838 genes were differentially expressed in fatal asthma vs. non-asthma-derived ASM at baseline, and vitamin D treatment compared to baseline conditions induced differential expression of 711 and 867 genes in fatal asthma- and non-asthma-derived ASM, respectively. Functional gene categories that were highly represented in all groups included extracellular matrix, and responses to steroid hormone stimuli and wounding. Genes differentially expressed by vitamin D also included cytokine and chemokine activity categories. Follow-up qPCR and individual analyte ELISA experiments were conducted for four cytokines (i.e. CCL2, CCL13, CXCL12, IL8) to measure TNFα-induced changes by asthma status and vitamin D treatment. Vitamin D inhibited TNFα-induced IL8 protein secretion levels to a comparable degree in fatal asthma- and non-asthma-derived ASM even though IL8 had significantly higher baseline levels in fatal asthma-derived ASM. Our findings identify vitamin D-specific gene targets and provide transcriptomic data to explore differences in the ASM of fatal asthma- and non-asthma-derived donors.
Collapse
Affiliation(s)
- Blanca E. Himes
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Cynthia Koziol-White
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Martin Johnson
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Christina Nikolos
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - William Jester
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | | | - Augusto A. Litonjua
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Kelan G. Tantisira
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Kevin Truskowski
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Kevin MacDonald
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Reynold A. Panettieri
- Airways Biology Initiative, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Scott T. Weiss
- Partners Personalized Medicine, Boston, MA, United States of America
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
47
|
Barochia AV, Kaler M, Cuento RA, Gordon EM, Weir NA, Sampson M, Fontana JR, MacDonald S, Moss J, Manganiello V, Remaley AT, Levine SJ. Serum apolipoprotein A-I and large high-density lipoprotein particles are positively correlated with FEV1 in atopic asthma. Am J Respir Crit Care Med 2015; 191:990-1000. [PMID: 25692941 DOI: 10.1164/rccm.201411-1990oc] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Although lipids, apolipoproteins, and lipoprotein particles are important modulators of inflammation, varying relationships exist between these parameters and asthma. OBJECTIVES To determine whether serum lipids and apolipoproteins correlate with the severity of airflow obstruction in subjects with atopy and asthma. METHODS Serum samples were obtained from 154 atopic and nonatopic subjects without asthma, and 159 subjects with atopy and asthma. Serum lipid and lipoprotein levels were quantified using standard diagnostic assays and nuclear magnetic resonance (NMR) spectroscopy. Airflow obstruction was assessed by FEV1% predicted. MEASUREMENTS AND MAIN RESULTS Serum lipid levels correlated with FEV1 only in the subjects with atopy and asthma. Serum levels of high-density lipoprotein (HDL) cholesterol and apolipoprotein A-I (apoA-I) were positively correlated with FEV1 in subjects with atopy and asthma, whereas a negative correlation existed between FEV1 and serum levels of triglycerides, low-density lipoprotein (LDL) cholesterol, apolipoprotein B (apoB), and the apoB/apoA-I ratio. NMR spectroscopy identified a positive correlation between FEV1 and HDLNMR particle size, as well as the concentrations of large HDLNMR particles and total IDLNMR (intermediate-density lipoprotein) particles in subjects with atopy and asthma. In contrast, LDLNMR particle size and concentrations of LDLNMR and VLDLNMR (very-low-density lipoprotein) particles were negatively correlated with FEV1 in subjects with atopy and asthma. CONCLUSIONS In subjects with atopy and asthma, serum levels of apoA-I and large HDLNMR particles are positively correlated with FEV1, whereas serum triglycerides, LDL cholesterol, and apoB are associated with more severe airflow obstruction. These results may facilitate future studies to assess whether apoA-I and large HDLNMR particles can reduce airflow obstruction and disease severity in asthma.
Collapse
|
48
|
Burnham ME, Esnault S, Roti Roti EC, Bates ME, Bertics PJ, Denlinger LC. Cholesterol selectively regulates IL-5 induced mitogen activated protein kinase signaling in human eosinophils. PLoS One 2014; 9:e103122. [PMID: 25121926 PMCID: PMC4133209 DOI: 10.1371/journal.pone.0103122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 06/26/2014] [Indexed: 01/21/2023] Open
Abstract
Eosinophils function contributes to human allergic and autoimmune diseases, many of which currently lack curative treatment. Development of more effective treatments for eosinophil-related diseases requires expanded understanding of eosinophil signaling and biology. Cell signaling requires integration of extracellular signals with intracellular responses, and is organized in part by cholesterol rich membrane microdomains (CRMMs), commonly referred to as lipid rafts. Formation of these organizational membrane domains is in turn dependent upon the amount of available cholesterol, which can fluctuate widely with a variety of disease states. We tested the hypothesis that manipulating membrane cholesterol content in primary human peripheral blood eosinophils (PBEos) would selectively alter signaling pathways that depend upon membrane-anchored signaling proteins localized within CRMMs (e.g., mitogen activated protein kinase [MAPK] pathway), while not affecting pathways that signal through soluble proteins, like the Janus Kinase/Signal Transducer and Activator of Transcription [JAK/STAT] pathway. Cholesterol levels were increased or decreased utilizing cholesterol-chelating methyl-β-cyclodextrin (MβCD), which can either extract membrane cholesterol or add exogenous membrane cholesterol depending on whether MβCD is preloaded with cholesterol. Human PBEos were pretreated with MβCD (cholesterol removal) or MβCD+Cholesterol (MβCD+Chol; cholesterol delivery); subsequent IL-5-stimulated signaling and physiological endpoints were assessed. MβCD reduced membrane cholesterol in PBEos, and attenuated an IL-5-stimulated p38 and extracellular-regulated kinase 1/2 phosphorylation (p-p38, p-ERK1/2), and an IL-5-dependent increase in interleukin-1β (IL-1β) mRNA levels. In contrast, MβCD+Chol treatment elevated PBEos membrane cholesterol levels and basal p-p38, but did not alter IL-5-stimulated phosphorylation of ERK1/2, STAT5, or STAT3. Furthermore, MβCD+Chol pretreatment attenuated an IL-5-induced increase in cell survival at 48 hours, measured as total cellular metabolism. The reduction in cell survival following cholesterol addition despite unaltered STAT phosphorylation contradicts the current dogma in which JAK/STAT activation is sufficient to promote eosinophil survival, and suggests an additional, unidentified mechanism critically regulates IL-5-mediated human PBEos survival.
Collapse
Affiliation(s)
- Mandy E. Burnham
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Stephane Esnault
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Elon C. Roti Roti
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Mary E. Bates
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Paul J. Bertics
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| | - Loren C. Denlinger
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI, United States of America
| |
Collapse
|
49
|
Martin RA, Hodgkins SR, Dixon AE, Poynter ME. Aligning mouse models of asthma to human endotypes of disease. Respirology 2014; 19:823-33. [PMID: 24811131 PMCID: PMC4107015 DOI: 10.1111/resp.12315] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 03/01/2014] [Accepted: 03/28/2014] [Indexed: 12/20/2022]
Abstract
Substantial gains in understanding the pathophysiologic mechanisms underlying asthma have been made using preclinical mouse models. However, because asthma is a complex, heterogeneous syndrome that is rarely due to a single allergen and that often presents in the absence of atopy, few of the promising therapeutics that demonstrated effectiveness in mouse models have translated into new treatments for patients. This has resulted in an urgent need to characterize T helper (Th) 2-low, non-eosinophilic subsets of asthma, to study models that are resistant to conventional treatments such as corticosteroids and to develop therapies targeting patients with severe disease. Classifying asthma based on underlying pathophysiologic mechanisms, known as endotyping, offers a stratified approach for the development of new therapies for asthma. In preclinical research, new models of asthma are being utilized that more closely resemble the clinical features of different asthma endotypes, including the presence of interleukin-17 and a Th17 response, a biomarker of severe disease. These models utilize more physiologically relevant sensitizing agents, exacerbating factors and allergens, as well as incorporate time points that better reflect the natural history and chronicity of clinical asthma. Importantly, some models better represent non-classical asthma endotypes that facilitate the study of non-Th2-driven pathology and resemble the complex nature of clinical asthma, including corticosteroid resistance. Placing mouse asthma models into the context of human asthma endotypes will afford a more relevant approach to the understanding of pathophysiological mechanisms of disease that will afford the development of new therapies for those asthmatics that remain difficult to treat.
Collapse
Affiliation(s)
- Rebecca A Martin
- Vermont Lung Center, Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, Burlington, Vermont, USA
| | | | | | | |
Collapse
|
50
|
Chen B, Moore TV, Li Z, Sperling AI, Zhang C, Andrade J, Rodriguez A, Bahroos N, Huang Y, Morrisey EE, Gruber PJ, Solway J. Gata5 deficiency causes airway constrictor hyperresponsiveness in mice. Am J Respir Cell Mol Biol 2014; 50:787-95. [PMID: 24199649 DOI: 10.1165/rcmb.2013-0294oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Gata5 is a transcription factor expressed in the lung, but its physiological role is unknown. To test whether and how Gata5 regulates airway constrictor responsiveness, we studied Gata5(-/-), Gata5(+/-), and wild-type mice on the C57BL/6J background. Cholinergic airway constrictor responsiveness was assessed invasively in mice without and with induction of allergic airway inflammation through ovalbumin sensitization and aerosol exposure. Gata5-deficient mice displayed native airway constrictor hyperresponsiveness (AHR) in the absence of allergen-induced inflammation. Gata5-deficient mice retained their relatively greater constrictor responsiveness even in ovalbumin-induced experimental asthma. Gata5 deficiency did not alter the distribution of cell types in bronchoalveolar lavage fluid, but bronchial epithelial mucus metaplasia was more prominent in Gata5(-/-) mice after allergen challenge. Gene expression profiles revealed that apolipoprotein E (apoE) was the fifth most down-regulated transcript in Gata5-deficient lungs, and quantitative RT-PCR and immunostaining confirmed reduced apoE expression in Gata5(-/-) mice. Quantitative RT-PCR also revealed increased IL-13 mRNA in the lungs of Gata5-deficient mice. These findings for the first time show that Gata5 regulates apoE and IL-13 expression in vivo and that its deletion causes AHR. Gata5-deficient mice exhibit an airway phenotype that closely resembles that previously reported for apoE(-/-) mice: both exhibit cholinergic AHR in native and experimental asthma states, and there is excessive goblet cell metaplasia after allergen sensitization and challenge. The Gata5-deficient phenotype also shares features that were previously reported for IL-13-treated mice. Together, these results indicate that Gata5 deficiency induces AHR, at least in part, by blunting apoE and increasing IL-13 expression.
Collapse
|