1
|
Zhang N, Liao H, Lin Z, Tang Q. Insights into the Role of Glutathione Peroxidase 3 in Non-Neoplastic Diseases. Biomolecules 2024; 14:689. [PMID: 38927092 PMCID: PMC11202029 DOI: 10.3390/biom14060689] [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: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROSs) are byproducts of normal cellular metabolism and play pivotal roles in various physiological processes. Disruptions in the balance between ROS levels and the body's antioxidant defenses can lead to the development of numerous diseases. Glutathione peroxidase 3 (GPX3), a key component of the body's antioxidant system, is an oxidoreductase enzyme. GPX3 mitigates oxidative damage by catalyzing the conversion of hydrogen peroxide into water. Beyond its antioxidant function, GPX3 is vital in regulating metabolism, modulating cell growth, inducing apoptosis and facilitating signal transduction. It also serves as a significant tumor suppressor in various cancers. Recent studies have revealed aberrant expression of GPX3 in several non-neoplastic diseases, associating it with multiple pathological processes. This review synthesizes the current understanding of GPX3 expression and regulation, highlighting its extensive roles in noncancerous diseases. Additionally, this paper evaluates the potential of GPX3 as a diagnostic biomarker and explores emerging therapeutic strategies targeting this enzyme, offering potential avenues for future clinical treatment of non-neoplastic conditions.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Haihan Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Zheng Lin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (N.Z.); (H.L.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
2
|
Zhang F, Lu JW, Lei WJ, Li MD, Pan F, Lin YK, Wang WS, Sun K. Paradoxical Induction of ALOX15/15B by Cortisol in Human Amnion Fibroblasts: Implications for Inflammatory Responses of the Fetal Membranes at Parturition. Int J Mol Sci 2023; 24:10881. [PMID: 37446059 DOI: 10.3390/ijms241310881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/23/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Inflammation of the fetal membranes is an indispensable event of parturition, with increasing prostaglandin E2 (PGE2) synthesis as one of the ultimate products that prime labor onset. In addition to PGE2, the fetal membranes also boast a large capacity for cortisol regeneration. It is intriguing how increased PGE2 synthesis is achieved in the presence of increasing amounts of classical anti-inflammatory glucocorticoids in the fetal membranes at parturition. 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) synthesized by lipoxygenase 15/15B (ALOX15/15B) has been shown to enhance inflammation-induced PGE2 synthesis in amnion fibroblasts. Here, we examined whether glucocorticoids could induce ALOX15/15B expression and 15(S)-HETE production to promote PGE2 synthesis in amnion fibroblasts at parturition. We found that cortisol and 15(S)-HETE abundance increased parallelly in the amnion at parturition. Cortisol induced ALOX15/15B expression and 15(S)-HETE production paradoxically in amnion fibroblasts. Mechanism study revealed that this paradoxical induction was mediated by p300-mediated histone acetylation and interaction of glucocorticoid receptor with transcription factors CREB and STAT3. Conclusively, cortisol regenerated in the fetal membranes can paradoxically induce ALOX15/15B expression and 15(S)-HETE production in human amnion fibroblasts, which may further assist in the induction of PGE2 synthesis in the inflammatory responses of the fetal membranes for parturition.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Jiang-Wen Lu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Wen-Jia Lei
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Meng-Die Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Fan Pan
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Yi-Kai Lin
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Wang-Sheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| |
Collapse
|
3
|
Queen K, Nguyen MN, Gilliland FD, Chun S, Raby BA, Millstein J. ACDC: a general approach for detecting phenotype or exposure associated co-expression. Front Med (Lausanne) 2023; 10:1118824. [PMID: 37275375 PMCID: PMC10235619 DOI: 10.3389/fmed.2023.1118824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Background Existing module-based differential co-expression methods identify differences in gene-gene relationships across phenotype or exposure structures by testing for consistent changes in transcription abundance. Current methods only allow for assessment of co-expression variation across a singular, binary or categorical exposure or phenotype, limiting the information that can be obtained from these analyses. Methods Here, we propose a novel approach for detection of differential co-expression that simultaneously accommodates multiple phenotypes or exposures with binary, ordinal, or continuous data types. Results We report an application to two cohorts of asthmatic patients with varying levels of asthma control to identify associations between gene co-expression and asthma control test scores. Results suggest that both expression levels and covariances of ADORA3, ALOX15, and IDO1 are associated with asthma control. Conclusion ACDC is a flexible extension to existing methodology that can detect differential co-expression across varying external variables.
Collapse
Affiliation(s)
- Katelyn Queen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - My-Nhi Nguyen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Frank D. Gilliland
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sung Chun
- Division of Pulmonary Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Benjamin A. Raby
- Division of Pulmonary Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
4
|
Venuto S, Coda ARD, González-Pérez R, Laselva O, Tolomeo D, Storlazzi CT, Liso A, Conese M. IGFBP-6 Network in Chronic Inflammatory Airway Diseases and Lung Tumor Progression. Int J Mol Sci 2023; 24:4804. [PMID: 36902237 PMCID: PMC10003725 DOI: 10.3390/ijms24054804] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
The lung is an accomplished organ for gas exchanges and directly faces the external environment, consequently exposing its large epithelial surface. It is also the putative determinant organ for inducing potent immune responses, holding both innate and adaptive immune cells. The maintenance of lung homeostasis requires a crucial balance between inflammation and anti-inflammation factors, and perturbations of this stability are frequently associated with progressive and fatal respiratory diseases. Several data demonstrate the involvement of the insulin-like growth factor (IGF) system and their binding proteins (IGFBPs) in pulmonary growth, as they are specifically expressed in different lung compartments. As we will discuss extensively in the text, IGFs and IGFBPs are implicated in normal pulmonary development but also in the pathogenesis of various airway diseases and lung tumors. Among the known IGFBPs, IGFBP-6 shows an emerging role as a mediator of airway inflammation and tumor-suppressing activity in different lung tumors. In this review, we assess the current state of IGFBP-6's multiple roles in respiratory diseases, focusing on its function in the inflammation and fibrosis in respiratory tissues, together with its role in controlling different types of lung cancer.
Collapse
Affiliation(s)
- Santina Venuto
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | | | - Ruperto González-Pérez
- Allergy Department, Hospital Universitario de Canarias, 38320 Tenerife, Spain
- Severe Asthma Unit, Hospital Universitario de Canarias, 38320 Tenerife, Spain
| | - Onofrio Laselva
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Doron Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
5
|
Laha A, Moitra S, Podder S. A review on aero-allergen induced allergy in India. Clin Exp Allergy 2023. [PMID: 36756731 DOI: 10.1111/cea.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 02/10/2023]
Abstract
Allergic diseases such as bronchial asthma, rhinitis and eczema are an important public health issue globally in 21st century. Their prevalence and impact in India is high, in terms of total numbers of people affected. Due to diverse climate and lifestyle, the Indian population is exposed to a rich and varied range of aerobiological particles. In the last two decades in India, 83 pollens, 34 fungi, six dust mite and 19 insect species were reported as allergenic, often contributing to heavy allergenic exposure in indoor and outdoor environments. There are currently few practitioners who offer allergy diagnosis and treatment in India, but there is active basic and molecular research aiming to improve the health of allergic patients. This review explores the field of aero-allergy in India during the last two decades, with special emphasis on prevalence across various centres and relevant scientific advances such as genetic associations and new therapeutics. This review will be helpful for clinicians to select aero-allergens for sensitisation testing in patients suffering from symptoms in different regions of India, and to support patients undertaking prophylaxis of allergic symptoms in India according to local environmental conditions. This study also helps to define patient selection for allergy testing and the role of test results in guiding allergic disease management in the Indian context. Finally, we summarize genetic findings relevant to aero-allergy in India and discuss the development of novel therapeutics for investigation and management of aero-allergy in India.
Collapse
Affiliation(s)
- Arghya Laha
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Bardhaman, West Bengal, India
| | - Saibal Moitra
- Apollo Multispecialty Hospitals, Kolkata, West Bengal, India
| | - Sanjoy Podder
- Ecology and Allergology Lab, Department of Zoology, The University of Burdwan, Bardhaman, West Bengal, India
| |
Collapse
|
6
|
Liso A, Venuto S, Coda ARD, Giallongo C, Palumbo GA, Tibullo D. IGFBP-6: At the Crossroads of Immunity, Tissue Repair and Fibrosis. Int J Mol Sci 2022; 23:ijms23084358. [PMID: 35457175 PMCID: PMC9030159 DOI: 10.3390/ijms23084358] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/04/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factors binding protein-6 (IGFBP-6) is involved in a relevant number of cellular activities and represents an important factor in the immune response, particularly in human dendritic cells (DCs). Over the past several years, significant insights into the IGF-independent effects of IGFBP-6 were discovered, such as the induction of chemotaxis, capacity to increase oxidative burst and neutrophils degranulation, ability to induce metabolic changes in DCs, and, more recently, the regulation of the Sonic Hedgehog (SHH) signaling pathway during fibrosis. IGFBP-6 has been implicated in different human diseases, and it plays a rather controversial role in the biology of tumors. Notably, well established relationships between immunity, stroma activity, and fibrosis are prognostic and predictive of response to cancer immunotherapy. This review aims at describing the current understanding of mechanisms that link IGFBP-6 and fibrosis development and at highlighting the multiple roles of IGFBP-6 to provide an insight into evolutionarily conserved mechanisms that can be relevant for inflammation, tumor immunity, and immunological diseases.
Collapse
Affiliation(s)
- Arcangelo Liso
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (S.V.); (A.R.D.C.)
- Correspondence:
| | - Santina Venuto
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (S.V.); (A.R.D.C.)
| | - Anna Rita Daniela Coda
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (S.V.); (A.R.D.C.)
| | - Cesarina Giallongo
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (C.G.); (G.A.P.)
| | - Giuseppe Alberto Palumbo
- Department of Medical Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy; (C.G.); (G.A.P.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
7
|
Watts AM, West NP, Smith PK, Zhang P, Cripps AW, Cox AJ. Nasal immune gene expression in response to azelastine and fluticasone propionate combination or monotherapy. Immun Inflamm Dis 2022; 10:e571. [PMID: 34813682 PMCID: PMC8926499 DOI: 10.1002/iid3.571] [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: 09/14/2021] [Revised: 10/28/2021] [Accepted: 11/13/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The combination of the antihistamine azelastine (AZE) with the corticosteroid fluticasone propionate (FP) in a single spray, has been reported to be significantly more effective at reducing allergic rhinitis (AR) symptoms than treatment with either corticosteroid or antihistamine monotherapy. However, the biological basis for enhanced symptom relief is not known. This study aimed to compare gene expression profiles (760 immune genes, performed with the NanoString nCounter) from peripheral blood and nasal brushing/lavage lysate samples in response to nasal spray treatment. METHODS Moderate/severe persistent dust mite AR sufferers received either AZE (125 μg/spray) nasal spray (n = 16), FP (50 μg/spray) nasal spray (n = 14) or combination spray AZE/FP (125 μg AZE and 50 μg FP/spray) (n = 14) for 7 days, twice daily. Self-reported symptom questionnaires were completed daily for the study duration. Gene expression analysis (760 immune genes) was performed with the NanoString nCounter on purified RNA from peripheral blood and nasal brushing/lavage lysate samples. RESULTS In nasal samples, 206 genes were significantly differentially expressed following FP treatment; 182 genes downregulated (-2.57 to -0.45 Log2 fold change [FC]), 24 genes upregulated (0.49-1.40 Log2 FC). In response to AZE/FP, only 16 genes were significantly differentially expressed; 10 genes downregulated (-1.53 to -0.58 Log2 FC), six genes upregulated (1.07-1.62 Log2 FC). Following AZE treatment only five genes were significantly differentially expressed; one gene downregulated (-1.68 Log2 FC), four genes upregulated (0.59-1.19 Log2 FC). Immune gene changes in peripheral blood samples following treatment were minimal. AR symptoms improved under all treatments, but improvements were less pronounced following AZE treatment. CONCLUSION AZE/FP, FP, and AZE had diverse effects on immune gene expression profiles in nasal mucosa samples. The moderate number of genes modulated by AZE/FP indicates alternative pathways in reducing AR symptoms whilst avoiding extensive local immune suppression.
Collapse
Affiliation(s)
- Annabelle M. Watts
- School of Medical ScienceGriffith UniversitySouthportQueenslandAustralia
| | - Nicholas P. West
- School of Medical ScienceGriffith UniversitySouthportQueenslandAustralia
- Menzies Health Institute of QueenslandGriffith UniversitySouthportQueenslandAustralia
| | - Peter K. Smith
- Queensland Allergy Services ClinicSouthportQueenslandAustralia
| | - Ping Zhang
- Menzies Health Institute of QueenslandGriffith UniversitySouthportQueenslandAustralia
| | - Allan W. Cripps
- Menzies Health Institute of QueenslandGriffith UniversitySouthportQueenslandAustralia
- School of MedicineGriffith UniversitySouthportQueenslandAustralia
| | - Amanda J. Cox
- School of Medical ScienceGriffith UniversitySouthportQueenslandAustralia
- Menzies Health Institute of QueenslandGriffith UniversitySouthportQueenslandAustralia
| |
Collapse
|
8
|
van der Ploeg EA, Melgert BN, Burgess JK, Gan CT. The potential of biomarkers of fibrosis in chronic lung allograft dysfunction. Transplant Rev (Orlando) 2021; 35:100626. [PMID: 33992914 DOI: 10.1016/j.trre.2021.100626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 11/27/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major long-term cause of morbidity and mortality after lung transplantation. Both bronchiolitis obliterans syndrome and restrictive lung allograft syndrome, two main types of CLAD, lead to fibrosis in either the small airways or alveoli and pleura. Pathological pathways in CLAD and other types of fibrosis, for example idiopathic pulmonary fibrosis, are assumed to overlap and therefore fibrosis biomarkers could aid in the early detection of CLAD. These biomarkers could help to differentiate between different phenotypes of CLAD and could, in comparison to biomarkers of inflammation, possibly distinguish an infectious event from CLAD when a decline in lung function is present. This review gives an overview of known CLAD specific biomarkers, describes new promising fibrosis biomarkers currently investigated in other types of fibrosis, and discusses the possible use of these fibrosis biomarkers for CLAD.
Collapse
Affiliation(s)
- Eline A van der Ploeg
- University of Groningen, University Medical Centre Groningen, Department of Pulmonary Medicine, PO Box 30. 001, 9700, RB, Groningen, the Netherlands.
| | - Barbro N Melgert
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, PO box 196, 9700, AD, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, PO Box 30.001, 9700, RB, Groningen, the Netherlands.
| | - Janette K Burgess
- University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, PO Box 30.001, 9700, RB, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, PO Box 30.001, 9700, RB, Groningen, the Netherlands.
| | - C Tji Gan
- University of Groningen, University Medical Centre Groningen, Department of Pulmonary Medicine, PO Box 30. 001, 9700, RB, Groningen, the Netherlands.
| |
Collapse
|
9
|
Knuplez E, Sturm EM, Marsche G. Emerging Role of Phospholipase-Derived Cleavage Products in Regulating Eosinophil Activity: Focus on Lysophospholipids, Polyunsaturated Fatty Acids and Eicosanoids. Int J Mol Sci 2021; 22:4356. [PMID: 33919453 PMCID: PMC8122506 DOI: 10.3390/ijms22094356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Eosinophils are important effector cells involved in allergic inflammation. When stimulated, eosinophils release a variety of mediators initiating, propagating, and maintaining local inflammation. Both, the activity and concentration of secreted and cytosolic phospholipases (PLAs) are increased in allergic inflammation, promoting the cleavage of phospholipids and thus the production of reactive lipid mediators. Eosinophils express high levels of secreted phospholipase A2 compared to other leukocytes, indicating their direct involvement in the production of lipid mediators during allergic inflammation. On the other side, eosinophils have also been recognized as crucial mediators with regulatory and homeostatic roles in local immunity and repair. Thus, targeting the complex network of lipid mediators offer a unique opportunity to target the over-activation and 'pro-inflammatory' phenotype of eosinophils without compromising the survival and functions of tissue-resident and homeostatic eosinophils. Here we provide a comprehensive overview of the critical role of phospholipase-derived lipid mediators in modulating eosinophil activity in health and disease. We focus on lysophospholipids, polyunsaturated fatty acids, and eicosanoids with exciting new perspectives for future drug development.
Collapse
Affiliation(s)
| | | | - Gunther Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria; (E.K.); (E.M.S.)
| |
Collapse
|
10
|
The Utility of Resolving Asthma Molecular Signatures Using Tissue-Specific Transcriptome Data. G3-GENES GENOMES GENETICS 2020; 10:4049-4062. [PMID: 32900903 PMCID: PMC7642926 DOI: 10.1534/g3.120.401718] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An integrative analysis focused on multi-tissue transcriptomics has not been done for asthma. Tissue-specific DEGs remain undetected in many multi-tissue analyses, which influences identification of disease-relevant pathways and potential drug candidates. Transcriptome data from 609 cases and 196 controls, generated using airway epithelium, bronchial, nasal, airway macrophages, distal lung fibroblasts, proximal lung fibroblasts, CD4+ lymphocytes, CD8+ lymphocytes from whole blood and induced sputum samples, were retrieved from Gene Expression Omnibus (GEO). Differentially regulated asthma-relevant genes identified from each sample type were used to identify (a) tissue-specific and tissue-shared asthma pathways, (b) their connection to GWAS-identified disease genes to identify candidate tissue for functional studies, (c) to select surrogate sample for invasive tissues, and finally (d) to identify potential drug candidates via connectivity map analysis. We found that inter-tissue similarity in gene expression was more pronounced at pathway/functional level than at gene level with highest similarity between bronchial epithelial cells and lung fibroblasts, and lowest between airway epithelium and whole blood samples. Although public-domain gene expression data are limited by inadequately annotated per-sample demographic and clinical information which limited the analysis, our tissue-resolved analysis clearly demonstrated relative importance of unique and shared asthma pathways, At the pathway level, IL-1b signaling and ERK signaling were significant in many tissue types, while Insulin-like growth factor and TGF-beta signaling were relevant in only airway epithelial tissue. IL-12 (in macrophages) and Immunoglobulin signaling (in lymphocytes) and chemokines (in nasal epithelium) were the highest expressed pathways. Overall, the IL-1 signaling genes (inflammatory) were relevant in the airway compartment, while pro-Th2 genes including IL-13 and STAT6 were more relevant in fibroblasts, lymphocytes, macrophages and bronchial biopsies. These genes were also associated with asthma in the GWAS catalog. Support Vector Machine showed that DEGs based on macrophages and epithelial cells have the highest and lowest discriminatory accuracy, respectively. Drug (entinostat, BMS-345541) and genetic perturbagens (KLF6, BCL10, INFB1 and BAMBI) negatively connected to disease at multi-tissue level could potentially repurposed for treating asthma. Collectively, our study indicates that the DEGs, perturbagens and disease are connected differentially depending on tissue/cell types. While most of the existing literature describes asthma transcriptome data from individual sample types, the present work demonstrates the utility of multi-tissue transcriptome data. Future studies should focus on collecting transcriptomic data from multiple tissues, age and race groups, genetic background, disease subtypes and on the availability of better-annotated data in the public domain.
Collapse
|
11
|
Pecak M, Korošec P, Kunej T. Multiomics Data Triangulation for Asthma Candidate Biomarkers and Precision Medicine. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:392-409. [PMID: 29927718 DOI: 10.1089/omi.2018.0036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Asthma is a common complex disorder and has been subject to intensive omics research for disease susceptibility and therapeutic innovation. Candidate biomarkers of asthma and its precision treatment demand that they stand the test of multiomics data triangulation before they can be prioritized for clinical applications. We classified the biomarkers of asthma after a search of the literature and based on whether or not a given biomarker candidate is reported in multiple omics platforms and methodologies, using PubMed and Web of Science, we identified omics studies of asthma conducted on diverse platforms using keywords, such as asthma, genomics, metabolomics, and epigenomics. We extracted data about asthma candidate biomarkers from 73 articles and developed a catalog of 190 potential asthma biomarkers (167 human, 23 animal data), comprising DNA loci, transcripts, proteins, metabolites, epimutations, and noncoding RNAs. The data were sorted according to 13 omics types: genomics, epigenomics, transcriptomics, proteomics, interactomics, metabolomics, ncRNAomics, glycomics, lipidomics, environmental omics, pharmacogenomics, phenomics, and integrative omics. Importantly, we found that 10 candidate biomarkers were apparent in at least two or more omics levels, thus promising potential for further biomarker research and development and precision medicine applications. This multiomics catalog reported herein for the first time contributes to future decision-making on prioritization of biomarkers and validation efforts for precision medicine in asthma. The findings may also facilitate meta-analyses and integrative omics studies in the future.
Collapse
Affiliation(s)
- Matija Pecak
- 1 Department of Animal Science, Biotechnical Faculty, University of Ljubljana , Domzale, Slovenia
| | - Peter Korošec
- 2 Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases , Golnik, Slovenia
| | - Tanja Kunej
- 1 Department of Animal Science, Biotechnical Faculty, University of Ljubljana , Domzale, Slovenia
| |
Collapse
|
12
|
Qi S, Liu G, Dong X, Huang N, Li W, Chen H. Microarray data analysis to identify differentially expressed genes and biological pathways associated with asthma. Exp Ther Med 2018; 16:1613-1620. [PMID: 30186379 PMCID: PMC6122392 DOI: 10.3892/etm.2018.6366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 02/17/2018] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to identify differentially expressed genes (DEGs) and biological processes (BPs) associated with asthma. DEGs between allergic asthma and healthy controls were screened from GSE15823. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed using Database for Annotation, Visualization and Integrated Discovery. A protein-protein interaction network was constructed, followed by module mining and functional analysis. Additionally, GSE41649 was downloaded to validate the reliability of the results. In GSE41649, DEGs were identified and compared with key DEGs identified in GSE15823. A total of 43 upregulated and 275 downregulated DEGs were obtained from GSE15823. Upregulated DEGs, such as nitric oxide synthase 2 (NOS2), were enriched in BPs related to oxidation reduction. Downregulated DEGs, such as chemokine (C-C motif) ligand 19 (CCL21) and Cys-X-Cys ligand (CXCL9), were enriched in immune response-related BPs. Protein tyrosine phosphatase receptor type C (PTPRC), CCL21, and CXCL9 were identified as hub genes. The DEGs in module 1 were significantly involved in the chemokine signaling pathway (P<0.05). The expression of the key genes obtained in GSE15823 demonstrated the same variation directions in the two datasets. The immune response, oxidants and nitric oxide metabolic pathways may have important roles in the progression of asthma. DEGs of PTPRC, CCL21, CXCL9 and NOS2 may be the potential targets for asthma diagnosis and treatment.
Collapse
Affiliation(s)
- Shanshan Qi
- Department of Allergy, Wuhan No. 1 Hospital, Wuhan, Hubei 430022, P.R. China
| | - Guanghui Liu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiang Dong
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Nan Huang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Wenjing Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hao Chen
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
13
|
Margaritte-Jeannin P, Babron MC, Laprise C, Lavielle N, Sarnowski C, Brossard M, Moffatt M, Gagné-Ouellet V, Etcheto A, Lathrop M, Just J, Cookson WO, Bouzigon E, Demenais F, Dizier MH. The COL5A3 and MMP9 genes interact in eczema susceptibility. Clin Exp Allergy 2017; 48:297-305. [PMID: 29168291 DOI: 10.1111/cea.13064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/10/2017] [Accepted: 11/07/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Genetic studies of eczema have identified many genes, which explain only 14% of the heritability. Missing heritability may be partly due to ignored gene-gene (G-G) interactions. OBJECTIVE Our aim was to detect new interacting genes involved in eczema. METHODS The search for G-G interaction in eczema was conducted using a two-step approach, which included as a first step, a biological selection of genes, which are involved either in the skin or epidermis development or in the collagen metabolism, and as a second step, an interaction analysis of the selected genes. Analyses were carried out at both SNP and gene levels in three asthma-ascertained family samples: the discovery dataset of 388 EGEA (Epidemiological study on the Genetics and Environment of Asthma) families and the two replication datasets of 253 SLSJ (Saguenay-Lac-Saint-Jean) families and 207 MRCA (Medical Research Council) families. RESULTS One pair of SNPs, rs2287807 in COL5A3 and rs17576 in MMP9, that were detected in EGEA at P ≤ 10-5 showed significant interaction by meta-analysis of EGEA, SLSJ and MRCA samples (P = 1.1 × 10-8 under the significant threshold of 10-7 ). Gene-based analysis confirmed strong interaction between COL5A3 and MMP9 (P = 4 × 10-8 under the significant threshold of 4 × 10-6 ) by meta-analysis of the three datasets. When stratifying the data on asthma, this interaction remained in both groups of asthmatic and non-asthmatic subjects. CONCLUSION This study identified significant interaction between two new genes, COL5A3 and MMP9, which may be accounted for by a degradation of COL5A3 by MMP9 influencing eczema susceptibility. Further confirmation of this interaction as well as functional studies is needed to better understand the role of these genes in eczema.
Collapse
Affiliation(s)
- P Margaritte-Jeannin
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - M-C Babron
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - C Laprise
- Université du Québec, Chicoutimi, Canada
| | - N Lavielle
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - C Sarnowski
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - M Brossard
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - M Moffatt
- National Heart Lung Institute, Imperial College, London, UK
| | | | - A Etcheto
- Rheumatology Department, Cochin Hospital, AP-HP, INSERM U1153, Sorbonne Paris-Cité, Paris Descartes University, Paris, France
| | - M Lathrop
- Mc Gill University and Genome Quebec's Innovation Centre, Montréal, Canada
| | - J Just
- Service d'Allergologie Pédiatrique, Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand-Trousseau-UPMC Paris 06, Paris, France
| | - W O Cookson
- National Heart Lung Institute, Imperial College, London, UK
| | - E Bouzigon
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - F Demenais
- Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| | - M-H Dizier
- Inserm, UMR-946, Genetic Variation and Human Diseases unit, Université Paris-Diderot, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Univ Paris Diderot, Paris, France
| |
Collapse
|
14
|
Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev 2017; 278:63-86. [PMID: 28658551 PMCID: PMC5501488 DOI: 10.1111/imr.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
15
|
Giziry DE, Zakaria NH, Kassem AH, Abdellatif MM. The study of fibulin-1 as a novel biomarker in bronchial asthma and its association with disease severity. EGYPTIAN JOURNAL OF CHEST DISEASES AND TUBERCULOSIS 2017. [DOI: 10.1016/j.ejcdt.2016.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
16
|
Zuiker RGJA, Tribouley C, Diamant Z, Boot JD, Cohen AF, Van Dyck K, De Lepeleire I, Rivas VM, Malkov VA, Burggraaf J, Ruddy MK. Sputum RNA signature in allergic asthmatics following allergen bronchoprovocation test. Eur Clin Respir J 2016; 3:31324. [PMID: 27421833 PMCID: PMC4947196 DOI: 10.3402/ecrj.v3.31324] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/30/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022] Open
Abstract
Background Inhaled allergen challenge is a validated disease model of allergic asthma offering useful pharmacodynamic assessment of pharmacotherapeutic effects in a limited number of subjects. Objectives To evaluate whether an RNA signature can be identified from induced sputum following an inhaled allergen challenge, whether a RNA signature could be modulated by limited doses of inhaled fluticasone, and whether these gene expression profiles would correlate with the clinical endpoints measured in this study. Methods Thirteen non-smoking, allergic subjects with mild-to-moderate asthma participated in a randomised, placebo-controlled, 2-period cross-over study following a single-blind placebo run-in period. Each period consisted of three consecutive days, separated by a wash-out period of at least 3 weeks. Subjects randomly received inhaled fluticasone ((FP) MDI; 500 mcg BID×5 doses in total) or placebo. On day 2, house dust mite extract was inhaled and airway response was measured by FEV1 at predefined time points until 7 h post-allergen. Sputum was induced by NaCl 4.5%, processed and analysed at 24 h pre-allergen and 7 and 24 h post-allergen. RNA was isolated from eligible sputum cell pellets (<80% squamous of 500 cells), amplified according to NuGEN technology, and profiled on Affymetrix arrays. Gene expression changes from baseline and fluticasone treatment effects were evaluated using a mixed effects ANCOVA model at 7 and at 24 h post-allergen challenge. Results Inhaled allergen-induced statistically significant gene expression changes in sputum, which were effectively blunted by fluticasone (adjusted p<0.025). Forty-seven RNA signatures were selected from these responses for correlation analyses and further validation. This included Th2 mRNA levels for cytokines, chemokines, high-affinity IgE receptor FCER1A, histamine receptor HRH4, and enzymes and receptors in the arachidonic pathway. Individual messengers from the 47 RNA signatures correlated significantly with lung function and sputum eosinophil counts. Conclusion Our RNA extraction and profiling protocols allowed reproducible assessments of inflammatory signatures in sputum including quantification of drug effects on this response in allergic asthmatics. This approach offers novel possibilities for the development of pharmacodynamic (PD) biomarkers in asthma.
Collapse
Affiliation(s)
| | - Catherine Tribouley
- Merck Research Laboratories, Rahway, New Jersey, USA.,Novartis, New York, NY, USA
| | - Zuzana Diamant
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Respiratory Medicine and Allergology, Skane University Hospital, Lund, Sweden.,Department of Clinical & Pharmacology, University Medical Center Groningen, Groningen, The Netherlands.,Department of General Practice, University Medical Center Groningen, Groningen, The Netherlands.,QPS Netherlands, Groningen, The Netherlands
| | - J Diderik Boot
- Centre for Human Drug Research, Leiden, The Netherlands.,Janssen Biologics B.V., Leiden, The Netherlands
| | - Adam F Cohen
- Centre for Human Drug Research, Leiden, The Netherlands
| | - K Van Dyck
- Merck Research Laboratories, Rahway, New Jersey, USA
| | | | | | | | | | - Marcella K Ruddy
- Merck Research Laboratories, Rahway, New Jersey, USA.,EMD Serono, Rockland, MA, USA
| |
Collapse
|
17
|
Piyadasa H, Altieri A, Basu S, Schwartz J, Halayko AJ, Mookherjee N. Biosignature for airway inflammation in a house dust mite-challenged murine model of allergic asthma. Biol Open 2016; 5:112-21. [PMID: 26740570 PMCID: PMC4823983 DOI: 10.1242/bio.014464] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
House dust mite (HDM) challenge is commonly used in murine models of allergic asthma for preclinical pathophysiological studies. However, few studies define objective readouts or biomarkers in this model. In this study we characterized immune responses and defined molecular markers that are specifically altered after HDM challenge. In this murine model, we used repeated HDM challenge for two weeks which induced hallmarks of allergic asthma seen in humans, including airway hyper-responsiveness (AHR) and elevated levels of circulating total and HDM-specific IgE and IgG1. Kinetic studies showed that at least 24 h after last HDM challenge results in significant AHR along with eosinophil infiltration in the lungs. Histologic assessment of lung revealed increased epithelial thickness and goblet cell hyperplasia, in the absence of airway wall collagen deposition, suggesting ongoing tissue repair concomitant with acute allergic lung inflammation. Thus, this model may be suitable to delineate airway inflammation processes that precede airway remodeling and development of fixed airway obstruction. We observed that a panel of cytokines e.g. IFN-γ, IL-1β, IL-4, IL-5, IL-6, KC, TNF-α, IL-13, IL-33, MDC and TARC were elevated in lung tissue and bronchoalveolar fluid, indicating local lung inflammation. However, levels of these cytokines remained unchanged in serum, reflecting lack of systemic inflammation in this model. Based on these findings, we further monitored the expression of 84 selected genes in lung tissues by quantitative real-time PCR array, and identified 31 mRNAs that were significantly up-regulated in lung tissue from HDM-challenged mice. These included genes associated with human asthma (e.g. clca3, ear11, il-13, il-13ra2, il-10, il-21, arg1 and chia1) and leukocyte recruitment in the lungs (e.g. ccl11, ccl12 and ccl24). This study describes a biosignature to enable broad and systematic interrogation of molecular mechanisms and intervention strategies for airway inflammation pertinent to allergic asthma that precedes and possibly potentiates airway remodeling and fibrosis. Summary: This study describes a systematic analysis of molecular end points in an murine model of allergic asthma. The biosignature described can be used to interrogate molecular mechanisms and intervention strategies for airway inflammation pertinent to allergic asthma that precedes and possibly potentiates airway remodeling and fibrosis.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada Department of Immunology, University of Manitoba, Winnipeg, Manitoba, R3E 0T5, Canada
| | - Anthony Altieri
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada Department of Immunology, University of Manitoba, Winnipeg, Manitoba, R3E 0T5, Canada
| | - Sujata Basu
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada
| | - Jacquie Schwartz
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada
| | - Andrew J Halayko
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, R3E 0T5, Canada Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada Canadian Respiratory Research Network
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada Department of Immunology, University of Manitoba, Winnipeg, Manitoba, R3E 0T5, Canada Biology of Breathing Group, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, R3E 3P4, Canada Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada Canadian Respiratory Research Network
| |
Collapse
|
18
|
Pediatric asthma and autism-genomic perspectives. Clin Transl Med 2015; 4:37. [PMID: 26668064 PMCID: PMC4678135 DOI: 10.1186/s40169-015-0078-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/29/2015] [Indexed: 02/06/2023] Open
Abstract
High-throughput technologies, ranging from microarrays to NexGen sequencing of RNA and genomic DNA, have opened new avenues for exploration of the pathobiology of human disease. Comparisons of the architecture of the genome, identification of mutated or modified sequences, and pre-and post- transcriptional regulation of gene expression as disease specific biomarkers are revolutionizing our understanding of the causes of disease and are guiding the development of new therapies. There is enormous heterogeneity in types of genomic variation that occur in human disease. Some are inherited, while others are the result of new somatic or germline mutations or errors in chromosomal replication. In this review, we provide examples of changes that occur in the human genome in two of the most common chronic pediatric disorders, autism and asthma. The incidence and economic burden of both of these disorders are increasing worldwide. Genomic variations have the potential to serve as biomarkers for personalization of therapy and prediction of outcomes.
Collapse
|
19
|
Gagné-Ouellet V, Guay SP, Boucher-Lafleur AM, Bouchard L, Laprise C. DNA methylation signature of interleukin 1 receptor type II in asthma. Clin Epigenetics 2015; 7:80. [PMID: 26246860 PMCID: PMC4526162 DOI: 10.1186/s13148-015-0114-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/13/2015] [Indexed: 12/25/2022] Open
Abstract
Interleukin 1 and its receptors are associated with allergic diseases such as asthma. In the present study, we measured DNA methylation at the IL1R1 and IL1R2 gene loci and assessed for associations with asthma-related phenotypes and gene expressions. We found that asthmatic and atopic individuals have higher IL1R2 promoter DNA methylation than control subjects. Additionally, we observed a negative correlation between DNA methylation at the IL1R2 promoter and IL1R2 mRNA expression. These results suggest for the first time that IL1R2 promoter DNA methylation is associated with its gene repression in allergic diseases such as asthma.
Collapse
Affiliation(s)
- Valérie Gagné-Ouellet
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Chicoutimi, QC Canada
| | - Simon-Pierre Guay
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC Canada ; ECOGENE-21 and Lipid Clinic, Hôpital de Chicoutimi, Saguenay, QC Canada
| | | | - Luigi Bouchard
- Department of Biochemistry, Université de Sherbrooke, Sherbrooke, QC Canada ; ECOGENE-21 and Lipid Clinic, Hôpital de Chicoutimi, Saguenay, QC Canada
| | - Catherine Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Chicoutimi, QC Canada
| |
Collapse
|
20
|
Persson H, Kwon AT, Ramilowski JA, Silberberg G, Söderhäll C, Orsmark-Pietras C, Nordlund B, Konradsen JR, de Hoon MJL, Melén E, Hayashizaki Y, Hedlin G, Kere J, Daub CO. Transcriptome analysis of controlled and therapy-resistant childhood asthma reveals distinct gene expression profiles. J Allergy Clin Immunol 2015; 136:638-48. [PMID: 25863981 DOI: 10.1016/j.jaci.2015.02.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 11/23/2014] [Accepted: 02/03/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Children with problematic severe asthma have poor disease control despite high doses of inhaled corticosteroids and additional therapy, leading to personal suffering, early deterioration of lung function, and significant consumption of health care resources. If no exacerbating factors, such as smoking or allergies, are found after extensive investigation, these children are given a diagnosis of therapy-resistant (or therapy-refractory) asthma (SA). OBJECTIVE We sought to deepen our understanding of childhood SA by analyzing gene expression and modeling the underlying regulatory transcription factor networks in peripheral blood leukocytes. METHODS Gene expression was analyzed by using Cap Analysis of Gene Expression in children with SA (n = 13), children with controlled persistent asthma (n = 15), and age-matched healthy control subjects (n = 9). Cap Analysis of Gene Expression sequencing detects the transcription start sites of known and novel mRNAs and noncoding RNAs. RESULTS Sample groups could be separated by hierarchical clustering on 1305 differentially expressed transcription start sites, including 816 known genes and several novel transcripts. Ten of 13 tested novel transcripts were validated by means of RT-PCR and Sanger sequencing. Expression of RAR-related orphan receptor A (RORA), which has been linked to asthma in genome-wide association studies, was significantly upregulated in patients with SA. Gene network modeling revealed decreased glucocorticoid receptor signaling and increased activity of the mitogen-activated protein kinase and Jun kinase cascades in patients with SA. CONCLUSION Circulating leukocytes from children with controlled asthma and those with SA have distinct gene expression profiles, demonstrating the possible development of specific molecular biomarkers and supporting the need for novel therapeutic approaches.
Collapse
Affiliation(s)
- Helena Persson
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Stockholm, Sweden
| | - Andrew T Kwon
- Omics Science Center,§ RIKEN Yokohama Institute, Yokohama, Japan; Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Jordan A Ramilowski
- Omics Science Center,§ RIKEN Yokohama Institute, Yokohama, Japan; Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Gilad Silberberg
- Unit of Computational Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Christina Orsmark-Pietras
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Stockholm, Sweden
| | - Björn Nordlund
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Jon R Konradsen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Michiel J L de Hoon
- Omics Science Center,§ RIKEN Yokohama Institute, Yokohama, Japan; Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Erik Melén
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Sachs' Children's Hospital, Stockholm, Sweden
| | - Yoshihide Hayashizaki
- Omics Science Center,§ RIKEN Yokohama Institute, Yokohama, Japan; Preventive Medicine and Diagnosis Innovation Program, RIKEN Research Cluster for Innovation, Wako, Japan
| | - Gunilla Hedlin
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Stockholm, Sweden; Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden; Folkhälsan Institute of Genetics, Helsinki, Finland; Research Programs Unit, University of Helsinki, Helsinki, Finland.
| | - Carsten O Daub
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Stockholm, Sweden; Omics Science Center,§ RIKEN Yokohama Institute, Yokohama, Japan; Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| |
Collapse
|
21
|
Malheiros D, Panepucci RA, Roselino AM, Araújo AG, Zago MA, Petzl-Erler ML. Genome-wide gene expression profiling reveals unsuspected molecular alterations in pemphigus foliaceus. Immunology 2014; 143:381-95. [PMID: 24813052 DOI: 10.1111/imm.12315] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 12/16/2022] Open
Abstract
Pemphigus foliaceus (PF) is a complex autoimmune disease characterized by bullous skin lesions and the presence of antibodies against desmoglein 1. In this study we sought to contribute to a better understanding of the molecular processes in endemic PF, as the identification of factors that participate in the pathogenesis is a prerequisite for understanding its biological basis and may lead to novel therapeutic interventions. CD4+ T lymphocytes are central to the development of the disease. Therefore, we compared genome-wide gene expression profiles of peripheral CD4+ T cells of various PF patient subgroups with each other and with that of healthy individuals. The patient sample was subdivided into three groups: untreated patients with the generalized form of the disease, patients submitted to immunosuppressive treatment, and patients with the localized form of the disease. Comparisons between different subgroups resulted in 135, 54 and 64 genes differentially expressed. These genes are mainly related to lymphocyte adhesion and migration, apoptosis, cellular proliferation, cytotoxicity and antigen presentation. Several of these genes were differentially expressed when comparing lesional and uninvolved skin from the same patient. The chromosomal regions 19q13 and 12p13 concentrate differentially expressed genes and are candidate regions for PF susceptibility genes and disease markers. Our results reveal genes involved in disease severity, potential therapeutic targets and previously unsuspected processes involved in the pathogenesis. Besides, this study adds original information that will contribute to the understanding of PF's pathogenesis and of the still poorly defined in vivo functions of most of these genes.
Collapse
Affiliation(s)
- Danielle Malheiros
- Human Molecular Genetics Laboratory, Department of Genetics, Federal University of Paraná, Curitiba, Paraná, Brazil
| | | | | | | | | | | |
Collapse
|
22
|
Wilson SJ, Rigden HM, Ward JA, Laviolette M, Jarjour NN, Djukanović R. The relationship between eosinophilia and airway remodelling in mild asthma. Clin Exp Allergy 2014; 43:1342-50. [PMID: 24261944 DOI: 10.1111/cea.12156] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 05/31/2013] [Accepted: 06/03/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Eosinophilia is a marker of corticosteroid responsiveness and risk of exacerbation in asthma; although it has been linked to submucosal matrix deposition, its relationship with other features of airway remodelling is less clear. OBJECTIVE The aim of this study was to investigate the relationship between airway eosinophilia and airway remodelling. METHODS Bronchial biopsies from subjects (n = 20 in each group) with mild steroid-naïve asthma, with either low (0-0.45 mm(-2)) ) or high submucosal eosinophil (23.43-46.28 mm(-2) ) counts and healthy controls were assessed for in vivo epithelial damage (using epidermal growth factor receptor staining), mucin expression, airway smooth muscle (ASM) hypertrophy and inflammatory cells within ASM. RESULTS The proportion of in vivo damaged epithelium was significantly greater (P = 0.02) in the high-eosinophil (27.37%) than the low-eosinophil (4.14%) group. Mucin expression and goblet cell numbers were similar in the two eosinophil groups; however, MUC-2 expression was increased (P = 0.002) in the high-eosinophil group compared with controls. The proportion of submucosa occupied by ASM was higher in both asthma groups (P = 0.021 and P = 0.046) compared with controls. In the ASM, eosinophil and T-lymphocyte numbers were higher (P < 0.05) in the high-eosinophil group than both the low-eosinophil group and the controls, whereas the numbers of mast cells were increased in the high-eosinophil group (P = 0.01) compared with controls. CONCLUSION Submucosal eosinophilia is a marker (and possibly a cause) of epithelial damage and is related to infiltration of ASM with eosinophils and T lymphocytes, but is unrelated to mucus metaplasia or smooth muscle hypertrophy.
Collapse
Affiliation(s)
- S J Wilson
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | | | |
Collapse
|
23
|
Bérubé JC, Bossé Y. Future clinical implications emerging from recent genome-wide expression studies in asthma. Expert Rev Clin Immunol 2014; 10:985-1004. [PMID: 25001610 DOI: 10.1586/1744666x.2014.932249] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Host susceptibility to environmental triggers is the most likely explanation for the development of asthma. Quantifying gene expression levels in disease-relevant tissues and cell types using fast evolving genomic technologies have generated new hypotheses about the pathogenesis of asthma and identified new therapeutic targets to treat asthma and asthma-exacerbations. New biomarkers and distinct transcriptomic phenotypes in blood, sputum and other tissues were also identified and proved effective to refine asthma classification and guide targeted therapies. The wealth of information provided by transcriptomic studies in asthma is yet to be fully exploited, but discoveries in this field may soon be implemented in clinical settings to improve diagnosis and treatment of patients afflicted with this common disease.
Collapse
Affiliation(s)
- Jean-Christophe Bérubé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Pavillon Marguerite-d'Youville, Y4190, 2725 Chemin Ste-Foy, Quebec, Canada, G1V 4G5
| | | |
Collapse
|
24
|
McErlean P, Favoreto S, Costa FF, Shen J, Quraishi J, Biyasheva A, Cooper JJ, Scholtens DM, Vanin EF, de Bonaldo MF, Xie H, Soares MB, Avila PC. Human rhinovirus infection causes different DNA methylation changes in nasal epithelial cells from healthy and asthmatic subjects. BMC Med Genomics 2014; 7:37. [PMID: 24947756 PMCID: PMC4080608 DOI: 10.1186/1755-8794-7-37] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/18/2014] [Indexed: 01/15/2023] Open
Abstract
Background Mechanisms underlying the development of virus-induced asthma exacerbations remain unclear. To investigate if epigenetic mechanisms could be involved in virus-induced asthma exacerbations, we undertook DNA methylation profiling in asthmatic and healthy nasal epithelial cells (NECs) during Human Rhinovirus (HRV) infection in vitro. Methods Global and loci-specific methylation profiles were determined via Alu element and Infinium Human Methylation 450 K microarray, respectively. Principal components analysis identified the genomic loci influenced the most by disease-status and infection. Real-time PCR and pyrosequencing were used to confirm gene expression and DNA methylation, respectively. Results HRV infection significantly increased global DNA methylation in cells from asthmatic subjects only (43.6% to 44.1%, p = 0.04). Microarray analysis revealed 389 differentially methylated loci either based on disease status, or caused by virus infection. There were disease-associated DNA methylation patterns that were not affected by HRV infection as well as HRV-induced DNA methylation changes that were unique to each group. A common methylation locus stood out in response to HRV infection in both groups, where the small nucleolar RNA, H/ACA box 12 (SNORA12) is located. Further analysis indicated that a relationship existed between SNORA12 DNA methylation and gene expression in response to HRV infection. Conclusions We describe for the first time that Human rhinovirus infection causes DNA methylation changes in airway epithelial cells that differ between asthmatic and healthy subjects. These epigenetic differences may possibly explain the mechanism by which respiratory viruses cause asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Pedro C Avila
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
25
|
McErlean P, Berdnikovs S, Favoreto S, Shen J, Biyasheva A, Barbeau R, Eisley C, Barczak A, Ward T, Schleimer RP, Erle DJ, Boushey HA, Avila PC. Asthmatics with exacerbation during acute respiratory illness exhibit unique transcriptional signatures within the nasal mucosa. Genome Med 2014; 6:1. [PMID: 24433494 PMCID: PMC3971347 DOI: 10.1186/gm520] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/08/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute respiratory illness is the leading cause of asthma exacerbations yet the mechanisms underlying this association remain unclear. To address the deficiencies in our understanding of the molecular events characterizing acute respiratory illness-induced asthma exacerbations, we undertook a transcriptional profiling study of the nasal mucosa over the course of acute respiratory illness amongst individuals with a history of asthma, allergic rhinitis and no underlying respiratory disease. METHODS Transcriptional profiling experiments were performed using the Agilent Whole Human Genome 4X44K array platform. Time point-based microarray and principal component analyses were conducted to identify and distinguish acute respiratory illness-associated transcriptional profiles over the course of our study. Gene enrichment analysis was conducted to identify biological processes over-represented within each acute respiratory illness-associated profile, and gene expression was subsequently confirmed by quantitative polymerase chain reaction. RESULTS We found that acute respiratory illness is characterized by dynamic, time-specific transcriptional profiles whose magnitudes of expression are influenced by underlying respiratory disease and the mucosal repair signature evoked during acute respiratory illness. Most strikingly, we report that people with asthma who experience acute respiratory illness-induced exacerbations are characterized by a reduced but prolonged inflammatory immune response, inadequate activation of mucosal repair, and the expression of a newly described exacerbation-specific transcriptional signature. CONCLUSION Findings from our study represent a significant contribution towards clarifying the complex molecular interactions that typify acute respiratory illness-induced asthma exacerbations.
Collapse
Affiliation(s)
- Peter McErlean
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sergejs Berdnikovs
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Silvio Favoreto
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Junqing Shen
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Assel Biyasheva
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rebecca Barbeau
- Sandler Asthma Basic Research (SABRE) Center Functional Genomics Core Facility, University of California San Francisco, San Francisco, CA, USA
| | - Chris Eisley
- Sandler Asthma Basic Research (SABRE) Center Functional Genomics Core Facility, University of California San Francisco, San Francisco, CA, USA
| | - Andrea Barczak
- Sandler Asthma Basic Research (SABRE) Center Functional Genomics Core Facility, University of California San Francisco, San Francisco, CA, USA
| | - Theresa Ward
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Robert P Schleimer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - David J Erle
- Sandler Asthma Basic Research (SABRE) Center Functional Genomics Core Facility, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Homer A Boushey
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Pedro C Avila
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
26
|
Sircar G, Saha B, Bhattacharya SG, Saha S. Allergic asthma biomarkers using systems approaches. Front Genet 2014; 4:308. [PMID: 24409194 PMCID: PMC3884215 DOI: 10.3389/fgene.2013.00308] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 12/19/2013] [Indexed: 12/19/2022] Open
Abstract
Asthma is characterized by lung inflammation caused by complex interaction between the immune system and environmental factors such as allergens and inorganic pollutants. Recent research in this field is focused on discovering new biomarkers associated with asthma pathogenesis. This review illustrates updated research associating biomarkers of allergic asthma and their potential use in systems biology of the disease. We focus on biomolecules with altered expression, which may serve as inflammatory, diagnostic and therapeutic biomarkers of asthma discovered in human or experimental asthma model using genomic, proteomic and epigenomic approaches for gene and protein expression profiling. These include high-throughput technologies such as state of the art microarray and proteomics Mass Spectrometry (MS) platforms. Emerging concepts of molecular interactions and pathways may provide new insights in searching potential clinical biomarkers. We summarized certain pathways with significant linkage to asthma pathophysiology by analyzing the compiled biomarkers. Systems approaches with this data can identify the regulating networks, which will eventually identify the key biomarkers to be used for diagnostics and drug discovery.
Collapse
Affiliation(s)
- Gaurab Sircar
- Division of Plant Biology, Bose Institute Kolkata, India
| | | | | | - Sudipto Saha
- Bioinformatics Center, Bose Institute Kolkata, India
| |
Collapse
|
27
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 653] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Esnault S, Kelly EA, Schwantes EA, Liu LY, DeLain LP, Hauer JA, Bochkov YA, Denlinger LC, Malter JS, Mathur SK, Jarjour NN. Identification of genes expressed by human airway eosinophils after an in vivo allergen challenge. PLoS One 2013; 8:e67560. [PMID: 23844029 PMCID: PMC3699655 DOI: 10.1371/journal.pone.0067560] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/20/2013] [Indexed: 01/21/2023] Open
Abstract
Background The mechanism for the contribution of eosinophils (EOS) to asthma pathophysiology is not fully understood. Genome-wide expression analysis of airway EOS by microarrays has been limited by the ability to generate high quality RNA from sufficient numbers of airway EOS. Objective To identify, by genome-wide expression analyses, a compendium of expressed genes characteristic of airway EOS following an in vivo allergen challenge. Methods Atopic, mild asthmatic subjects were recruited for these studies. Induced sputum was obtained before and 48h after a whole lung allergen challenge (WLAC). Individuals also received a segmental bronchoprovocation with allergen (SBP-Ag) 1 month before and after administering a single dose of mepolizumab (anti-IL-5 monoclonal antibody) to reduce airway EOS. Bronchoalveolar lavage (BAL) was performed before and 48 h after SBP-Ag. Gene expression of sputum and BAL cells was analyzed by microarrays. The results were validated by qPCR in BAL cells and purified BAL EOS. Results A total of 299 transcripts were up-regulated by more than 2-fold in total BAL cells following SBP-Ag. Mepolizumab treatment resulted in a reduction of airway EOS by 54.5% and decreased expression of 99 of the 299 transcripts. 3 of 6 post-WLAC sputum samples showed increased expression of EOS-specific genes, along with the expression of 361 other genes. Finally, the intersection of the 3 groups of transcripts (increased in BAL post SBP-Ag (299), decreased after mepolizumab (99), and increased in sputum after WLAC (365)) was composed of 57 genes characterizing airway EOS gene expression. Conclusion We identified 57 genes that were highly expressed by BAL EOS compared to unseparated BAL cells after in vivo allergen challenge. 41 of these genes had not been previously described in EOS and are thus potential new candidates to elucidate EOS contribution to airway biology.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine, Allergy, Pulmonary, and Critical Care Medicine Division, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ali S, Hirschfeld AF, Mayer ML, Fortuno ES, Corbett N, Kaplan M, Wang S, Schneiderman J, Fjell CD, Yan J, Akhabir L, Aminuddin F, Marr N, Lacaze-Masmonteil T, Hegele RG, Becker A, Chan-Yeung M, Hancock REW, Kollmann TR, Daley D, Sandford AJ, Lavoie PM, Turvey SE. Functional genetic variation in NFKBIA and susceptibility to childhood asthma, bronchiolitis, and bronchopulmonary dysplasia. THE JOURNAL OF IMMUNOLOGY 2013; 190:3949-58. [PMID: 23487427 DOI: 10.4049/jimmunol.1201015] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Respiratory diseases are the most frequent chronic illnesses in babies and children. Although a vigorous innate immune system is critical for maintaining lung health, a balanced response is essential to minimize damaging inflammation. We investigated the functional and clinical impact of human genetic variants in the promoter of NFKBIA, which encodes IκBα, the major negative regulator of NF-κB. In this study, we quantified the functional impact of NFKBIA promoter polymorphisms (rs3138053, rs2233406, and rs2233409) on promoter-driven protein expression, allele-specific and total NFKBIA mRNA expression, IκBα protein expression, and TLR responsiveness; mapped innate immune regulatory networks active during respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia; and genotyped and analyzed independent cohorts of children with respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia. Genetic variants in the promoter of NFKBIA influenced NFKBIA gene expression, IκBα protein expression, and TLR-mediated inflammatory responses. Using a systems biology approach, we demonstrated that NFKBIA/IκBα is a central hub in transcriptional responses of prevalent childhood lung diseases, including respiratory syncytial virus infection, asthma, and bronchopulmonary dysplasia. Finally, by examining independent pediatric lung disease cohorts, we established that this immunologically relevant genetic variation in the promoter of NFKBIA is associated with differential susceptibility to severe bronchiolitis following infection with respiratory syncytial virus, airway hyperresponsiveness, and severe bronchopulmonary dysplasia. These data highlight the importance of negative innate immune regulators, such as NFKBIA, in pediatric lung disease and begin to unravel common aspects in the genetic predisposition to bronchopulmonary dysplasia, bronchiolitis, and childhood asthma.
Collapse
Affiliation(s)
- Salman Ali
- Department of Pediatrics, British Columbia Children's Hospital and Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia V6H 3V4, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
The ANO3/MUC15 locus is associated with eczema in families ascertained through asthma. J Allergy Clin Immunol 2013; 129:1547-53.e3. [PMID: 22657408 DOI: 10.1016/j.jaci.2012.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 04/05/2012] [Accepted: 04/09/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND A previous genome-wide linkage scan in 295 families of the French Epidemiological Study on the Genetics and Environment of Asthma (EGEA) reported strong evidence of linkage of 11p14 to eczema. OBJECTIVE Our purpose was to conduct fine-scale mapping of the 11p14 region to identify the genetic variants associated with eczema. METHODS Association analyses were first conducted in the family sample from the French EGEA by using 2 methods: the family-based association method and logistic regression. Replication of the EGEA findings was sought in French Canadian and United Kingdom family samples, which, similarly to EGEA samples, were ascertained through asthma. We also tested for association in 2 German samples ascertained through eczema. RESULTS We found significant association of eczema with 11p14 genetic variants in the vicinity of the linkage peak in EGEA (P = 10(-4) for rs1050153 by using the family-based association method, which reached the multiple testing-corrected threshold of 10(-4); P = .003 with logistic regression). Pooled analysis of the 3 asthma-ascertained samples showed strong improvement in the evidence for association (P = 6 × 10(-6) for rs293974, P = 3 × 10(-5) for rs1050153, and P = 8 × 10(-5) for rs15783). No association was observed in the eczema-ascertained samples. CONCLUSION The significant single nucleotide polymorphisms are located within the overlapping anoctamin 3 (ANO3) and mucin 15 (MUC15) genes. Several lines of evidence suggest that MUC15 is a strong candidate for eczema. Further investigation is needed to confirm our findings and to better understand the role of the ANO3/MUC15 locus in eczema and its relationship with respect to asthma.
Collapse
|
31
|
Vaillancourt VT, Bordeleau M, Laviolette M, Laprise C. From expression pattern to genetic association in asthma and asthma-related phenotypes. BMC Res Notes 2012; 5:630. [PMID: 23148572 PMCID: PMC3532380 DOI: 10.1186/1756-0500-5-630] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 11/06/2012] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Asthma is a complex disease characterized by hyperresponsiveness, obstruction and inflammation of the airways. To date, several studies using different approaches as candidate genes approach, genome wide association studies, linkage analysis and genomic expression leaded to the identification of over 300 genes involved in asthma pathophysiology. Combining results from two studies of genomic expression, this study aims to perform an association analysis between genes differently expressed in bronchial biopsies of asthmatics compared to controls and asthma-related phenotypes using the same French-Canadian Caucasian population. RESULTS Before correction, 31 of the 85 genes selected were associated with at least one asthma-related phenotype. We found four genes that survived the correction for multiple testing. The rs11630178 in aggrecan gene (AGC1) is associated with atopy (p=0.0003) and atopic asthma (p=0.0001), the rs1247653 in the interferon alpha-inducible protein 6 (IFI6), the rs1119529 in adrenergic, alpha-2A-, receptor (ADRA2A) and the rs13103321 in the alcohol dehydrogenase 1B (class I), beta polypeptide (ADH1B), are associated with asthma (p=0.019; 0.01 and 0.002 respectively). CONCLUSION To our knowledge, this is the first time those genes are associated with asthma and related traits. Consequently, our study confirms that genetic and expression studies are complementary to identify new candidate genes and to investigate their role to improve the comprehension of the complexity of asthma pathophysiology.
Collapse
Affiliation(s)
- Vanessa T Vaillancourt
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 boulevard de l'Université, Chicoutimi, Saguenay, Québec, G7H 2B1, Canada
| | | | | | | |
Collapse
|
32
|
Brunnström Å, Backman L, Tryselius Y, Claesson HE. Biosynthesis of eoxin C4 by porcine leukocytes. Prostaglandins Leukot Essent Fatty Acids 2012; 87:159-63. [PMID: 22921794 DOI: 10.1016/j.plefa.2012.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 06/11/2012] [Accepted: 07/07/2012] [Indexed: 11/23/2022]
Abstract
Human 15-lipoxygenase-1 (LO) possesses mainly 15-lipoxygenase activity whereas the animal ortholog 12/15-LO possesses mainly 12-lipoxygenase activity. These findings have raised the question if studies on animals can predict the function of 15-LO-1 in human. In this study we have characterized the arachidonic acid metabolites formed by porcine 12/15-LO. Mini pigs were infected with a parasite to increase the number of blood eosinophils, which highly express 12/15-LO. Isolated porcine polymorphonuclear leukocytes (PMNL) were incubated with arachidonic acid and the produced metabolites were analysed with HPLC and mass spectrometry (MS). The cells were found to produce 15-hydroxyeicosatetraenoic acid (HETE) and 12-HETE at a ratio of 1:5. Furthermore 8,15-dihydroxyeicosatetraenoic acids (DiHETEs) and 14,15-DiHETE were formed. Based on HPLC, UV-spectroscopy and MS analysis it was found that porcine PMNL also produced eoxin (EX) C4. These results demonstrate that although porcine 12/15-LO possesses primarily 12-lipoxygenase activity, the enzyme can catalyse the formation of EXC(4).
Collapse
Affiliation(s)
- Åsa Brunnström
- Department of Medicine, Karolinska University Hospital and Karolinska Institutet, Solna, SE-171 76 Stockholm, Sweden.
| | | | | | | |
Collapse
|
33
|
MiR-375 is downregulated in epithelial cells after IL-13 stimulation and regulates an IL-13-induced epithelial transcriptome. Mucosal Immunol 2012; 5:388-96. [PMID: 22453679 PMCID: PMC4154234 DOI: 10.1038/mi.2012.16] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interleukin 13 (IL-13)-induced epithelial gene and protein expression changes are central to the pathogenesis of multiple allergic diseases. Herein, using human esophageal squamous and bronchial columnar epithelial cells, we identified microRNAs (miRNAs) that were differentially regulated after IL-13 stimulation. Among the IL-13-regulated miRNAs, miR-375 showed a conserved pattern of downregulation. Furthermore, miR-375 was downregulated in the lung of IL-13 lung transgenic mice. We subsequently analyzed miR-375 levels in a human disease characterized by IL-13 overproduction--the allergic disorder eosinophilic esophagitis (EE)--and observed downregulation of miR-375 in EE patient samples compared with control patients. MiR-375 expression levels reflected disease activity, normalized with remission, and inversely correlated with the degree of allergic inflammation. Using a lentiviral strategy and whole-transcriptome analysis in epithelial cells, miR-375 overexpression was sufficient to markedly modify IL-13-associated immunoinflammatory pathways in epithelial cells in vitro, further substantiating interactions between miR-375 and IL-13. Taken together, our results support a key role of miRNAs, particularly miR-375, in regulating and fine-tuning IL-13-mediated responses.
Collapse
|
34
|
Abstract
Immunoglobulin E (IgE) antibodies and mast cells have been so convincingly linked to the pathophysiology of anaphylaxis and other acute allergic reactions that it can be difficult to think of them in other contexts. However, a large body of evidence now suggests that both IgE and mast cells are also key drivers of the long-term pathophysiological changes and tissue remodeling associated with chronic allergic inflammation in asthma and other settings. Such potential roles include IgE-dependent regulation of mast-cell functions, actions of IgE that are largely independent of mast cells and roles of mast cells that do not directly involve IgE. In this review, we discuss findings supporting the conclusion that IgE and mast cells can have both interdependent and independent roles in the complex immune responses that manifest clinically as asthma and other allergic disorders.
Collapse
Affiliation(s)
- Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, California, USA.
| | | |
Collapse
|
35
|
Miglino N, Roth M, Tamm M, Borger P. Asthma and COPD - The C/EBP Connection. Open Respir Med J 2012; 6:1-13. [PMID: 22715349 PMCID: PMC3377872 DOI: 10.2174/1874306401206010001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 04/06/2012] [Accepted: 04/11/2012] [Indexed: 12/11/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are the two most prominent chronic inflammatory lung diseases with increasing prevalence. Both diseases are associated with mild or severe remodeling of the airways. In this review, we postulate that the pathologies of asthma and COPD may result from inadequate responses and/or a deregulated balance of a group of cell differentiation regulating factors, the CCAAT/Enhancer Binding Proteins (C/EBPs). In addition, we will argue that the exposure to environmental factors, such as house dust mite and cigarette smoke, changes the response of C/EBPs and are different in diseased cells. These novel insights may lead to a better understanding of the etiology of the diseases and may provide new aspects for therapies.
Collapse
Affiliation(s)
| | | | | | - Peter Borger
- Pulmonary Cell Research, Departments of Biomedicine and Pneumology, University Hospital Basel,
Switzerland
| |
Collapse
|
36
|
How can microarrays unlock asthma? J Allergy (Cairo) 2012; 2012:241314. [PMID: 22500180 PMCID: PMC3303677 DOI: 10.1155/2012/241314] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 09/30/2011] [Accepted: 10/12/2011] [Indexed: 01/10/2023] Open
Abstract
Asthma is a complex disease regulated by the interplay of a large number of underlying mechanisms which contribute to the overall pathology. Despite various breakthroughs identifying genes related to asthma, our understanding of the importance of the genetic background remains limited. Although current therapies for asthma are relatively effective, subpopulations of asthmatics do not respond to these regimens. By unlocking the role of these underlying mechanisms, a source of novel and more effective treatments may be identified. In the new age of high-throughput technologies, gene-expression microarrays provide a quick and effective method of identifying novel genes and pathways, which would be impossible to discover using an individual gene screening approach. In this review we follow the history of expression microarray technologies and describe their contributions to advancing our current knowledge and understanding of asthma pathology.
Collapse
|
37
|
Yu M, Eckart MR, Morgan AA, Mukai K, Butte AJ, Tsai M, Galli SJ. Identification of an IFN-γ/mast cell axis in a mouse model of chronic asthma. J Clin Invest 2011; 121:3133-43. [PMID: 21737883 DOI: 10.1172/jci43598] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 05/11/2011] [Indexed: 01/10/2023] Open
Abstract
Asthma is considered a Th2 cell–associated disorder. Despite this, both the Th1 cell–associated cytokine IFN-γ and airway neutrophilia have been implicated in severe asthma. To investigate the relative contributions of different immune system components to the pathogenesis of asthma, we previously developed a model that exhibits several features of severe asthma in humans, including airway neutrophilia and increased lung IFN-γ. In the present studies, we tested the hypothesis that IFN-γ regulates mast cell function in our model of chronic asthma. Engraftment of mast cell–deficient KitW(-sh/W-sh) mice, which develop markedly attenuated features of disease, with wild-type mast cells restored disease pathology in this model of chronic asthma. However, disease pathology was not fully restored by engraftment with either IFN-γ receptor 1–null (Ifngr1–/–) or Fcε receptor 1γ–null (Fcer1g–/–) mast cells. Additional analysis, including gene array studies, showed that mast cell expression of IFN-γR contributed to the development of many FcεRIγ-dependent and some FcεRIγ-independent features of disease in our model, including airway hyperresponsiveness, neutrophilic and eosinophilic inflammation, airway remodeling, and lung expression of several cytokines, chemokines, and markers of an alternatively activated macrophage response. These findings identify a previously unsuspected IFN-γ/mast cell axis in the pathology of chronic allergic inflammation of the airways in mice.
Collapse
Affiliation(s)
- Mang Yu
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305-5176, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
O'Neil SE, Sitkauskiene B, Babusyte A, Krisiukeniene A, Stravinskaite-Bieksiene K, Sakalauskas R, Sihlbom C, Ekerljung L, Carlsohn E, Lötvall J. Network analysis of quantitative proteomics on asthmatic bronchi: effects of inhaled glucocorticoid treatment. Respir Res 2011; 12:124. [PMID: 21939520 PMCID: PMC3206435 DOI: 10.1186/1465-9921-12-124] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/22/2011] [Indexed: 11/10/2022] Open
Abstract
Background Proteomic studies of respiratory disorders have the potential to identify protein biomarkers for diagnosis and disease monitoring. Utilisation of sensitive quantitative proteomic methods creates opportunities to determine individual patient proteomes. The aim of the current study was to determine if quantitative proteomics of bronchial biopsies from asthmatics can distinguish relevant biological functions and whether inhaled glucocorticoid treatment affects these functions. Methods Endobronchial biopsies were taken from untreated asthmatic patients (n = 12) and healthy controls (n = 3). Asthmatic patients were randomised to double blind treatment with either placebo or budesonide (800 μg daily for 3 months) and new biopsies were obtained. Proteins extracted from the biopsies were digested and analysed using isobaric tags for relative and absolute quantitation combined with a nanoLC-LTQ Orbitrap mass spectrometer. Spectra obtained were used to identify and quantify proteins. Pathways analysis was performed using Ingenuity Pathway Analysis to identify significant biological pathways in asthma and determine how the expression of these pathways was changed by treatment. Results More than 1800 proteins were identified and quantified in the bronchial biopsies of subjects. The pathway analysis revealed acute phase response signalling, cell-to-cell signalling and tissue development associations with proteins expressed in asthmatics compared to controls. The functions and pathways associated with placebo and budesonide treatment showed distinct differences, including the decreased association with acute phase proteins as a result of budesonide treatment compared to placebo. Conclusions Proteomic analysis of bronchial biopsy material can be used to identify and quantify proteins using highly sensitive technologies, without the need for pooling of samples from several patients. Distinct pathophysiological features of asthma can be identified using this approach and the expression of these features is changed by inhaled glucocorticoid treatment. Quantitative proteomics may be applied to identify mechanisms of disease that may assist in the accurate and timely diagnosis of asthma. Trial registration ClinicalTrials.gov registration NCT01378039
Collapse
Affiliation(s)
- Serena E O'Neil
- Krefting Research Centre, Department of Internal Medicine, University of Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ueta M, Sotozono C, Kinoshita S. Expression of interleukin-4 receptor α in human corneal epithelial cells. Jpn J Ophthalmol 2011; 55:405-410. [PMID: 21617960 DOI: 10.1007/s10384-011-0030-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 01/12/2011] [Indexed: 11/30/2022]
Abstract
PURPOSE We previously reported that human conjunctival epithelial cells expressed functioning interleukin-4 receptor α (IL-4Rα). In this study, we investigated whether human corneal epithelial cells also express functioning IL-4Rα. METHODS The presence of IL-4Rα mRNA and protein in human corneal epithelium was examined by reverse-transcriptase polymerase chain reaction (RT-PCR) and immunohistology, respectively. The cell surface expression of IL-4Rα and the transcripts upregulated upon IL-4Rα ligand (IL-4 or IL-13) stimulation were examined by flow cytometry and quantitative RT-PCR, respectively, using immortalized human corneal-limbal epithelial (HCLE) cells. RESULTS The mRNA and protein of IL-4Rα were detected in human corneal epithelium. Flow cytometry analysis showed the cell surface expression of IL-4Rα protein. Quantitative RT-PCR assay of HCLE cells showed the upregulation of the transcripts tumor necrosis factor alpha-induced protein 6 (TNFAIP6), RAS guanyl-releasing protein 1 (RASGRP1), carbonic anhydrase II (CA2), cytokine-inducible SH2-containing protein (CISH), hyaluronan synthase 3 (HAS3), calpain 14 (CAPN14), endothelin receptor type A (EDNRA), cathepsin C (CTSC), and lecithin retinol acyltransferase (LRAT) as well as human conjunctival epithelial cells. CONCLUSION Human corneal epithelial cells expressed functioning IL-4Rα, and stimulation of its ligands, IL-4 and IL-13, could induce the expression of various genes, e.g., antiinflammatory molecule genes such as TNFAIP6 and CISH and cellular differentiation and proliferation-related molecule genes such as RASGRP1, HAS3, EDNRA, and LRAT.
Collapse
Affiliation(s)
- Mayumi Ueta
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan. .,Research Center for Inflammation and Regenerative Medicine, Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan.
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| | - Shigeru Kinoshita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Hirokoji-agaru, Kawaramachi-dori, Kamigyo-ku, Kyoto, 602-0841, Japan
| |
Collapse
|
40
|
Shin SW, Oh TJ, Park SM, Park JS, Jang AS, Park SW, Uh ST, An S, Park CS. Asthma-predictive genetic markers in gene expression profiling of peripheral blood mononuclear cells. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2011; 3:265-72. [PMID: 21966607 PMCID: PMC3178825 DOI: 10.4168/aair.2011.3.4.265] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 03/04/2011] [Indexed: 12/11/2022]
Abstract
PURPOSE We sought to identify asthma-related genes and to examine the potential of these genes to predict asthma, based on expression levels. METHODS The subjects were 42 asthmatics and 10 normal healthy controls. PBMC RNA was subjected to microarray analysis using a 35K array; t-tests were used to identify genes that were expressed differentially between the two groups. A multiple logistic regression analysis was applied to the differentially expressed genes, and area under the curve (AUC) values from receiver operating characteristic (ROC) curves were obtained. RESULTS In total, 170 genes were selected using the following criteria: P≤0.001 and ≥2-fold change. Among these genes, 57 were up-regulated and 113 were down-regulated in asthmatics versus normal controls. A multiple logistic regression analysis was done using more stringent criteria (P≤0.001 and ≥5-fold change), and eight genes were selected as candidate asthma biomarkers. Using these genes, 255 models (2(8)-1) were generated. Among them, only 85 showed P≤0.05 by multiple logistic regression analysis. Based on the AUCs from ROC curves for the 85 models, we found that the best model consisted of the genes MEPE, MLSTD1, and TRIM37. The model showed 0.9928 of the AUC with 98% sensitivity and 80% specificity. CONCLUSIONS MEPE, MLSTD1, and TRIM37 may be useful biomarkers for asthma.
Collapse
Affiliation(s)
- Seung Woo Shin
- Genome Research Center for Allergy and Respiratory Disease, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Choy DF, Modrek B, Abbas AR, Kummerfeld S, Clark HF, Wu LC, Fedorowicz G, Modrusan Z, Fahy JV, Woodruff PG, Arron JR. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. THE JOURNAL OF IMMUNOLOGY 2010; 186:1861-9. [PMID: 21187436 DOI: 10.4049/jimmunol.1002568] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Asthma is canonically thought of as a disorder of excessive Th2-driven inflammation in the airway, although recent studies have described heterogeneity with respect to asthma pathophysiology. We have previously described distinct phenotypes of asthma based on the presence or absence of a three-gene "Th2 signature" in bronchial epithelium, which differ in terms of eosinophilic inflammation, mucin composition, subepithelial fibrosis, and corticosteroid responsiveness. In the present analysis, we sought to describe Th2 inflammation in human asthmatic airways quantitatively with respect to known mediators of inflammation and intercellular communication. Using whole-genome microarray and quantitative real-time PCR analysis of endobronchial biopsies from 27 mild-to-moderate asthmatics and 13 healthy controls with associated clinical and demographic data, we found that asthmatic Th2 inflammation is expressed over a variable continuum, correlating significantly with local and systemic measures of allergy and eosinophilia. We evaluated a composite metric describing 79 coexpressed genes associated with Th2 inflammation against the biological space comprising cytokines, chemokines, and growth factors, identifying distinctive patterns of inflammatory mediators as well as Wnt, TGF-β, and platelet-derived growth factor family members. This integrated description of the factors regulating inflammation, cell migration, and tissue remodeling in asthmatic airways has important consequences for the pathophysiological and clinical impacts of emerging asthma therapeutics targeting Th2 inflammation.
Collapse
Affiliation(s)
- David F Choy
- Immunology, Tissue Growth, and Repair Biomarker Discovery, Genentech, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lau JY, Oliver BG, Baraket M, Beckett EL, Hansbro NG, Moir LM, Wilton SD, Williams C, Foster PS, Hansbro PM, Black JL, Burgess JK. Fibulin-1 is increased in asthma--a novel mediator of airway remodeling? PLoS One 2010; 5:e13360. [PMID: 20967215 PMCID: PMC2954173 DOI: 10.1371/journal.pone.0013360] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 09/13/2010] [Indexed: 11/24/2022] Open
Abstract
Background The extracellular matrix is a dynamic and complex network of macromolecules responsible for maintaining and influencing cellular functions of the airway. The role of fibronectin, an extracellular matrix protein, is well documented in asthma. However, the expression and function of fibulin-1, a secreted glycoprotein which interacts with fibronectin, has not been reported. Fibulin-1 is widely expressed in basement membranes in many organs including the lung. There are four isoforms in humans (A–D) of which fibulin-1C and 1D predominate. The objective of this study was to study the expression of fibulin-1 in volunteers with and without asthma, and to examine its function in vitro. Methodology/Principal Findings We used immunohistochemistry and dot-blots to examine fibulin-1 levels in bronchial biopsies, bronchoalveolar lavage fluid and serum. Real-time PCR for fibulin-1C and 1D, and ELISA and western blotting for fibulin-1 were used to study the levels in airway smooth muscle cells. The function of fibulin-1C was determined by assessing its role, using an antisense oligonucleotide, in cell proliferation, migration and wound healing. A murine model of airway hyperresponsiveness (AHR) was used to explore the biological significance of fibulin-1. Levels of fibulin-1 were significantly increased in the serum and bronchoalveolar lavage fluid of 21 asthmatics compared with 11 healthy volunteers. In addition fibulin-1 was increased in asthma derived airway smooth muscle cells and fibulin-1C contributed to the enhanced proliferation and wound repair in these cells. These features were reversed when fibulin-1C was suppressed using an antisense oligomer. In a mouse model of AHR, treatment with an AO inhibited the development of AHR to methacholine. Conclusions Our data collectively suggest fibulin-1C may be worthy of further investigation as a target for airway remodeling in asthma.
Collapse
Affiliation(s)
- Justine Y. Lau
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
| | - Brian G. Oliver
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Melissa Baraket
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Emma L. Beckett
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Nicole G. Hansbro
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Lyn M. Moir
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Steve D. Wilton
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, Perth, Western Australia, Australia
- Lung Institute of Western Australia and the Centre for Asthma, Allergy and Respiratory Research, University of Western Australia, Perth, Western Australia, Australia
| | - Carolyn Williams
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Lung Institute of Western Australia and the Centre for Asthma, Allergy and Respiratory Research, University of Western Australia, Perth, Western Australia, Australia
| | - Paul S. Foster
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Philip M. Hansbro
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Centre for Asthma and Respiratory Disease and Hunter Medical Research Institute, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Judith L. Black
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
| | - Janette K. Burgess
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- Discipline of Pharmacology, The University of Sydney, Sydney, New South Wales, Australia
- Woolcock Institute of Medical Research, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
43
|
Madore AM, Laprise C. Immunological and genetic aspects of asthma and allergy. J Asthma Allergy 2010; 3:107-21. [PMID: 21437045 PMCID: PMC3047903 DOI: 10.2147/jaa.s8970] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Indexed: 12/21/2022] Open
Abstract
Prevalence of allergy and allergic asthma are increasing worldwide. More than half of the US population has a positive skin prick test and approximately 10% are asthmatics. Many studies have been conducted to define immunological pathways underlying allergy and asthma development and to identify the main genetic determinants. In the effort to find missing pieces of the puzzle, new genomic approaches and more standardized ones, such as the candidate gene approach, have been used collectively. This article proposes an overview of the actual knowledge about immunological and genetic aspects of allergy and asthma. Special attention has been drawn to the challenges linked to genetic research in complex traits such as asthma and to the contribution of new genomic approaches.
Collapse
Affiliation(s)
- Anne-Marie Madore
- Université du Québec à Chicoutimi, Département des sciences fondamentales, Saguenay, Canada
| | | |
Collapse
|
44
|
Auffray C, Adcock IM, Chung KF, Djukanovic R, Pison C, Sterk PJ. An integrative systems biology approach to understanding pulmonary diseases. Chest 2010; 137:1410-6. [PMID: 20525651 DOI: 10.1378/chest.09-1850] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Chronic inflammatory pulmonary diseases such as COPD and asthma are highly prevalent and associated with a major health burden worldwide. Despite a wealth of biologic and clinical information on normal and pathologic airway structure and function, the primary causes and mechanisms of disease remain to a large extent unknown, preventing the development of more efficient diagnosis and treatment. We propose to overcome these limitations through an integrative systems biology research strategy designed to identify the functional and regulatory pathways that play central roles in respiratory pathophysiology, starting with severe asthma. This approach relies on global genome, transcriptome, proteome, and metabolome data sets collected in cross-sectional patient cohorts with high-throughput measurement platforms and integrated with biologic and clinical data to inform predictive multiscale models ranging from the molecular to the organ levels. Working hypotheses formulated on the mechanisms and pathways involved in various disease states are tested through perturbation experiments using model simulation combined with targeted and global technologies in cellular and animal models. The responses observed are compared with those predicted by the initial models, which are refined to account better for the results. Novel perturbation experiments are designed and tested both computationally and experimentally to arbitrate between competing hypotheses. The process is iterated until the derived knowledge allows a better classification and subphenotyping of severe asthma using complex biomarkers, which will facilitate the development of novel diagnostic and therapeutic interventions targeting multiple components of the molecular and cellular pathways involved. This can be tested and validated in prospective clinical trials.
Collapse
Affiliation(s)
- Charles Auffray
- Functional Genomics and Systems Biology for Health, CNRS Institute of Biological Sciences, 7 rue Guy Moquet-BP8, 94801 Villejuif cedex, France.
| | | | | | | | | | | |
Collapse
|
45
|
Plager DA, Kahl JC, Asmann YW, Nilson AE, Pallanch JF, Friedman O, Kita H. Gene transcription changes in asthmatic chronic rhinosinusitis with nasal polyps and comparison to those in atopic dermatitis. PLoS One 2010; 5:e11450. [PMID: 20625511 PMCID: PMC2897889 DOI: 10.1371/journal.pone.0011450] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 05/12/2010] [Indexed: 01/21/2023] Open
Abstract
Background Asthmatic chronic rhinosinusitis with nasal polyps (aCRSwNP) is a common disruptive eosinophilic disease without effective medical treatment. Therefore, we sought to identify gene expression changes, particularly those occurring early, in aCRSwNP. To highlight expression changes associated with eosinophilic epithelial inflammation, we further compared the changes in aCRSwNP with those in a second eosinophilic epithelial disease, atopic dermatitis (AD), which is also closely related to asthma. Methods/Principal Findings Genome-wide mRNA levels measured by exon array in both nasosinus inflamed mucosa and adjacent polyp from 11 aCRSwNP patients were compared to those in nasosinus tissue from 17 normal or rhinitis subjects without polyps. Differential expression of selected genes was confirmed by qRT-PCR or immunoassay, and transcription changes common to AD were identified. Comparison of aCRSwNP inflamed mucosa and polyp to normal/rhinitis tissue identified 447 differentially transcribed genes at ≥2 fold-change and adjusted p-value<0.05. These included increased transcription of chemokines localized to chromosome 17q11.2 (CCL13, CCL2, CCL8, and CCL11) that favor eosinophil and monocyte chemotaxis and chemokines (CCL18, CCL22, and CXCL13) that alternatively-activated monocyte-derived cells have been shown to produce. Additional transcription changes likely associated with Th2-like eosinophilic inflammation were prominent and included increased IL1RL1 (IL33 receptor) and EMR1&3 and decreased CRISP2&3. A down-regulated PDGFB-centric network involving several smooth muscle-associated genes was also implicated. Genes at 17q11.2, genes associated with alternative activation or smooth muscle, and the IL1RL1 gene were also differentially transcribed in AD. Conclusions/Significance Our data implicate several genes or gene sets in aCRSwNP and eosinophilic epithelial inflammation, some that likely act in the earlier stages of inflammation. The identified gene expression changes provide additional diagnostic and therapeutic targets for aCRSwNP and other eosinophilic epithelial diseases.
Collapse
Affiliation(s)
- Douglas A Plager
- Allergic Diseases Research Laboratory, Mayo Clinic, Rochester, Minnesota, United States of America.
| | | | | | | | | | | | | |
Collapse
|
46
|
A microarray study of gene expression profiles in nasal polyps. Auris Nasus Larynx 2010; 38:58-64. [PMID: 20554417 DOI: 10.1016/j.anl.2010.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Revised: 03/18/2010] [Accepted: 05/11/2010] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Nasal polyposis (NP) is a multifactorial disease manifesting in chronic inflammation of upper respiratory tract of unknown etiology. We studied mRNA gene expression profiles in NP compared with normal mucosa as well as pointed at genes characteristic of different expression in examined tissues. MATERIAL AND METHODS Fifty-three patients with NP (36 eosinophilic and 17 neutrophilic NP) were included into the study. Transcriptional activity of genes was analyzed using oligonucleotide microarray in 17 NP and 8 cases of normal nasal mucosa. A study of mRNA expression of selected genes was performed using QRT-PCR. RESULTS We identified 556 genes, which were differentially expressed between the studied and the control group. Among them 217 showed significantly higher expression, whereas 339 lower expression in NP than in controls. The microarray and QRT-PCR results were compatible for 7 of 8 evaluated genes. In NP strongly significant higher transcriptional activity of MMP10, NOS2A, ALOX15 and IL-8 genes was observed. In the control group, significantly higher expression of DMBT1, ALOX12 and LTF genes was detected. CONCLUSION The analysis of gene expression in inflammatory changed nasal polyp tissues may become a supplementary method in diagnostics and treatment. Molecular alterations may indicate changes during the clinical course of the disease.
Collapse
|
47
|
Kicic A, Hallstrand TS, Sutanto EN, Stevens PT, Kobor MS, Taplin C, Paré PD, Beyer RP, Stick SM, Knight DA. Decreased fibronectin production significantly contributes to dysregulated repair of asthmatic epithelium. Am J Respir Crit Care Med 2010; 181:889-98. [PMID: 20110557 DOI: 10.1164/rccm.200907-1071oc] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Damage to airway epithelium is followed by deposition of extracellular matrix (ECM) and migration of adjacent epithelial cells. We have shown that epithelial cells from children with asthma fail to heal a wound in vitro. OBJECTIVES To determine whether dysregulated ECM production by the epithelium plays a role in aberrant repair in asthma. METHODS Airway epithelial cells (AEC) from children with asthma (n = 36), healthy atopic control subjects (n = 23), and healthy nonatopic control subjects (n = 53) were investigated by microarray, gene expression and silencing, transcript regulation analysis, and ability to close mechanical wounds. MEASUREMENTS AND MAIN RESULTS Time to repair a mechanical wound in vitro by AEC from healthy and atopic children was not significantly different and both were faster than AEC from children with asthma. Microarray analysis revealed differential expression of multiple gene sets associated with repair and remodeling in asthmatic AEC. Fibronectin (FN) was the only ECM component whose expression was significantly lower in asthmatic AEC. Expression differences were verified by quantitative polymerase chain reaction and ELISA, and reduced FN expression persisted in asthmatic cells over passage. Silencing of FN expression in nonasthmatic AEC inhibited wound repair, whereas addition of FN to asthmatic AEC restored reparative capacity. Asthmatic AEC failed to synthesize FN in response to wounding or cytokine/growth factor stimulation. Exposure to 5', 2'deoxyazacytidine had no effect on FN expression and subsequent analysis of the FN promoter did not show evidence of DNA methylation. CONCLUSIONS These data show that the reduced capacity of asthmatic epithelial cells to secrete FN is an important contributor to the dysregulated AEC repair observed in these cells.
Collapse
Affiliation(s)
- Anthony Kicic
- Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, 6001, Western Australia, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hallstrand TS, Wurfel MM, Lai Y, Ni Z, Gelb MH, Altemeier WA, Beyer RP, Aitken ML, Henderson WR. Transglutaminase 2, a novel regulator of eicosanoid production in asthma revealed by genome-wide expression profiling of distinct asthma phenotypes. PLoS One 2010; 5:e8583. [PMID: 20052409 PMCID: PMC2797392 DOI: 10.1371/journal.pone.0008583] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 12/08/2009] [Indexed: 11/21/2022] Open
Abstract
Background A frequent manifestation of asthma, exercise-induced bronchoconstriction (EIB), occurs in 30–50% of asthmatics and is characterized by increased release of inflammatory eicosanoids. The objective of this study was to identify genes differentially expressed in EIB and to understand the function of these genes in the biology of asthma. Methodology/Principal Findings Genome-wide expression profiling of airway leukocytes and epithelial cells obtained by induced sputum was conducted in two groups of subjects with asthma with and without EIB (n = 7 per group), at baseline and following exercise challenge. Based on the results of the gene expression study, additional comparisons were made with a normal control group (n = 10). Localization studies were conducted on epithelial brushings and biopsies from an additional group of asthmatics with EIB (n = 3). Genes related to epithelial repair and mast cell infiltration including β-tryptase and carboxypeptidase A3 were upregulated by exercise challenge in the asthma group with EIB. A gene novel to asthma pathogenesis, transglutaminase 2 (TGM2), was the most differentially expressed at baseline between the groups. In vivo studies confirmed the increased expression of TGM2 in airway cells and airway lining fluid, and demonstrate that TGM2 is avidly expressed in the asthmatic airway epithelium. In vitro studies using recombinant human enzymes reveal that TGM2 augments the enzymatic activity of secreted phospholipase A2 (PLA2) group X (sPLA2-X), an enzyme recently implicated in asthma pathogenesis. Conclusions/Significance This study found that TGM2, a mediator that is novel to asthma pathogenesis, is overexpressed in asthmatic airways and functions to increase sPLA2-X enzymatic activity. Since PLA2 serves as the first rate-limiting step leading to eicosanoid formation, these results suggest that TGM2 may be a key initiator of the airway inflammatory cascade in asthma.
Collapse
Affiliation(s)
- Teal S Hallstrand
- Division of Pulmonary and Critical Care, Department of Medicine, University of Washington, Seattle, Washington, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chamberland A, Madore AM, Tremblay K, Laviolette M, Laprise C. A comparison of two sets of microarray experiments to define allergic asthma expression pattern. Exp Lung Res 2009; 35:399-410. [PMID: 19842841 DOI: 10.1080/01902140902745174] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Allergic asthma is a complex trait. Several approaches have been used to identify biomarkers involved in this disease. This study aimed at demonstrating the relevance and validity of microarrays in the definition of allergic asthma expression pattern. The authors compared the transcript expressions of bronchial biopsy of 2 different microarray experiments done 2 years apart, both including nonallergic healthy and allergic asthmatic subjects (n = 4 in each experiment). U95Av2 and U133A GeneChips detected respectively 89 and 40 differentially expressed genes. Fifty-five percent of the U133A genes were previously identified with the U95Av2 arrays. The immune signaling molecules and the proteolytic enzymes were the most preserved categories between the 2 experiments, because 3/4 of the genes identified by the U133A were also significant in the U95Av2 study for both categories. These results demonstrate the relevance of microarray experiments using bronchial tissues in allergic asthma. The comparison of these GeneChip studies suggests that earlier microarray results are as relevant as actual ones to target new genes of interest, particularly in function categories linked to the studied disease. Moreover, it demonstrates that microarrays are a valuable technology to target novel allergic asthma pathways as well as biomarkers.
Collapse
Affiliation(s)
- Annie Chamberland
- Département des Sciences Fondamentales, Université du Québec à Chicoutimi, Chicoutimi, Quebec, Canada
| | | | | | | | | |
Collapse
|
50
|
Array of hope: expression profiling identifies disease biomarkers and mechanism. Biochem Soc Trans 2009; 37:855-62. [PMID: 19614607 DOI: 10.1042/bst0370855] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
High-throughput, genome-wide analytical technologies are now commonly used in all fields of medical research. The most commonly applied of these technologies, gene expression microarrays, have been shown to be both accurate and precise when properly implemented. For over a decade, microarrays have provided novel insight into many complex human diseases. Microarray-based discovery can be classified into three components, biomarker detection, disease (sub)classification and identification of causal mechanism, in order of accomplishment. Within the respiratory system, the application of microarrays has achieved significant success in all components, particularly with respect to lung cancer. Numerous studies over the last half-decade have applied this technology to the characterization of non-malignant respiratory diseases, animal models of respiratory disease and normal developmental processes. Studies of obstructive lung diseases by many groups, including our own, have yielded not only disease biomarkers, but also some novel putative pathogenic mechanisms. We have successfully used an integrative genomics approach, combining microarray analysis with human genetics, to identify susceptibility genes for COPD (chronic obstructive pulmonary disease). Interestingly, we find that the assessment of quantitative phenotypic variables enhances gene discovery. Our studies contribute to the identification of obstructive lung disease biomarkers, provide data associated with disease phenotypes and support the use of an integrated approach to move beyond marker identification to mechanism discovery.
Collapse
|