1
|
Liu J, Xu L, Lu J, Shen X, Li D, Bai L, Li X, Yu Z, Li H. Roles of Adam8 in Neuroinflammation in experimental ischemic Stroke: Insights from single-cell and ribosome-bound mRNA sequencing. Exp Neurol 2025; 388:115207. [PMID: 40064361 DOI: 10.1016/j.expneurol.2025.115207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/21/2025]
Abstract
Stroke remains a leading cause of global mortality, with neuroinflammation significantly exacerbating clinical outcomes. Microglia serve as key mediators of post-stroke neuroinflammation, though the mechanisms driving their migration to injury sites remain poorly understood. In this study, using publicly available single-cell sequencing data (GSE234052), we identified a migration-associated microglial subtype in a murine model of distal middle cerebral artery occlusion (dMCAO). Additionally, ribosome-bound mRNA sequencing data (GSE225110) from microglia isolated from peri-infarct cortical tissue uncovered dMCAO-induced alterations in microglial mRNA translation. By integrating these datasets, we identified A Disintegrin And Metalloproteinase 8 (Adam8) as a key gene upregulated at both the transcriptional and translational levels post-dMCAO. Protein analysis revealed that both the precursor and active forms of Adam8 were predominantly expressed in microglia and significantly upregulated in peri-infarct regions following dMCAO. Notably, Adam8 inhibition with BK-1361 significantly reduced Adam8 cleavage, M1 microglial migration, inflammation, infarct size, and improved neurological outcomes. Bioinformatics analysis further identified Myo1e as a potential interacting partner of Adam8, a finding validated through immunofluorescence co-localization. These findings highlight Adam8 as a promising therapeutic target for mitigating post-stroke neuroinflammation and offer new insights into the mechanisms of microglial migration.
Collapse
Affiliation(s)
- Jiale Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Li Xu
- Intensive Care Unit of the Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xi Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Di Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
2
|
Garcia FM, de Sousa VP, Silva-Dos-Santos PPE, Fernandes IS, Serpa FS, de Paula F, Mill JG, Bueno MRP, Errera FIV. Copy Number Variation in Asthma: An Integrative Review. Clin Rev Allergy Immunol 2025; 68:4. [PMID: 39755867 DOI: 10.1007/s12016-024-09015-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 01/06/2025]
Abstract
Asthma is a complex disease with varied clinical manifestations resulting from the interaction between environmental and genetic factors. While chronic airway inflammation and hyperresponsiveness are central features, the etiology of asthma is multifaceted, leading to a diversity of phenotypes and endotypes. Although most research into the genetics of asthma focused on the analysis of single nucleotide polymorphisms (SNPs), studies highlight the importance of structural variations, such as copy number variations (CNVs), in the inheritance of complex characteristics, but their role has not yet been fully elucidated in asthma. In this context, an integrative review was conducted to identify the genes and pathways involved, the location, size, and classes of CNVs, as well as their contribution to asthma risk, severity, control, and response to treatment. As a result of the review, 16 articles were analyzed, from different types of observational studies, such as case-control, cohort studies and genotyped-proband or trios design, that have been carried out in populations from different countries, ethnicities, and ages. Chromosomes 12 and 17 were the most studied in three publications each. CNVs located on 12 chromosomes were associated with asthma, the majority being found on chromosome 6p and 17q, of the deletion type, encompassing 30 different coding-protein genes and one pseudogene region. Six genes with CNVs were identified as significant expression quantitative locus (eQTLs) with mean expression in asthma-related tissues, such as the lung and whole blood. The phenotypic variability of asthma may hinder the clinical application of these findings, but the research shows the importance of investigating these genetic variations as possible biomarkers in asthma patients.
Collapse
Affiliation(s)
- Fernanda Mariano Garcia
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil.
| | - Valdemir Pereira de Sousa
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Priscila Pinto E Silva-Dos-Santos
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Izadora Silveira Fernandes
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Faradiba Sarquis Serpa
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
| | - Flávia de Paula
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Maria Rita Passos Bueno
- Department of Genetics and Evolutionary Biology, University of São Paulo (USP), São Paulo, São Paulo, Brazil
- Human Genome and Stem Cell Research Center, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Flávia Imbroisi Valle Errera
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| |
Collapse
|
3
|
Liu P, Liu M, Sun Y, Lei W, Xu Y. Assessing the predictive potential of ADAM8 for disease control in chronic rhinosinusitis with nasal polyps. FRONTIERS IN ALLERGY 2024; 5:1488441. [PMID: 39588470 PMCID: PMC11586353 DOI: 10.3389/falgy.2024.1488441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/30/2024] [Indexed: 11/27/2024] Open
Abstract
Background A disintegrin and metalloproteinase 8 (ADAM8) has been implicated in eosinophilic inflammation; however, its role in chronic rhinosinusitis with nasal polyps (CRSwNP) remains to be elucidated. This study aimed to investigate the predictive significance of ADAM8 levels in nasal secretions for the endotypes and disease control status of CRSwNP. Methods A cohort comprising 120 CRSwNP patients and 45 healthy controls (HCs) was assembled, delineating 53 non-eosinophilic CRSwNP (neCRSwNP) and 67 eosinophilic CRSwNP (eCRSwNP) patients. Immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) were utilized to measure ADAM8 levels in nasal mucosal tissues and secretions from all participants. The receiver operating characteristic (ROC) curves and Pearson correlation analysis were employed to assess the predictive capability of ADAM8 levels in predictiving CRSwNP endotypes and disease control status. Results ADAM8 levels in nasal secretions were elevated in CRSwNP patients compared to HCs, with a more pronounced increase observed in eCRSwNP patients. Elevated ADAM8 concentrations in nasal secretions were positively correlated with peripheral blood eosinophil counts and percentages, tissue eosinophil counts, serum total IgE, Lund-Mackay scores, and Lund-Kennedy scores. Ultimately, 103 CRSwNP patients completed the follow-up protocol, with 72 classified as the controlled group and 31 as the uncontrolled group. Uncontrolled CRSwNP patients exhibited significantly higher ADAM8 levels in nasal secretions compared to the controlled group. The ROC curves indicated that ADAM8 in nasal secretions exhibits robust discriminatory capacity for eCRSwNP and postoperative disease control status. Conclusion ADAM8 in nasal secretions emerges as a potential novel biomarker for the prognostication of CRSwNP endotypes and the postoperative disease control status.
Collapse
Affiliation(s)
- Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meng Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yibin Sun
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiwei Lei
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, China
| |
Collapse
|
4
|
Cook L, Gharzia FG, Bartsch JW, Yildiz D. A jack of all trades - ADAM8 as a signaling hub in inflammation and cancer. FEBS J 2024; 291:3989-4008. [PMID: 38097912 DOI: 10.1111/febs.17034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
As a member of the family of A Disintegrin And Metalloproteinases (ADAM) ADAM8 is preferentially expressed in lymphatic organs, immune cells, and tumor cells. The substrate spectrum for ADAM8 proteolytic activity is not exclusive but is related to effectors of inflammation and signaling in the tumor microenvironment. In addition, complexes of ADAM8 with extracellular binding partners such as integrin β-1 cause an extensive intracellular signaling in tumor cells, thereby activating kinase pathways with STAT3, ERK1/2, and Akt signaling, which causes increased cell survival and enhanced motility. The cytoplasmic domain of ADAM8 harbors five SRC homology-3 (SH3) domains that can potentially interact with several proteins involved in actin dynamics and cell motility, including Myosin 1F (MYO1F), which is essential for neutrophil motility. The concept of ADAM8 thus involves immune cell recruitment, in most cases leading to an enhancement of inflammatory (asthma, COPD) and tumor (including pancreatic and breast cancers) pathologies. In this review, we report on available studies that qualify ADAM8 as a therapeutic target in different pathologies. As a signaling hub, ADAM8 controls extracellular, intracellular, and intercellular communication, the latter one mainly mediated by the release of extracellular vesicles with ADAM8 as cargo. Here, we will dissect the contribution of different domains to these distinct ways of communication in several pathologies. We conclude that therapeutic targeting attempts for ADAM8 should consider blocking more than a single domain and that this requires a thorough evaluation of potent molecules targeting ADAM8 in an in vivo setting.
Collapse
Affiliation(s)
- Lena Cook
- Department of Neurosurgery, Philipps University Marburg, Germany
| | - Federico Guillermo Gharzia
- Experimental and Clinical Pharmacology and Toxicology Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Germany
| | - Daniela Yildiz
- Experimental and Clinical Pharmacology and Toxicology Center for Molecular Signaling (PZMS), Saarland University, Homburg, Germany
| |
Collapse
|
5
|
Qian Z, Li R, Zhao T, Xie K, Li P, Li G, Shen N, Gong J, Hong X, Yang L, Li H. Blockade of the ADAM8-Fra-1 complex attenuates neuroinflammation by suppressing the Map3k4/MAPKs axis after spinal cord injury. Cell Mol Biol Lett 2024; 29:75. [PMID: 38755530 PMCID: PMC11100242 DOI: 10.1186/s11658-024-00589-3] [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: 02/05/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Mechanical spinal cord injury (SCI) is a deteriorative neurological disorder, causing secondary neuroinflammation and neuropathy. ADAM8 is thought to be an extracellular metalloproteinase, which regulates proteolysis and cell adherence, but whether its intracellular region is involved in regulating neuroinflammation in microglia after SCI is unclear. METHODS Using animal tissue RNA-Seq and clinical blood sample examinations, we found that a specific up-regulation of ADAM8 in microglia was associated with inflammation after SCI. In vitro, microglia stimulated by HMGB1, the tail region of ADAM8, promoted microglial inflammation, migration and proliferation by directly interacting with ERKs and Fra-1 to promote activation, then further activated Map3k4/JNKs/p38. Using SCI mice, we used BK-1361, a specific inhibitor of ADAM8, to treat these mice. RESULTS The results showed that administration of BK-1361 attenuated the level of neuroinflammation and reduced microglial activation and recruitment by inhibiting the ADAM8/Fra-1 axis. Furthermore, treatment with BK-1361 alleviated glial scar formation, and also preserved myelin and axonal structures. The locomotor recovery of SCI mice treated with BK-1361 was therefore better than those without treatment. CONCLUSIONS Taken together, the results showed that ADAM8 was a critical molecule, which positively regulated neuroinflammatory development and secondary pathogenesis by promoting microglial activation and migration. Mechanically, ADAM8 formed a complex with ERK and Fra-1 to further activate the Map3k4/JNK/p38 axis in microglia. Inhibition of ADAM8 by treatment with BK-1361 decreased the levels of neuroinflammation, glial formation, and neurohistological loss, leading to favorable improvement in locomotor functional recovery in SCI mice.
Collapse
Affiliation(s)
- Zhanyang Qian
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, China
| | - Rulin Li
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- School of Postgraduate, Dalian Medical University, Dalian, China
| | - Tianyu Zhao
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- School of Postgraduate, Dalian Medical University, Dalian, China
| | - Kunxin Xie
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - PengFei Li
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- School of Postgraduate, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guangshen Li
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Na Shen
- School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Jiamin Gong
- School of Basic Medicine, Nanjing Medical University, Nanjing, China
| | - Xin Hong
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, China
| | - Lei Yang
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| | - Haijun Li
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| |
Collapse
|
6
|
Li D, Wang T, Ma Q, Zhou L, Le Y, Rao Y, Jin L, Pei Y, Cheng Y, Huang C, Gai X, Sun Y. IL-17A Promotes Epithelial ADAM9 Expression in Cigarette Smoke-Related COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:2589-2602. [PMID: 36267325 PMCID: PMC9578481 DOI: 10.2147/copd.s375006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022] Open
Abstract
Background It has been reported that a disintegrin and metalloproteinase 9 (ADAM9) is involved in the pathogenesis of cigarette smoke (CS)-associated chronic obstructive pulmonary disease (COPD). But how CS exposure leads to upregulation of ADAM9 remains unknown. Methods Patients who underwent lobectomy for a solitary pulmonary nodule were enrolled and divided into three groups: non-smokers with normal lung function, smokers without COPD and smoker patients with COPD. Immunoreactivity of interleukin (IL)-17A and ADAM9 in small airways and alveolar walls was measured by immunohistochemistry. Wild-type and Il17a−/− C57BL/6 mice were exposed to CS for six months, and ADAM9 expression in the airway epithelia was measured by immunoreactivity. In addition, the protein and mRNA expression levels of IL-17A and ADAM9 were assessed in CS extract (CSE) and/or IL-17A-treated human bronchial epithelial (HBE) cells. Results The immunoreactivity of ADAM9 was increased in the airway epithelia and alveolar walls of patients with COPD compared to that of the controls. The expression of IL-17A was also upregulated in airway epithelial cells of patients with COPD and correlated positively with the level of ADAM9. The results from the animal model showed that Il17a−/− mice were protected from emphysema induced by CS exposure, together with a reduced level of ADAM9 expression in the airway epithelia, suggesting a possible link between ADAM9 and IL-17A. Consistently, our in vitro cell model showed that CSE stimulated the expression of ADAM9 and IL-17A in HBE cells in a dose- and time-dependent manner. Recombinant IL-17A induced ADAM9 upregulation in HBE cells and had a synergistic effect with CSE, whereas blocking IL-17A inhibited CSE-induced ADAM9 expression. Further analysis revealed that IL-17A induced c-Jun N-terminal kinase (JNK) phosphorylation, thereby increasing ADAM9 expression. Conclusion Our results revealed a novel role of IL-17A in CS-related COPD, where IL-17A contributes to ADAM9 expression by activating JNK signaling.
Collapse
Affiliation(s)
- Danyang Li
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Tong Wang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Qianli Ma
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Liang Jin
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yuqiang Pei
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Yaning Cheng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, People’s Republic of China
| | - Chen Huang
- Center of Basic Medical Research, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| | - Xiaoyan Gai
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China,Correspondence: Xiaoyan Gai; Yongchang Sun, Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, People’s Republic of China, Email ;
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People’s Republic of China
| |
Collapse
|
7
|
Zhang Y, Tian Z, Gerard D, Yao L, Shofer FS, Cs-Szabo G, Qin L, Pacifici M, Enomoto-Iwamoto M. Elevated inflammatory gene expression in intervertebral disc tissues in mice with ADAM8 inactivated. Sci Rep 2021; 11:1804. [PMID: 33469101 PMCID: PMC7815795 DOI: 10.1038/s41598-021-81495-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
We found ADAM8 enzymatic activity elevated in degenerative human intervertebral disc (IVD). Here, we examined the discs in ADAM8-inactivation mice that carry a mutation preventing self-activation of the enzyme. Surprisingly, elevated gene expression for inflammatory markers (Cxcl1, IL6) was observed in injured discs of ADAM8 mutant mice, along with elevated expression of type 2 collagen gene (Col2a1), compared with wild type controls. Injured annulus fibrosus of mutant and wild type mice contained a higher proportion of large collagen fibers compared with intact discs, as documented by microscopic examination under circular polarized light. In the intact IVDs, Adam8EQ mouse AF contained lower proportion of yellow (intermediate) fiber than WT mice. This suggests that ADAM8 may regulate inflammation and collagen fiber assembly. The seemingly contradictory findings of elevated inflammatory markers in mutant mice and excessive ADAM8 activity in human degenerative discs suggest that ADAM8 may interact with other enzymatic and pro-inflammatory processes needed for tissue maintenance and repair. As a future therapeutic intervention to retard intervertebral disc degeneration, partial inhibition of ADAM8 proteolysis may be more desirable than complete inactivation of this enzyme.
Collapse
Affiliation(s)
- Yejia Zhang
- Department of Physical Medicine & Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
- Translational Musculoskeletal Research Center (TMRC), Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA.
| | - Zuozhen Tian
- Department of Physical Medicine & Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David Gerard
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Lutian Yao
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Department of Orthopaedics/Sports Medicine and Joint Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | - Frances S Shofer
- Emergency Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Gabriella Cs-Szabo
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Ling Qin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Maurizio Pacifici
- Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, USA
| | | |
Collapse
|
8
|
Charting Extracellular Transcriptomes in The Human Biofluid RNA Atlas. Cell Rep 2020; 33:108552. [PMID: 33378673 DOI: 10.1016/j.celrep.2020.108552] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/14/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular RNAs present in biofluids have emerged as potential biomarkers for disease. Where most studies focus on blood-derived fluids, other biofluids may be more informative. We present an atlas of messenger, circular, and small RNA transcriptomes of a comprehensive collection of 20 human biofluids. By means of synthetic spike-in controls, we compare RNA content across biofluids, revealing a 10,000-fold difference in concentration. The circular RNA fraction is increased in most biofluids compared to tissues. Each biofluid transcriptome is enriched for RNA molecules derived from specific tissues and cell types. Our atlas enables an informed selection of the most relevant biofluid to monitor particular diseases. To verify the biomarker potential in these biofluids, four validation cohorts representing a broad spectrum of diseases were profiled, revealing numerous differential RNAs between case and control subjects. Spike-normalized data are publicly available in the R2 web portal for further exploration.
Collapse
|
9
|
Hachim MY, Elemam NM, Ramakrishnan RK, Salameh L, Olivenstein R, Hachim IY, Venkatachalam T, Mahboub B, Al Heialy S, Halwani R, Hamid Q, Hamoudi R. Blood and Salivary Amphiregulin Levels as Biomarkers for Asthma. Front Med (Lausanne) 2020; 7:561866. [PMID: 33195308 PMCID: PMC7659399 DOI: 10.3389/fmed.2020.561866] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Background: Amphiregulin (AREG) expression in asthmatic airways and sputum was shown to increase and correlate with asthma. However, no studies were carried out to evaluate the AREG level in blood and saliva of asthmatic patients. Objective: To measure circulating AREG mRNA and protein concentrations in blood, saliva, and bronchial biopsies samples from asthmatic patients. Methods: Plasma and Saliva AREG protein concentrations were measured using ELISA while PBMCs, and Saliva mRNA expression was measured by RT qPCR in non-severe, and severe asthmatic patients compared to healthy controls. Primary asthmatic bronchial epithelial cells and fibroblasts were assessed for AREG mRNA expression and released soluble AREG in their conditioned media. Tissue expression of AREG was evaluated using immunohistochemistry of bronchial biopsies from asthmatic patients and healthy controls. Publicly available transcriptomic databases were explored for the global transcriptomic profile of bronchial epithelium, and PBMCs were explored for AREG expression in asthmatic vs. healthy controls. Results: Asthmatic patients had higher AREG protein levels in blood and saliva compared to control subjects. Higher mRNA expression in saliva and primary bronchial epithelial cells plus higher AREG immunoreactivity in bronchial biopsies were also observed. Both blood and saliva AREG levels showed positive correlations with allergic rhinitis status, atopy status, eczema status, plasma periostin, neutrophilia, Montelukast sodium use, ACT score, FEV1, and FEV1/FVC. In silico analysis showed that severe asthmatic bronchial epithelium with high AREG gene expression is associated with higher neutrophils infiltration. Conclusion: AREG levels measured in a minimally invasive blood sample and a non-invasive saliva sample are higher in non-allergic severe asthma. CLINICAL IMPLICATIONS This is the first report to show the higher level of AREG levels in blood and saliva of non-allergic severe asthma.
Collapse
Affiliation(s)
- Mahmood Yaseen Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Noha Mousaad Elemam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rakhee K. Ramakrishnan
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Laila Salameh
- Rashid Hospital, Dubai Health Authority, Dubai, United Arab Emirates
| | | | - Ibrahim Yaseen Hachim
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Thenmozhi Venkatachalam
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bassam Mahboub
- Rashid Hospital, Dubai Health Authority, Dubai, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Saba Al Heialy
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
| | - Rabih Halwani
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Sharjah Institute for Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, UCL, London, United Kingdom
| |
Collapse
|
10
|
Ning X, Zhang Y, Wu H, Bai L, Gong C, Wang Z. Genetic association of ADAM33 polymorphisms with childhood asthma in Chinese Han population: A case-control study. Medicine (Baltimore) 2019; 98:e17327. [PMID: 31626088 PMCID: PMC6824736 DOI: 10.1097/md.0000000000017327] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
To explore the association of a disintegrin and metalloprotease 33 (ADAM33) polymorphisms with childhood asthma susceptibility, we conducted this case-control study.In this case-control study, we selected 96 asthma children and 86 healthy children to conduct the genotyping of ADAM33 polymorphisms through polymerase chain reaction-direct sequencing (PCR-DS). Hardy-Weinberg equilibrium (HWE) status in the control group was detected adopting chi-square test. Frequency differences of genotypes, alleles, and haplotypes were compared by chi-square test between the case and control groups. Linkage disequilibrium (LD) between polymorphisms was checked using Haploview software. Association intensity of the polymorphisms with the disease risk was assessed by odds ratio (OR) and 95% confidence interval (95%CI).The frequency of rs678881 GA genotype was obviously higher in cases than in controls (P = .03) and the carriage of this genotype conferred higher risk of asthma among children than GG genotype (OR = 2.03, 95%CI = 1.05-3.91). However, neither rs2280089 nor rs2853209 polymorphism was significantly associated with the risk of childhood asthma. Strong LD was found among rs678881, rs2280089 and rs2853209, and haplotype GGT was distinctly associated with the risk of asthma in children (OR = 0.28, 95%CI = 0.13-0.57).ADAM33 rs678881 polymorphism is significantly correlated with increased susceptibility to asthma in Chinese Han children. Besides, haplotype GGT among the 3 polymorphisms was obviously associated with decreased risk of childhood asthma.
Collapse
Affiliation(s)
| | - Yunxia Zhang
- Pediatric Intensive Care Unit, Xingtai People's Hospital, Hebei Xingtai, China
| | | | | | | | | |
Collapse
|
11
|
Cho JL, Medoff BD. Chronic Obstructive Pulmonary Disease: Back to the Basics. Am J Respir Crit Care Med 2019; 198:1241-1242. [PMID: 30011221 DOI: 10.1164/rccm.201806-1148ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Josalyn L Cho
- 1 Division of Pulmonary, Critical Care and Occupational Medicine University of Iowa Iowa City, Iowa and
| | - Benjamin D Medoff
- 2 Division of Pulmonary and Critical Care Medicine Massachusetts General Hospital Boston, Massachusetts
| |
Collapse
|
12
|
Bright LA, Dittmar W, Nanduri B, McCarthy FM, Mujahid N, Costa LR, Burgess SC, Swiderski CE. Modeling the pasture-associated severe equine asthma bronchoalveolar lavage fluid proteome identifies molecular events mediating neutrophilic airway inflammation. VETERINARY MEDICINE-RESEARCH AND REPORTS 2019; 10:43-63. [PMID: 31119093 PMCID: PMC6504673 DOI: 10.2147/vmrr.s194427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/12/2019] [Indexed: 12/11/2022]
Abstract
Background: Pasture-associated severe equine asthma is a warm season, environmentally-induced respiratory disease characterized by reversible airway obstruction, persistent and non-specific airway hyper-responsiveness, and chronic neutrophilic airway inflammation. During seasonal exacerbation, signs vary from mild to life-threatening episodes of wheezing, coughing, and chronic debilitating labored breathing. Purpose: In human asthma, neutrophilic airway inflammation is associated with more severe and steroid-refractory asthma phenotypes, highlighting a need to decipher the mechanistic basis of this disease characteristic. We hypothesize that the collective biological activities of proteins in bronchoalveolar lavage fluid (BALF) of horses with pasture-associated severe asthma predict changes in neutrophil functions that contribute to airway neutrophilic inflammation. Methods: Using shotgun proteomics, we identified 1,003 unique proteins in cell-free BALF from six horses experiencing asthma exacerbation and six control herdmates. Contributions of each protein to ten neutrophil functions were modeled using manual biocuration to determine each protein’s net effect on the respective neutrophil functions. Results: A total of 417 proteins were unique to asthmatic horses, 472 proteins were unique to control horses (p<0.05), and 114 proteins were common in both groups. Proteins whose biological activities are responsible for increasing neutrophil migration, chemotaxis, cell spreading, transmigration, and infiltration, which would collectively bring neutrophils to airways, were over-represented in the BALF of asthmatic relative to control horses. By contrast, proteins whose biological activities support neutrophil activation, adhesion, phagocytosis, respiratory burst, and apoptosis, which would collectively shorten neutrophil lifespan, were under-represented in BALF of asthmatic relative to control horses. Interaction networks generated using Ingenuity® Pathways Analysis further support the results of our biocuration. Conclusion: Congruent with our hypothesis, the collective biological functions represented in differentially expressed proteins of BALF from horses with pasture-associated severe asthma support neutrophilic airway inflammation. This illustrates the utility of systems modeling to organize functional genomics data in a manner that characterizes complex molecular events associated with clinically relevant disease.
Collapse
Affiliation(s)
- Lauren A Bright
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Wellesley Dittmar
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Bindu Nanduri
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Fiona M McCarthy
- School of Animal Comparative and Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Nisma Mujahid
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Lais Rr Costa
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Shane C Burgess
- School of Animal Comparative and Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Cyprianna E Swiderski
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| |
Collapse
|
13
|
Polverino F, Rojas-Quintero J, Wang X, Petersen H, Zhang L, Gai X, Higham A, Zhang D, Gupta K, Rout A, Yambayev I, Pinto-Plata V, Sholl LM, Cunoosamy D, Celli BR, Goldring J, Singh D, Tesfaigzi Y, Wedzicha J, Olsson H, Owen CA. A Disintegrin and Metalloproteinase Domain-8: A Novel Protective Proteinase in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2018; 198:1254-1267. [PMID: 29750543 PMCID: PMC6290938 DOI: 10.1164/rccm.201707-1331oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 05/11/2018] [Indexed: 11/16/2022] Open
Abstract
RATIONALE ADAM8 (a disintegrin and metalloproteinase domain-8) is expressed by leukocytes and epithelial cells in health, but its contribution to the pathogenesis of chronic obstructive pulmonary disease (COPD) is unknown. OBJECTIVES To determine whether the expression of ADAM8 is increased in the lungs of patients with COPD and cigarette smoke (CS)-exposed mice, and whether ADAM8 promotes the development of COPD. METHODS ADAM8 levels were measured in lung, sputum, plasma, and/or BAL fluid samples from patients with COPD, smokers, and nonsmokers, and wild-type (WT) mice exposed to CS versus air. COPD-like lung pathologies were compared in CS-exposed WT versus Adam8-/- mice. MEASUREMENTS AND MAIN RESULTS ADAM8 immunostaining was reduced in macrophages, and alveolar and bronchial epithelial cells in the lungs of patients with COPD versus control subjects, and CS- versus air-exposed WT mice. ADAM8 levels were similar in plasma, sputum, and BAL fluid samples from patients with COPD and control subjects. CS-exposed Adam8-/- mice had greater airspace enlargement and airway mucus cell metaplasia than WT mice, but similar small airway fibrosis. CS-exposed Adam8-/- mice had higher lung macrophage counts, oxidative stress levels, and alveolar septal cell death rates, but lower alveolar septal cell proliferation rates and soluble epidermal growth factor receptor BAL fluid levels than WT mice. Adam8 deficiency increased lung inflammation by reducing CS-induced activation of the intrinsic apoptosis pathway in macrophages. Human ADAM8 proteolytically shed the epidermal growth factor receptor from bronchial epithelial cells to reduce mucin expression in vitro. Adam8 bone marrow chimera studies revealed that Adam8 deficiency in leukocytes and lung parenchymal cells contributed to the exaggerated COPD-like disease in Adam8-/- mice. CONCLUSIONS Adam8 deficiency increases CS-induced lung inflammation, emphysema, and airway mucus cell metaplasia. Strategies that increase or prolong ADAM8's expression in the lung may have therapeutic efficacy in COPD.
Collapse
Affiliation(s)
- Francesca Polverino
- Division of Pulmonary and Critical Care Medicine and
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | | | - Xiaoyun Wang
- Division of Pulmonary and Critical Care Medicine and
| | - Hans Petersen
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Li Zhang
- Division of Pulmonary and Critical Care Medicine and
| | - Xiaoyan Gai
- Division of Pulmonary and Critical Care Medicine and
| | - Andrew Higham
- Medicines Evaluation Unit, University of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Duo Zhang
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | | | - Amit Rout
- Division of Pulmonary and Critical Care Medicine and
| | | | | | - Lynette M. Sholl
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Danen Cunoosamy
- Respiratory, Inflammation and Autoimmunity Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Bartolomé R. Celli
- Division of Pulmonary and Critical Care Medicine and
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | | | - Dave Singh
- Medicines Evaluation Unit, University of South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | | | - Jadwiga Wedzicha
- Imperial College London, National Heart and Lung Institute, London, United Kingdom
| | - Henric Olsson
- Respiratory, Inflammation and Autoimmunity Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine and
- The Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| |
Collapse
|
14
|
|
15
|
Wang X, Polverino F, Rojas-Quintero J, Zhang D, Sánchez J, Yambayev I, Lindqvist E, Virtala R, Djukanovic R, Davies DE, Wilson S, O'Donnell R, Cunoosamy D, Hazon P, Higham A, Singh D, Olsson H, Owen CA. A Disintegrin and A Metalloproteinase-9 (ADAM9): A Novel Proteinase Culprit with Multifarious Contributions to COPD. Am J Respir Crit Care Med 2018; 198:1500-1518. [PMID: 29864380 PMCID: PMC6298633 DOI: 10.1164/rccm.201711-2300oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Proteinases with a disintegrin and a metalloproteinase domain (ADAMs) have not been well studied in COPD. We investigated whether ADAM9 is linked to COPD in humans and mice. METHODS ADAM9 blood and lung levels were measured in COPD patients versus controls, and air- versus cigarette smoke (CS)-exposed wild-type (WT) mice. WT and Adam9-/- mice were exposed to air or CS for 1-6 months, and COPD-like lung pathologies were measured. RESULTS ADAM9 staining was increased in lung epithelial cells and macrophages in smokers and even more so in COPD patients and correlated directly with pack-year smoking history and inversely with airflow obstruction and/or FEV1 % predicted. Bronchial epithelial cell ADAM9 mRNA levels were higher in COPD patients than controls and correlated directly with pack-year smoking history. Plasma, BALF and sputum ADAM9 levels were similar in COPD patients and controls. CS exposure increased Adam9 levels in WT murine lungs. Adam9-/- mice were protected from emphysema development, small airway fibrosis, and airway mucus metaplasia. CS-exposed Adam9-/- mice had reduced lung macrophage counts, alveolar septal cell apoptosis, lung elastin degradation, and shedding of VEGFR2 and EGFR in BALF samples. Recombinant ADAM9 sheds EGF and VEGF receptors from epithelial cells to reduce activation of the Akt pro-survival pathway and increase cellular apoptosis. CONCLUSIONS ADAM9 levels are increased in COPD lungs and linked to key clinical variables. Adam9 promotes emphysema development, and large and small airway disease in mice. Inhibition of ADAM9 could be a therapeutic approach for multiple COPD phenotypes.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Brigham and Women's Hospital, 1861, Boston, Massachusetts, United States
| | - Francesca Polverino
- Brigham and Women's Hospital, Harvard Medical School, Medicine, Boston, Massachusetts, United States
| | - Joselyn Rojas-Quintero
- Brigham and Women's Hospital, Harvard Medical School, Medicine, Boston, Massachusetts, United States
| | - Duo Zhang
- Boston University, 1846, Boston, Massachusetts, United States
| | - José Sánchez
- AstraZeneca R&D, Quantitative Biology, Discovery Sciences, Gothenburgh, Sweden
| | - Ilyas Yambayev
- Brigham and Women's Hospital, 1861, Boston, Massachusetts, United States
| | - Eva Lindqvist
- AstraZeneca R&D , Department of Translational Biology, Respiratory, Inflammation & Autoimmunity IMED, Gothenburg, Sweden
| | - Robert Virtala
- AstraZeneca R&D , Department of Translational Biology, Respiratory, Inflammation & Autoimmunity IMED, Gothenburg, Sweden
| | - Ratko Djukanovic
- Southampton University, Clinical and Experimental Sciences and Southampton NIHR Respiratory Biomedical Research Unit, Southampton, United Kingdom of Great Britain and Northern Ireland
| | - Donna E Davies
- Brooke Laboratory, Infection, Inflammation & Repair, Southampton, Hampshire, United Kingdom of Great Britain and Northern Ireland
| | - Susan Wilson
- University of Southampton, 7423, Southampton, United Kingdom of Great Britain and Northern Ireland
| | | | - Danen Cunoosamy
- AstraZeneca, Respiratory, Inflammation and Autoimmune iMed, Molndal, Sweden
| | - Petra Hazon
- AstraZeneca R&D , Department of Translational Biology, Respiratory, Inflammation & Autoimmunity IMED, Gothenburg, Sweden
| | - Andrew Higham
- University of South Manchester NHS Foundation Trust, Medicines Evaluation Unit, Manchester, United Kingdom of Great Britain and Northern Ireland
| | - Dave Singh
- North West Lung Research Centre, Manchester, United Kingdom of Great Britain and Northern Ireland
| | - Henric Olsson
- AstraZeneca R&D , Department of Translational Biology, Respiratory, Inflammation & Autoimmunity IMED, Gothenburg, Sweden
| | - Caroline A Owen
- Brigham and Women's Hospital, Boston, Massachusetts, United States ;
| |
Collapse
|
16
|
Bagnasco D, Ferrando M, Caminati M, Bragantini A, Puggioni F, Varricchi G, Passalacqua G, Canonica GW. Targeting Interleukin-5 or Interleukin-5Rα: Safety Considerations. Drug Saf 2018; 40:559-570. [PMID: 28321782 DOI: 10.1007/s40264-017-0522-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Asthma is a highly prevalent chronic disease of the airways; approximately 10% of patients with asthma will experience a severe form of the disease. New understanding of the pathogenesis of asthma has enabled the development of novel drugs and provided hope for patients with asthma. Interleukin (IL)-5 and IL-5 receptor subunit α (IL-5-Rα) plays a crucial role in the development, maturation, and operation of eosinophils so were the first important therapeutic target of these new drugs. While the results of early clinical trials of these drugs were not promising, results improved once researchers discovered the drugs worked best in patients with high eosinophil levels. Patients treated with both anti-IL-5 and IL-5-Rα experienced significant decreases in exacerbations. Trials have also demonstrated promising safety profiles; adverse events have been few and frequently only observed with placebo or considered unrelated to the study drug. The positive efficacy and safety profiles of these drugs has led to trials with interesting results in other diseases that are also secondary to the action of eosinophils: Churg-Strauss syndrome, hypereosinophilic syndrome, nasal polyposis, chronic obstructive pulmonary disease, atopic dermatitis, and esophagitis. In this review, we explore the main clinical trials of anti-IL-5 and IL-5-Rα, both in asthma and in other pathologies, with particular reference to the interesting safety and efficacy results.
Collapse
Affiliation(s)
- Diego Bagnasco
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Matteo Ferrando
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Marco Caminati
- Allergy Unit, University Hospital of Verona, Verona, Italy
| | - Alice Bragantini
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Francesca Puggioni
- Respiratory Disease Clinic, Department of Internal Medicine, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy
| | - Gilda Varricchi
- Division of Clinical Immunology and Allergy, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Giovanni Passalacqua
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - Giorgio Walter Canonica
- Allergy and Respiratory Diseases, Department of Internal Medicine, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy.
- Respiratory Disease Clinic, Department of Internal Medicine, IRCCS Humanitas Clinical and Research Center, Humanitas University, Milan, Italy.
| |
Collapse
|
17
|
da Silva Antunes R, Mehta AK, Madge L, Tocker J, Croft M. TNFSF14 (LIGHT) Exhibits Inflammatory Activities in Lung Fibroblasts Complementary to IL-13 and TGF-β. Front Immunol 2018; 9:576. [PMID: 29616048 PMCID: PMC5868327 DOI: 10.3389/fimmu.2018.00576] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 03/07/2018] [Indexed: 12/21/2022] Open
Abstract
The cytokine TNFSF14 [homologous to Lymphotoxin, exhibits Inducible expression and competes with HSV Glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes (LIGHT)] has been shown in mouse models to be important for development of lung tissue remodeling that is characteristic of asthma, idiopathic pulmonary fibrosis (IPF), and systemic sclerosis (SSc). However, its cellular targets are not fully delineated. In the present report, we show that LTβR and HVEM, the receptors for LIGHT, are constitutively expressed in primary human lung fibroblasts (HLFs). We asked whether LIGHT could promote inflammatory and remodeling-relevant activity in HLFs and how this was similar to, or distinct from, IL-13 or TGF-β, two cytokines strongly implicated in the pathogenesis of asthma, IPF, and SSc. Accumulation of myofibroblasts expressing alpha smooth muscle actin is a feature of lung inflammatory diseases. LIGHT promoted cell cycle progression and proliferation of HLFs, but not alpha smooth muscle actin expression. In contrast, TGF-β upregulated alpha smooth muscle actin but did not drive their proliferation. LIGHT also increased the gene or protein expression of a number of proinflammatory mediators, including ICAM-1 and VCAM-1, IL-6 and GM-CSF, the chemokines CCL5 and 20, and CXCL5, 11, and 12, and lung remodeling-associated proteinases MMP-9 and ADAM8. These were dependent on LTβR but not HVEM. LIGHT displayed overlapping and synergistic activities with IL-13 for a number of the activities, but LIGHT additionally enhanced the gene expression of several molecules, including the innate cytokines IL-33 and TSLP, which were not upregulated by IL-13. Our results highlight the varied and pleiotropic effects of LIGHT in HLFs. LIGHT might then be a therapeutic target for modulation of inflammation and remodeling associated with asthma and other similar diseases of the lung that involve fibroblasts.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Amit K Mehta
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Lisa Madge
- Janssen Research and Development, LLC, Immunology Discovery Research, Spring House, PA, United States
| | - Joel Tocker
- Janssen Research and Development, LLC, Immunology Discovery Research, Spring House, PA, United States
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States.,Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
18
|
Uwaezuoke SN, Ayuk AC, Eze JN. Severe bronchial asthma in children: a review of novel biomarkers used as predictors of the disease. J Asthma Allergy 2018; 11:11-18. [PMID: 29398922 PMCID: PMC5774744 DOI: 10.2147/jaa.s149577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Severe asthma or therapy-resistant asthma in children is a heterogeneous disease that affects all age-groups. Given its heterogeneity, precision in diagnosis and treatment has become imperative, in order to achieve better outcomes. If one is thus able to identify specific patient phenotypes and endotypes using the appropriate biomarkers, it will assist in providing the patient with more personalized and appropriate treatment. However, there appears to be a huge diagnostic gap in severe asthma, as there is no single test yet that accurately determines disease phenotype. In this paper, we review the published literature on some of these biomarkers and their possible role in bridging this diagnostic gap. We also highlight the cellular and molecular mechanisms involved in severe asthma, in order to show the basis for the novel biomarkers. Some markers useful for monitoring therapy and assessing airway remodeling in the disease are also discussed. A review of the literature was conducted with PubMed to gather baseline data on the subject. The literature search extended to articles published within the last 40 years. Although biomarkers specific to different severe asthma phenotypes have been identified, progress in their utility remains slow, because of several disease mechanisms, the variation of biomarkers at different levels of inflammation, changes in relying on one test over time (eg, from sputum eosinophilia to blood eosinophilia), and the degree of invasive tests required to collect biomarkers, which limits their applicability in clinical settings. In conclusion, several biomarkers remain useful in recognizing various asthma phenotypes. However, due to disease heterogeneity, identification and utilization of ideal and defined biomarkers in severe asthma are still inconclusive. The development of novel serum/sputum-based biomarker panels with enhanced sensitivity and specificity may lead to prompt diagnosis of the disease in the future.
Collapse
Affiliation(s)
- Samuel N Uwaezuoke
- Department of Pediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Nigeria
| | - Adaeze C Ayuk
- Department of Pediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Nigeria
| | - Joy N Eze
- Department of Pediatrics, University of Nigeria Teaching Hospital, Ituku-Ozalla, Nigeria
| |
Collapse
|
19
|
Theodorou K, van der Vorst EPC, Gijbels MJ, Wolfs IMJ, Jeurissen M, Theelen TL, Sluimer JC, Wijnands E, Cleutjens JP, Li Y, Jansen Y, Weber C, Ludwig A, Bentzon JF, Bartsch JW, Biessen EAL, Donners MMPC. Whole body and hematopoietic ADAM8 deficiency does not influence advanced atherosclerotic lesion development, despite its association with human plaque progression. Sci Rep 2017; 7:11670. [PMID: 28916789 PMCID: PMC5601942 DOI: 10.1038/s41598-017-10549-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 08/11/2017] [Indexed: 01/18/2023] Open
Abstract
Although A Disintegrin And Metalloproteinase 8 (ADAM8) is not crucial for tissue development and homeostasis, it has been implicated in various inflammatory diseases by regulating processes like immune cell recruitment and activation. ADAM8 expression has been associated with human atherosclerosis development and myocardial infarction, however a causal role of ADAM8 in atherosclerosis has not been investigated thus far. In this study, we examined the expression of ADAM8 in early and progressed human atherosclerotic lesions, in which ADAM8 was significantly upregulated in vulnerable lesions. In addition, ADAM8 expression was most prominent in the shoulder region of human atherosclerotic lesions, characterized by the abundance of foam cells. In mice, Adam8 was highly expressed in circulating neutrophils and in macrophages. Moreover, ADAM8 deficient mouse macrophages displayed reduced secretion of inflammatory mediators. Remarkably, however, neither hematopoietic nor whole-body ADAM8 deficiency in mice affected atherosclerotic lesion size. Additionally, except for an increase in granulocyte content in plaques of ADAM8 deficient mice, lesion morphology was unaffected. Taken together, whole body and hematopoietic ADAM8 does not contribute to advanced atherosclerotic plaque development, at least in female mice, although its expression might still be valuable as a diagnostic/prognostic biomarker to distinguish between stable and unstable lesions.
Collapse
Affiliation(s)
- Kosta Theodorou
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Emiel P C van der Vorst
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion J Gijbels
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,Department of Molecular Genetics, CARIM, Maastricht University, Maastricht, The Netherlands.,Department of Medical Biochemistry, AMC, Amsterdam, Netherlands
| | - Ine M J Wolfs
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Mike Jeurissen
- Department of Molecular Genetics, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Thomas L Theelen
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Judith C Sluimer
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Erwin Wijnands
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Jack P Cleutjens
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Yu Li
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Biochemistry, CARIM, Maastricht University, Maastricht, Netherlands.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Jacob F Bentzon
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jörg W Bartsch
- Department of Neurosurgery, Philipps University Marburg, Marburg, Germany
| | - Erik A L Biessen
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen, Aachen, Germany
| | - Marjo M P C Donners
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
20
|
Chen JY, Lin CH, Chen BC. Hypoxia-induced ADAM 17 expression is mediated by RSK1-dependent C/EBPβ activation in human lung fibroblasts. Mol Immunol 2017. [PMID: 28646679 DOI: 10.1016/j.molimm.2017.06.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hypoxia was identified as a mediator of lung fibrosis in patients with chronic obstructive asthma (COA). Overexpression of a disintegrin and metalloproteinase 17 (ADAM 17) and connective tissue growth factor (CTGF) leads to development of tissue fibrosis. However, the signaling pathway in hypoxia-induced ADAM 17 expression remains poorly defined. In this study, we investigated the roles that ribosomal S-6 kinase 1 (RSK1)/CCAAT/enhancer-binding protein β (C/EBPβ)-dependent ADAM 17 expression plays in hypoxia-induced CTGF expression in human lung fibroblasts. We observed that hypoxia caused increases in ADAM 17 expression and ADAM 17-luciferase activity in WI-38 cells. Hypoxia-induced CTGF-luciferase activity and CTGF expression were reduced in cells transfected with small interfering (si)RNA of ADAM 17 in WI-38 cells. Moreover, hypoxia-induced ADAM 17 expression was reduced by RSK1 siRNA and C/EBPβ siRNA. Hypoxia caused time-dependent increases in RSK1 phosphorylation at Thr359/Ser363. Exposure of cells to hypoxia resulted in increased C/EBPβ phosphorylation at Thr266 and C/EBPβ-luciferase activity in time-dependent manners, and these effects were suppressed by RSK1 siRNA. Hypoxia induced recruitment of C/EBPβ to the ADAM 17 promoter. Furthermore, CTGF-luciferase activity induced by hypoxia was attenuated by RSK1 siRNA and C/EBPβ siRNA. These results suggest that hypoxia instigates the RSK1-dependent C/EBPβ signaling pathway, which in turn initiates binding of C/EBPβ to the ADAM 17 promoter and ultimately induces ADAM 17 expression in human lung fibroblasts. Moreover, RSK1/C/EBPβ-dependent ADAM 17 expression is involved in hypoxia-induced CTGF expression. Our results suggest possible therapeutic approaches for treating hypoxia-mediated lung fibrosis in COA.
Collapse
Affiliation(s)
- Jing-Yun Chen
- Gradual Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Huang Lin
- Gradual Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Bing-Chang Chen
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
21
|
Johansson MW, Khanna M, Bortnov V, Annis DS, Nguyen CL, Mosher DF. IL-5-stimulated eosinophils adherent to periostin undergo stereotypic morphological changes and ADAM8-dependent migration. Clin Exp Allergy 2017; 47:1263-1274. [PMID: 28378503 DOI: 10.1111/cea.12934] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/28/2017] [Accepted: 03/28/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND IL-5 causes suspended eosinophils to polarize with filamentous (F)-actin and granules at one pole and the nucleus in a specialized uropod, the "nucleopod," which is capped with P-selectin glycoprotein ligand-1 (PSGL-1). IL-5 enhances eosinophil adhesion and migration on periostin, an extracellular matrix protein upregulated in asthma by type 2 immunity mediators. OBJECTIVE Determine how the polarized morphology evolves to foster migration of IL-5-stimulated eosinophils on a surface coated with periostin. METHODS Blood eosinophils adhering to adsorbed periostin were imaged at different time points by fluorescent microscopy, and migration of eosinophils on periostin was assayed. RESULTS After 10 minutes in the presence of IL-5, adherent eosinophils were polarized with PSGL-1 at the nucleopod tip and F-actin distributed diffusely at the opposite end. After 30-60 minutes, the nucleopod had dissipated such that PSGL-1 was localized in a crescent or ring away from the cell periphery, and F-actin was found in podosome-like structures. The periostin layer, detected with monoclonal antibody Stiny-1, shown here to recognize the FAS1 4 module, was cleared in wide areas around adherent eosinophils. Clearance was attenuated by metalloproteinase inhibitors or antibodies to disintegrin metalloproteinase 8 (ADAM8), a major eosinophil metalloproteinase previously implicated in asthma pathogenesis. ADAM8 was not found in podosome-like structures, which are associated with proteolytic activity in other cell types. Instead, immunoblotting demonstrated proteoforms of ADAM8 that lack the cytoplasmic tail in the supernatant. Anti-ADAM8 inhibited migration of IL-5-stimulated eosinophils on periostin. CONCLUSIONS AND CLINICAL RELEVANCE Migrating IL-5-activated eosinophils on periostin exhibit loss of nucleopodal features and appearance of prominent podosomes along with clearance of the Stiny-1 periostin epitope. Migration and epitope clearance are both attenuated by inhibitors of ADAM8. We propose, therefore, that eosinophils remodel and migrate on periostin-rich extracellular matrix in the asthmatic airway in an ADAM8-dependent manner, making ADAM8 a possible therapeutic target.
Collapse
Affiliation(s)
- M W Johansson
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - M Khanna
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - V Bortnov
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - D S Annis
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - C L Nguyen
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA
| | - D F Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI, USA.,Department of Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
22
|
Hales BJ, Hizawa N, Jenmalm M, Sverremark-Ekström E, Wardlaw AJ. Developments in the field of allergy in 2014 through the eyes of Clinical and Experimental Allergy. Clin Exp Allergy 2016; 45:1723-45. [PMID: 26492197 DOI: 10.1111/cea.12663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of asthma continues to be a major topic of interest to our authors with reviews and original papers on the role of viruses, mechanisms of inflammation, biomarkers, and phenotypes of asthma being major topics. A number of papers described new treatments for asthma focusing on blocking the Th2 response reflecting the fact that two decades of work in this area is finally bearing fruit. The pathogenesis of chronic rhinosinusitis is a growing area of interest, but there has been less on the genetics of airways disease than in previous years possibly reflecting the degree of rigour (and therefore a smaller body of work), with which these sorts of studies are now being undertaken. There continues to be a wide range of papers dealing with mechanisms of allergic disease ranging from clinical-based studies to basic research and the use of in vivo animal models especially mice. As before, mechanisms and new approaches to immunotherapy are common themes. Several were published in the allergens section investigating modification of allergens to increase their effectiveness and reduce the risk of adverse events. Risk factors for allergic disease was a common theme in the epidemiology section and food allergy a common theme in clinical allergy with papers on the development of protocols to induce tolerance and attempts to find biomarkers to distinguish sensitization from allergic disease. This was another exciting year for the editors, and we hope the readers of the journal.
Collapse
Affiliation(s)
- B J Hales
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - N Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - M Jenmalm
- Unit of Autoimmunity and Immune Regulation, Division of Clinical Immunology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - E Sverremark-Ekström
- M.C., Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - A J Wardlaw
- Department of Infection Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester, UK.,Department of Respiratory Medicine, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
23
|
Davies ER, Kelly JFC, Howarth PH, Wilson DI, Holgate ST, Davies DE, Whitsett JA, Haitchi HM. Soluble ADAM33 initiates airway remodeling to promote susceptibility for allergic asthma in early life. JCI Insight 2016; 1. [PMID: 27489884 PMCID: PMC4968941 DOI: 10.1172/jci.insight.87632] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Asthma is a chronic inflammatory airways disease that usually begins in early life and involves gene-environment interactions. Although most asthma exhibits allergic inflammation, many allergic individuals do not have asthma. Here, we report how the asthma gene a disintegrin and metalloprotease 33 (ADAM33) acts as local tissue susceptibility gene that promotes allergic asthma. We show that enzymatically active soluble ADAM33 (sADAM33) is increased in asthmatic airways and plays a role in airway remodeling, independent of inflammation. Furthermore, remodeling and inflammation are both suppressed in Adam33-null mice after allergen challenge. When induced in utero or added ex vivo, sADAM33 causes structural remodeling of the airways, which enhances postnatal airway eosinophilia and bronchial hyperresponsiveness following subthreshold challenge with an aeroallergen. This substantial gene-environment interaction helps to explain the end-organ expression of allergic asthma in genetically susceptible individuals. Finally, we show that sADAM33-induced airway remodeling is reversible, highlighting the therapeutic potential of targeting ADAM33 in asthma. Loss of ADAM33 suppresses airway remodeling and allergic inflammation in mice, suggesting the therapeutic potential of targeting ADAM33 in asthma.
Collapse
Affiliation(s)
- Elizabeth R Davies
- The Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joanne F C Kelly
- The Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Peter H Howarth
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.; Institute for Life Sciences, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - David I Wilson
- Institute for Life Sciences, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.; Centre for Human Development, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stephen T Holgate
- National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.; Institute for Life Sciences, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Donna E Davies
- The Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.; National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.; Institute for Life Sciences, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Hans Michael Haitchi
- The Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.; Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.; National Institute for Health Research (NIHR) Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom.; Institute for Life Sciences, Stem Cells and Regeneration, Human Genetics, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
24
|
Froidure A, Mouthuy J, Durham SR, Chanez P, Sibille Y, Pilette C. Asthma phenotypes and IgE responses. Eur Respir J 2015; 47:304-19. [PMID: 26677936 DOI: 10.1183/13993003.01824-2014] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 10/14/2015] [Indexed: 01/18/2023]
Abstract
The discovery of IgE represented a major breakthrough in allergy and asthma research, whereas the clinical interest given to IgE in asthma has been blurred until the arrival of anti-IgE biotherapy. Novel facets of the complex link between IgE and asthma have been highlighted by the effect of this treatment and by basic research. In parallel, asthma phenotyping recently evolved to the concept of endotypes, relying on identified/suspected pathobiological mechanisms to phenotype patients, but has not yet clearly positioned IgE among biomarkers of asthma.In this review, we first summarise recent knowledge about the regulation of IgE production and its main receptor, FcεRI. In addition to allergens acting as classical IgE inducers, viral infections as well as air pollution may trigger the IgE pathway, notably resetting the threshold of IgE sensitivity by regulating FcεRI expression. We then analyse the place of IgE in different asthma endo/phenotypes and discuss the potential interest of IgE among biomarkers in asthma.
Collapse
Affiliation(s)
- Antoine Froidure
- Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, Université catholique de Louvain, Brussels and Walloon Institute for Excellence in Lifesciences and Biotechnology (WELBIO), Belgium Dept of Chest Medicine, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium These authors contributed equally to this work
| | - Jonathan Mouthuy
- Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, Université catholique de Louvain, Brussels and Walloon Institute for Excellence in Lifesciences and Biotechnology (WELBIO), Belgium Dept of Chest Medicine, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium These authors contributed equally to this work
| | - Stephen R Durham
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Pascal Chanez
- INSERM U 1067, CNRS UMR 7333 Aix Marseille Université and Dépt des Maladies Respiratoires, Assistance Publique des Hôpitaux de Marseille, Marseille, France
| | - Yves Sibille
- Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, Université catholique de Louvain, Brussels and Walloon Institute for Excellence in Lifesciences and Biotechnology (WELBIO), Belgium Dept of Chest Medicine, Centre Hospitalier Universitaire de Mont-Godinne, Université catholique de Louvain, Yvoir, Belgium
| | - Charles Pilette
- Institut de Recherche Expérimentale et Clinique, Pôle de Pneumologie, Université catholique de Louvain, Brussels and Walloon Institute for Excellence in Lifesciences and Biotechnology (WELBIO), Belgium Dept of Chest Medicine, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
25
|
De Ferrari L, Chiappori A, Bagnasco D, Riccio AM, Passalacqua G, Canonica GW. Molecular phenotyping and biomarker development: are we on our way towards targeted therapy for severe asthma? Expert Rev Respir Med 2015; 10:29-38. [PMID: 26566089 DOI: 10.1586/17476348.2016.1111763] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although different phenotypes of severe asthma can be identified, all are characterized by common symptoms. Due to their heterogeneity, they exhibit differences in pathogenesis, etiology and clinical responses to therapeutic approaches. The identification of distinct molecular phenotypes to define severe asthmatic patients will allow us to better understand the pathophysiology of the disease and thus to more precisely target the treatment for each patient. To achieve this goal, a systematic search for new, reliable and stable biomarkers specific for each phenotype is essential. This review focuses on the current known molecular phenotypes of severe asthma and highlights the need for biomarkers that could (either alone or in combination) be predictive of the treatment outcome.
Collapse
Affiliation(s)
- Laura De Ferrari
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| | - Alessandra Chiappori
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| | - Diego Bagnasco
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| | - Anna Maria Riccio
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| | - Giovanni Passalacqua
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| | - Giorgio Walter Canonica
- a Allergy and Respiratory Diseases, IRCCS San Martino-IST , University of Genoa , Genoa , Italy
| |
Collapse
|
26
|
Chiappori A, De Ferrari L, Folli C, Mauri P, Riccio AM, Canonica GW. Biomarkers and severe asthma: a critical appraisal. Clin Mol Allergy 2015; 13:20. [PMID: 26430389 PMCID: PMC4590266 DOI: 10.1186/s12948-015-0027-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 01/17/2023] Open
Abstract
Severe asthma (SA) is a clinically and etiologically heterogeneous respiratory disease which affects among 5–10 % of asthmatic patients. Despite high-dose therapy, a large patients percentage is not fully controlled and has a poor quality of life. In this review, we describe the biomarkers actually known in scientific literature and used in clinical practice for SA assessment and management: neutrophils, eosinophils, periostin, fractional exhaled nitric oxide, exhaled breath condensate and galectins. Moreover, we give an overview on clinical and biological features characterizing severe asthma, paying special attention to the potential use of these ones as reliable markers. We finally underline the need to define different biomarkers panels to select patients affected by severe asthma for specific and personalized therapeutic approach.
Collapse
Affiliation(s)
- Alessandra Chiappori
- DIMI-Department of Internal Medicine, Respiratory Diseases and Allergy Clinic, University of Genoa, IRCCS AOU S.Martino-IST, Genoa, Italy
| | - Laura De Ferrari
- DIMI-Department of Internal Medicine, Respiratory Diseases and Allergy Clinic, University of Genoa, IRCCS AOU S.Martino-IST, Genoa, Italy
| | - Chiara Folli
- DIMI-Department of Internal Medicine, Respiratory Diseases and Allergy Clinic, University of Genoa, IRCCS AOU S.Martino-IST, Genoa, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies, CNR, Segrate, Milan, Italy
| | - Anna Maria Riccio
- DIMI-Department of Internal Medicine, Respiratory Diseases and Allergy Clinic, University of Genoa, IRCCS AOU S.Martino-IST, Genoa, Italy
| | - Giorgio Walter Canonica
- DIMI-Department of Internal Medicine, Respiratory Diseases and Allergy Clinic, University of Genoa, IRCCS AOU S.Martino-IST, Genoa, Italy
| |
Collapse
|
27
|
da Silva Antunes R, Madge L, Soroosh P, Tocker J, Croft M. The TNF Family Molecules LIGHT and Lymphotoxin αβ Induce a Distinct Steroid-Resistant Inflammatory Phenotype in Human Lung Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26209626 DOI: 10.4049/jimmunol.1500356] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lung epithelial cells are considered important sources of inflammatory molecules and extracellular matrix proteins that contribute to diseases such as asthma. Understanding the factors that stimulate epithelial cells may lead to new insights into controlling lung inflammation. This study sought to investigate the responsiveness of human lung epithelial cells to the TNF family molecules LIGHT and lymphotoxin αβ (LTαβ). Bronchial and alveolar epithelial cell lines, and primary human bronchial epithelial cells, were stimulated with LIGHT and LTαβ, and expression of inflammatory cytokines and chemokines and markers of epithelial-mesenchymal transition and fibrosis/remodeling was measured. LTβ receptor, the receptor shared by LIGHT and LTαβ, was constitutively expressed on all epithelial cells. Correspondingly, LIGHT and LTαβ strongly induced a limited but highly distinct set of inflammatory genes in all epithelial cells tested, namely the adhesion molecules ICAM-1 and VCAM-1; the chemokines CCL5, CCL20, CXCL1, CXCL3, CXCL5, and CXCL11; the cytokines IL-6, activin A and GM-CSF; and metalloproteinases matrix metalloproteinase-9 and a disintegrin and metalloproteinase domain-8. Importantly, induction of the majority of these inflammatory molecules was insensitive to the suppressive effects of the corticosteroid budesonide. LIGHT and LTαβ also moderately downregulated E-cadherin, a protein associated with maintaining epithelial integrity, but did not significantly drive production of extracellular matrix proteins or α-smooth muscle actin. Thus, LIGHT and LTαβ induce a distinct steroid-resistant inflammatory signature in airway epithelial cells via constitutively expressed LTβ receptor. These findings support our prior murine studies that suggested the receptors for LIGHT and LTαβ contribute to development of lung inflammation characteristic of asthma and idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Lisa Madge
- Janssen Research and Development, Immunology Discovery Research, Spring House, PA 19002; and
| | | | - Joel Tocker
- Janssen Research and Development, Immunology Discovery Research, Spring House, PA 19002; and
| | - Michael Croft
- Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037;
| |
Collapse
|
28
|
Dreymueller D, Uhlig S, Ludwig A. ADAM-family metalloproteinases in lung inflammation: potential therapeutic targets. Am J Physiol Lung Cell Mol Physiol 2014; 308:L325-43. [PMID: 25480335 DOI: 10.1152/ajplung.00294.2014] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acute and chronic lung inflammation is driven and controlled by several endogenous mediators that undergo proteolytic conversion from surface-expressed proteins to soluble variants by a disintegrin and metalloproteinase (ADAM)-family members. TNF and epidermal growth factor receptor ligands are just some of the many substrates by which these proteases regulate inflammatory or regenerative processes in the lung. ADAM10 and ADAM17 are the most prominent members of this protease family. They are constitutively expressed in most lung cells and, as recent research has shown, are the pivotal shedding enzymes mediating acute lung inflammation in a cell-specific manner. ADAM17 promotes endothelial and epithelial permeability, transendothelial leukocyte migration, and inflammatory mediator production by smooth muscle and epithelial cells. ADAM10 is critical for leukocyte migration and alveolar leukocyte recruitment. ADAM10 also promotes allergic asthma by driving B cell responses. Additionally, ADAM10 acts as a receptor for Staphylococcus aureus (S. aureus) α-toxin and is crucial for bacterial virulence. ADAM8, ADAM9, ADAM15, and ADAM33 are upregulated during acute or chronic lung inflammation, and recent functional or genetic analyses have linked them to disease development. Pharmacological inhibitors that allow us to locally or systemically target and differentiate ADAM-family members in the lung suppress acute and asthmatic inflammatory responses and S. aureus virulence. These promising results encourage further research to develop therapeutic strategies based on selected ADAMs. These studies need also to address the role of the ADAMs in repair and regeneration in the lung to identify further therapeutic opportunities and possible side effects.
Collapse
Affiliation(s)
- Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| |
Collapse
|