1
|
Del Duca E, Dahabreh D, Kim M, Bar J, Da Rosa JC, Rabinowitz G, Facheris P, Gómez-Arias PJ, Chang A, Utti V, Chowdhury A, Liu Y, Estrada YD, Laculiceanu A, Agache I, Guttman-Yassky E. Transcriptomic evaluation of skin tape-strips in children with allergic asthma uncovers epidermal barrier dysfunction and asthma-associated biomarkers abnormalities. Allergy 2024; 79:1516-1530. [PMID: 38375886 PMCID: PMC11247382 DOI: 10.1111/all.16060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/08/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Tape-strips, a minimally invasive method validated for the evaluation of several skin diseases, may help identify asthma-specific biomarkers in the skin of children with allergic asthma. METHODS Skin tape-strips were obtained and analyzed with RNA-Seq from children with moderate allergic asthma (MAA) (n = 11, mean age 7.00; SD = 1.67), severe allergic asthma (SAA) (n = 9, mean age 9.11; SD = 2.37), and healthy controls (HCs) (n = 12, mean age 7.36; SD = 2.03). Differentially expressed genes (DEGs) were identified by fold change ≥2 with a false discovery rate <0.05. Transcriptomic biomarkers were analyzed for their accuracy in distinguishing asthma from HCs, their relationships with asthma-related outcomes (exacerbation rate, lung function-FEV1, IOS-R5-20, and lung inflammation-FeNO), and their links to skin (barrier and immune response) and lung (remodeling, metabolism, aging) pathogenetic pathways. RESULTS RNA-Seq captured 1113 in MAA and 2117 DEGs in SAA. Epidermal transcriptomic biomarkers for terminal differentiation (FLG/filaggrin), cell adhesion (CDH19, JAM2), lipid biosynthesis/metabolism (ACOT2, LOXL2) were significantly downregulated. Gene set variation analysis revealed enrichment of Th1/IFNγ pathways (p < .01). MAA and SAA shared downregulation of G-protein-coupled receptor (OR4A16, TAS1R3), upregulation of TGF-β/ErbB signaling-related (ACVR1B, EGFR, ID1/2), and upregulation of mitochondrial-related (HIGD2A, VDAC3, NDUFB9) genes. Skin transcriptomic biomarkers correlated with the annualized exacerbation rate and with lung function parameters. A two-gene classifier (TSSC4-FAM212B) was able to differentiate asthma from HCs with 100% accuracy. CONCLUSION Tape-strips detected epithelial barrier and asthma-associated signatures in normal-appearing skin from children with allergic asthma and may serve as an alternative to invasive approaches for evaluating asthma endotypes.
Collapse
Affiliation(s)
- Ester Del Duca
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Dante Dahabreh
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Madeline Kim
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Jonathan Bar
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Joel Correa Da Rosa
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Grace Rabinowitz
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Paola Facheris
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
- Department of Dermatology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Pedro Jesús Gómez-Arias
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
- Department of Dermatology, Reina Sofía University Hospital, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Annie Chang
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Vivian Utti
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Amira Chowdhury
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Ying Liu
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Yeriel D Estrada
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| | - Alexandru Laculiceanu
- Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Ioana Agache
- Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at the Mount Sinai, New York, New York, USA
| |
Collapse
|
2
|
Gayen S, Dachert S, Lashari BH, Gordon M, Desai P, Criner GJ, Cardet JC, Shenoy K. Critical Care Management of Severe Asthma Exacerbations. J Clin Med 2024; 13:859. [PMID: 38337552 PMCID: PMC10856115 DOI: 10.3390/jcm13030859] [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: 01/11/2024] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Severe asthma exacerbations, including near-fatal asthma (NFA), have high morbidity and mortality. Mechanical ventilation of patients with severe asthma is difficult due to the complex pathophysiology resulting from severe bronchospasm and dynamic hyperinflation. Life-threatening complications of traditional ventilation strategies in asthma exacerbations include the development of systemic hypotension from hyperinflation, air trapping, and pneumothoraces. Optimizing pharmacologic techniques and ventilation strategies is crucial to treat the underlying bronchospasm. Despite optimal pharmacologic management and mechanical ventilation, the mortality rate of patients with severe asthma in intensive care units is 8%, suggesting a need for advanced non-pharmacologic therapies, including extracorporeal life support (ECLS). This review focuses on the pathophysiology of acute asthma exacerbations, ventilation management including non-invasive ventilation (NIV) and invasive mechanical ventilation (IMV), the pharmacologic management of acute asthma, and ECLS. This review also explores additional advanced non-pharmacologic techniques and monitoring tools for the safe and effective management of critically ill adult asthmatic patients.
Collapse
Affiliation(s)
- Shameek Gayen
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Stephen Dachert
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Bilal H. Lashari
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Matthew Gordon
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Parag Desai
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Gerard J. Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL 33602, USA;
| | - Kartik Shenoy
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University Hospital, Philadelphia, PA 19140, USA; (S.D.); (B.H.L.); (M.G.); (P.D.); (G.J.C.); (K.S.)
| |
Collapse
|
3
|
Pathak MP, Patowary P, Chattopadhyay P, Barbhuiyan PA, Islam J, Gogoi J, Wankhar W. Obesity-associated Airway Hyperresponsiveness: Mechanisms Underlying Inflammatory Markers and Possible Pharmacological Interventions. Endocr Metab Immune Disord Drug Targets 2024; 24:1053-1068. [PMID: 37957906 DOI: 10.2174/0118715303256440231028072049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 11/15/2023]
Abstract
Obesity is rapidly becoming a global health problem affecting about 13% of the world's population affecting women and children the most. Recent studies have stated that obese asthmatic subjects suffer from an increased risk of asthma, encounter severe symptoms, respond poorly to anti-asthmatic drugs, and ultimately their quality-of-life decreases. Although, the association between airway hyperresponsiveness (AHR) and obesity is a growing concern among the public due to lifestyle and environmental etiologies, however, the precise mechanism underlying this association is yet to establish. Apart from aiming at the conventional antiasthmatic targets, treatment should be directed towards ameliorating obesity pathogenesis too. Understanding the pathogenesis underlying the association between obesity and AHR is limited, however, a plethora of obesity pathologies have been reported viz., increased pro-inflammatory and decreased anti-inflammatory adipokines, depletion of ROS controller Nrf2/HO-1 axis, NLRP3 associated macrophage polarization, hypertrophy of WAT, and down-regulation of UCP1 in BAT following down-regulated AMPKα and melanocortin pathway that may be correlated with AHR. Increased waist circumference (WC) or central obesity was thought to be related to severe AHR, however, some recent reports suggest body mass index (BMI), not WC tends to exaggerate airway closure in AHR due to some unknown mechanisms. This review aims to co-relate the above-mentioned mechanisms that may explain the copious relation underlying obesity and AHR with the help of published reports. A proper understanding of these mechanisms discussed in this review will ensure an appropriate treatment plan for patients through advanced pharmacological interventions.
Collapse
Affiliation(s)
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India
| | | | | | - Johirul Islam
- Department of Pharmaceutical Sciences, School of Health Sciences, Assam Kaziranga University, Jorhat, India
| | - Jyotchna Gogoi
- Department of Biochemistry, Faculty of Science, Assam Down Town University, Guwahati, India
| | - Wankupar Wankhar
- Department of Dialysis, Faculty of Paramedical Science, Assam Down Town University, Guwahati, India
| |
Collapse
|
4
|
Cai Y, Tian J, Su Y, Shi X. MiR-506 targets polypyrimidine tract-binding protein 1 to inhibit airway inflammatory response and remodeling via mediating Wnt/β-catenin signaling pathway. Allergol Immunopathol (Madr) 2023; 51:15-24. [PMID: 37169555 DOI: 10.15586/aei.v51i3.676] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/12/2022] [Indexed: 05/13/2023]
Abstract
BACKGROUND Airway remodeling, which contributes to the clinical course of childhood asthma, occurs due to airway inflammation and is featured by anomalous biological behaviors of airway smooth muscle cells (ASMCs). microRNA (miRNA) plays an essential role in the etiopathogenesis of asthma. OBJECTIVE This research was aimed to characterize miR-506 in asthma and uncover potential regulatory machinery. MATERIAL AND METHODS The asthmatic cell model was established by treating ASMCs with transforming growth factor-beta1 (TGF-β1) and assessed by the levels of interleukin (IL)-1β and interferon gamma (IFN-γ). Using real-time quantitative polymerase chain reaction, mRNA expression of miR-506 and polypyrimidine tract-binding protein 1 (PTBP1) was measured. Cell counting kit-8 and Transwell migration tests were used for estimating the capacity of ASMCs to proliferate and migrate. Luciferase reporter assay was used to corroborate whether miR-506 was directly bound to PTBP1. Expression of PTBP1, collagen I and III, and essential proteins of the wingless-related integration (Wnt)/β-catenin pathway (β-catenin, c-MYC and cyclin D1) was accomplished by Western blot analysis. The involvement of Wnt/β-catenin signaling in asthma was confirmed by Wnt signaling pathway inhibitor (IWR-1). RESULTS miR-506 was poorly expressed in asthmatic tissues and cell model. Functionally, overexpression of miR-506 reduced aberrant proliferation, migration, inflammation and collagen deposition of ASMCs triggered by TGF-β1. Mechanically, miR-506 directly targeted the 3' untranslated region (3-UTR) of PTBP1 and had a negative regulation on PTBP1 expression. Moreover, overexpression of miR-506 suppressed the induction of Wnt/β-catenin pathway. The administration of IWR-1 further validated negative correlation between miR-506 and the Wnt/β-catenin pathway in asthma. CONCLUSION Our data indicated that targeting miR-506/PTBP1/Wnt/β-catenin axis might point in a helpful direction for treating asthma in children.
Collapse
Affiliation(s)
- Yuxiang Cai
- Department of Emergency, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Jifeng Tian
- Department of Integrated Traditional Chinese and Western Medicine, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Yufei Su
- Department of Emergency, Xi'an Children's Hospital, Xi'an, Shaanxi, China
| | - Xiaolan Shi
- Department of Respiratory Asthma Center, Xi'an Children's Hospital, Xi'an, Shaanxi, China;
| |
Collapse
|
5
|
Lee HW, Baek MG, Choi S, Ahn YH, Bang JY, Sohn KH, Kang MG, Jung JW, Choi JH, Cho SH, Yi H, Kang HR. Peripheral blood transcriptomic clusters uncovered immune phenotypes of asthma. Respir Res 2022; 23:237. [PMID: 36076228 PMCID: PMC9461267 DOI: 10.1186/s12931-022-02156-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transcriptomic analysis has been used to elucidate the complex pathogenesis of heterogeneous disease and may also contribute to identify potential therapeutic targets by delineating the hub genes. This study aimed to investigate whether blood transcriptomic clustering can distinguish clinical and immune phenotypes of asthmatics, and microbiome in asthmatics. METHODS Transcriptomic expression of peripheral blood mononuclear cells (PBMCs) from 47 asthmatics and 21 non-asthmatics was measured using RNA sequencing. A hierarchical clustering algorithm was used to classify asthmatics. Differentially expressed genes, clinical phenotypes, immune phenotypes, and microbiome of each transcriptomic cluster were assessed. RESULTS In asthmatics, three distinct transcriptomic clusters with numerously different transcriptomic expressions were identified. The proportion of severe asthmatics was highest in cluster 3 as 73.3%, followed by cluster 2 (45.5%) and cluster 1 (28.6%). While cluster 1 represented clinically non-severe T2 asthma, cluster 3 tended to include severe non-T2 asthma. Cluster 2 had features of both T2 and non-T2 asthmatics characterized by the highest serum IgE level and neutrophil-dominant sputum cell population. Compared to non-asthmatics, cluster 1 showed higher CCL23 and IL1RL1 expression while the expression of TREML4 was suppressed in cluster 3. CTSD and ALDH2 showed a significant positive linear relationship across three clusters in the order of cluster 1 to 3. No significant differences in the diversities of lung and gut microbiomes were observed among transcriptomic clusters of asthmatics and non-asthmatics. However, our study has limitations in that small sample size data were analyzed with unmeasured confounding factors and causal relationships or function pathways were not verified. CONCLUSIONS Genetic clustering based on the blood transcriptome may provide novel immunological insight, which can be biomarkers of asthma immune phenotypes. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Hyun Woo Lee
- Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Min-Gyung Baek
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Korea
| | - Sungmi Choi
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Korea
| | - Yoon Hae Ahn
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul, 110-744, Korea
| | - Ji-Young Bang
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Kyoung-Hee Sohn
- Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Min-Gyu Kang
- Department of Internal Medicine, Chungbuk National University College of Medicine, Chungbuk National University Hospital, Cheongju, Korea
| | - Jae-Woo Jung
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Jeong-Hee Choi
- Department of Pulmonology and Allergy, Allergy and Clinical Immunology Research Center, Hallym University College of Medicine, Chuncheon, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul, 110-744, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Hana Yi
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, Korea. .,School of Biosystems and Biomedical Sciences, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Korea.
| | - Hye-Ryun Kang
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-Gu, Seoul, 110-744, Korea. .,Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea. .,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Mo BW, Li XM, Li SM, Xiao B, Yang J, Li HM. m6A echoes with DNA methylation: Coordinated DNA methylation and gene expression data analysis identified critical m6A genes associated with asthma. Gene 2022; 828:146457. [PMID: 35421547 DOI: 10.1016/j.gene.2022.146457] [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: 10/21/2021] [Revised: 01/30/2022] [Accepted: 03/25/2022] [Indexed: 11/04/2022]
Abstract
Asthma is a chronic inflammatory disease that involves complex gene-environment interactions. Methylation of nucleotides, such as 5-methylcytosine (5mC) in DNA and N6-methyladenosine (m6A) in mRNA, carries important information for gene regulation. Our study screened m6A genes and genes associated with asthma from the Gene Expression Omnibus (GEO) databases GSE63383, GSE119580, GSE38003, GSE34313, GSE13168, and GSE35643. GSE52778, GSE35643, GSE40996, and GSE64744), and DNA methylation data from GSE85568 and GSE146377. We screened out 6 m6A related genes (FTO, IGF2BP2, RBM15, RBMX, WTAP, and YTHDC1) that were significantly dysregulated in asthma or proinflammatory conditions. A correlation study showed a high correlation between m6A genes and gene pairs such as WTAP, IL7R, and TLR2; RBMX, SLC22A4, IL33, TNC, FLG, and IL6R (|r| ≥ 0.8). Following DNA methylation dataset analysis, we proposed several DNA methylation-m6A modification asthma-related gene axes such as cg19032951/cg15153914-IGF2BP2-SMAD3. Interestingly, several target genes, such as SMAD3, possess the ability to participate in DNA methylation processes, which may reciprocally regulate the expression of m6A genes and form a closed-loop regulation axis. Some classic DNA methylation-related genes, such as TET1, UHRF1, and ZBTB4, were also involved. We identified an integrated profile of m6A gene expression in asthma and proposed a novel potential interplay between DNA methylation and m6A modification in asthma pathogenesis. Using the CMAP database, we found that resveratrol may target these dysregulated m6A genes, and therefore may serve as a potential therapeutic agent for asthma.
Collapse
Affiliation(s)
- Bi-Wen Mo
- The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, PR China; Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, PR China
| | - Xiao-Mang Li
- Guilin Medical University, Guilin 541000, PR China
| | - Shen-Mei Li
- Guilin Medical University, Guilin 541000, PR China
| | - Bo Xiao
- Affiliated Hospital of Guilin Medical University, Guilin 541000, PR China; Key Laboratory of Respiratory Diseases (Guilin Medical University, Education Department of Guangxi Zhuang Autonomous Region), PR China
| | - Jie Yang
- Guilin Medical University, Guilin 541000, PR China
| | - Hui-Min Li
- Guilin Medical University, Guilin 541000, PR China.
| |
Collapse
|
7
|
Development and validation of an RNA-seq-based transcriptomic risk score for asthma. Sci Rep 2022; 12:8643. [PMID: 35606385 PMCID: PMC9126925 DOI: 10.1038/s41598-022-12199-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022] Open
Abstract
Recent progress in RNA sequencing (RNA-seq) allows us to explore whole-genome gene expression profiles and to develop predictive model for disease risk. The objective of this study was to develop and validate an RNA-seq-based transcriptomic risk score (RSRS) for disease risk prediction that can simultaneously accommodate demographic information. We analyzed RNA-seq gene expression data from 441 asthmatic and 254 non-asthmatic samples. Logistic least absolute shrinkage and selection operator (Lasso) regression analysis in the training set identified 73 differentially expressed genes (DEG) to form a weighted RSRS that discriminated asthmatics from healthy subjects with area under the curve (AUC) of 0.80 in the testing set after adjustment for age and gender. The 73-gene RSRS was validated in three independent RNA-seq datasets and achieved AUCs of 0.70, 0.77 and 0.60, respectively. To explore their biological and molecular functions in asthma phenotype, we examined the 73 genes by enrichment pathway analysis and found that these genes were significantly (p < 0.0001) enriched for DNA replication, recombination, and repair, cell-to-cell signaling and interaction, and eumelanin biosynthesis and developmental disorder. Further in-silico analyses of the 73 genes using Connectivity map shows that drugs (mepacrine, dactolisib) and genetic perturbagens (PAK1, GSR, RBM15 and TNFRSF12A) were identified and could potentially be repurposed for treating asthma. These findings show the promise for RNA-seq risk scores to stratify and predict disease risk.
Collapse
|
8
|
Diana P, Carvalheira GMG. NIBAN1, Exploring its Roles in Cell Survival Under Stress Context. Front Cell Dev Biol 2022; 10:867003. [PMID: 35517496 PMCID: PMC9062034 DOI: 10.3389/fcell.2022.867003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Cell survival must quickly activate specific mechanisms that enable to detect changes in the cellular microenvironment. The impact of these cell alteration has direct consequences on cellular homeostasis. Cellular stress, as well as its regulation and implication, has been studied in different pathologies. In this sense, the alteration in NIBAN1 expression seems to act in response to different cellular disturbances. Over the years, the knowledge of NIBAN1 functions has improved, demonstrating its important cell roles, favoring the cell survival under stress context. In response to the disturbances, NIBAN1 seems to be involved in the decision-making process between cell survival and death. The increase in NIBAN1 expression has been related to cellular mechanisms that seek to minimize the damage caused to cellular homeostasis. In this review, the main biological insights attributed to the NIBAN1 gene in different cellular contexts and its role as a mediator of cellular stress are discussed.
Collapse
|
9
|
Adulthood asthma as a consequence of childhood adversity: a systematic review of epigenetically affected genes. J Dev Orig Health Dis 2022; 13:674-682. [PMID: 35256035 DOI: 10.1017/s2040174422000083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
There is an accumulating data that shows relation between childhood adversity and vulnerability to chronic diseases as well as epigenetic influences that in turn give rise to these diseases. Asthma is one of the chronic diseases that is influenced from genetic regulation of the inflammatory biomolecules and therefore the hypothesis in this research was childhood adversity might have caused epigenetic differentiation in the asthma-related genes in the population who had childhood trauma. To test this hypothesis, the literature was systematically reviewed to extract epigenetically modified gene data of the adults who had childhood adversity, and affected genes were further evaluated for their association with asthma. PRISMA guidelines were adopted and PubMed and Google Scholar were included in the searched databases, to evaluate epigenetic modifications in asthma-related genes of physically, emotionally or sexually abused children. After retrieving a total of 5245 articles, 36 of them were included in the study. Several genes and pathways that may contribute to pathogenesis of asthma development, increased inflammation, or response to asthma treatment were found epigenetically affected by childhood traumas. Childhood adversity, causing epigenetic changes in DNA, may lead to asthma development or influence the course of the disease and therefore should be taken into account for the prolonged health consequences.
Collapse
|
10
|
Huang JQ, Wang F, Wang LT, Li YM, Lu JL, Chen JY. Circular RNA ERBB2 Contributes to Proliferation and Migration of Airway Smooth Muscle Cells via miR-98-5p/IGF1R Signaling in Asthma. J Asthma Allergy 2021; 14:1197-1207. [PMID: 34616159 PMCID: PMC8488044 DOI: 10.2147/jaa.s326058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/01/2021] [Indexed: 12/30/2022] Open
Abstract
Background Asthma belongs to chronic inflammatory respiratory diseases characterized by airway inflammation and remodeling. Circular RNAs (circRNAs) are promising therapeutic targets for various diseases, including asthma. In this work, we aim to investigate the role of circular RNA Erb-B2 receptor tyrosine kinase 2 (circERBB2) during progression of asthma. Methods Human airway smooth muscle cells (ASMCs) were treated with platelet-derived growth factor BB (PDGF-BB) to mimic cell remodeling. The expression of circERBB2, microRNA-98-5p (miR-98-5p), and insulin-like growth factor 1 receptor (IGF1R) was measured by qRT-PCR. Cell proliferation, migration and apoptosis were determined by cell counting-8 (CCK-8), transwell, and flow cytometry. Protein levels of PCNA, MMP-9, IGF1R were evaluated using Western blotting. The levels of tumor necrosis factor‐α (TNF‐α), interleukin‐1β (IL‐1β), and IL‐6 were detected by enzyme‐linked immunosorbent assay (ELISA). Luciferase reporter gene experiment was adopted to evaluate the targeting relationship between miR-98-5p with circERBB2 and IGF1R. Interaction between RNAs was determined by RNA pulldown and RIP assay. Results The depletion of circERBB2 attenuated the proliferation, migration, and levels of inflammatory factors induced by PDGF-BB and cell apoptosis. CircERBB2 was identified to directly interact with miR-98-5p, and overexpression of miR-98-5p abolished the function of circERBB2 on PDGF-BB-stimulated ASMCs. IGF1R was identified as a target of miR-98-5p, and knockdown of IGF1R relieved the PDGF-BB-induced ASMCs proliferation and migration. Conclusion Our work disclosed that knockdown of circERBB2 suppressed PDGF-BB-caused proliferation, migration and inflammatory response of ASMCs, through regulating miR-98-5p/IGF1R signaling, presented circERBB2 as a promising therapeutic target for asthma.
Collapse
Affiliation(s)
- Jun-Qian Huang
- Department of Respiratory and Critical Medicine, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, People's Republic of China
| | - Fang Wang
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, People's Republic of China
| | - Long-Tao Wang
- Department of Critical Care Medicine, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, People's Republic of China
| | - Yong-Mei Li
- Department of Clinical Pharmacy, Qingdao Chengyang District People's Hospital, Qingdao, Shandong, People's Republic of China
| | - Jun-Li Lu
- Qingdao Chengyang District People's Hospital, Qingdao, Shandong, People's Republic of China
| | - Jian-You Chen
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
11
|
Cheng Y, Wang N, Zhao L, Liu C, Wang J, Ma C, Shi X. Knockdown of NOVA1 inhibits inflammation and migration of asthmatic airway smooth muscle cells to regulate PTEN/Akt pathway by targeting PTBP1. Mol Immunol 2021; 138:31-37. [PMID: 34332183 DOI: 10.1016/j.molimm.2021.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/07/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
NOVA1 (neuro-oncological ventral antigen 1) is a neuron specific RNA binding protein, belonging to the Nova family, which plays an important role in various diseases. However, the role of NOVA1 in childhood asthma remains unclear. This study was aimed to investigate the role of NOVA1 in TGF-β1-induced ASMCs proliferation and migration as well as the potential mechanisms. In our study, the NOVA1 expression was significantly increased in asthmatic tissues and TGF-β1-induced ASMCs. Inhibition of NOVA1 significantly inhibited TGF-β1-induced ASMCs cell proliferation and migration, and alleviates TGF-β1-induced inflammation. NOVA1 positively regulated the PTBP1 expression and si-NOVA1 inhibited the activation of PTEN/AKT signal pathway. Importantly, the overexpression of PTBP1 partially reversed the effect of NOVA1 on cell viability, migration, inflammation and the activation of PTEN/AKT signal pathway. Generally, our study demonstrated that si-NOVA1 inhibited TGF-β1-induced inflammation and migration in ASMCs through PTBP1/PTEN/AKT pathway. Therefore, inhibition of NOVA1 may be useful for the prevention or treatment of asthma airway remodeling.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Pediatrics, Weinan Maternal and Child Health Hospital, Weinan City, Shaanxi Province, 714000, China
| | - Ning Wang
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China
| | - Long Zhao
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China
| | - Cuicui Liu
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China
| | - Jing Wang
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China
| | - Cailing Ma
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China
| | - Xiaolan Shi
- Respiratory Asthma Center of Xi'an Children's Hospital, Xi'an City, Shaanxi Province, 710043, China.
| |
Collapse
|
12
|
Banerjee P, Balraj P, Ambhore NS, Wicher SA, Britt RD, Pabelick CM, Prakash YS, Sathish V. Network and co-expression analysis of airway smooth muscle cell transcriptome delineates potential gene signatures in asthma. Sci Rep 2021; 11:14386. [PMID: 34257337 PMCID: PMC8277837 DOI: 10.1038/s41598-021-93845-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Airway smooth muscle (ASM) is known for its role in asthma exacerbations characterized by acute bronchoconstriction and remodeling. The molecular mechanisms underlying multiple gene interactions regulating gene expression in asthma remain elusive. Herein, we explored the regulatory relationship between ASM genes to uncover the putative mechanism underlying asthma in humans. To this end, the gene expression from human ASM was measured with RNA-Seq in non-asthmatic and asthmatic groups. The gene network for the asthmatic and non-asthmatic group was constructed by prioritizing differentially expressed genes (DEGs) (121) and transcription factors (TFs) (116). Furthermore, we identified differentially connected or co-expressed genes in each group. The asthmatic group showed a loss of gene connectivity due to the rewiring of major regulators. Notably, TFs such as ZNF792, SMAD1, and SMAD7 were differentially correlated in the asthmatic ASM. Additionally, the DEGs, TFs, and differentially connected genes over-represented in the pathways involved with herpes simplex virus infection, Hippo and TGF-β signaling, adherens junctions, gap junctions, and ferroptosis. The rewiring of major regulators unveiled in this study likely modulates the expression of gene-targets as an adaptive response to asthma. These multiple gene interactions pointed out novel targets and pathways for asthma exacerbations.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | - Premanand Balraj
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA
| | | | - Sarah A Wicher
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Rodney D Britt
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Christina M Pabelick
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Sudro 108A, Fargo, ND, 58108-6050, USA.
| |
Collapse
|
13
|
Acevedo N, Scala G, Merid SK, Frumento P, Bruhn S, Andersson A, Ogris C, Bottai M, Pershagen G, Koppelman GH, Melén E, Sonnhammer E, Alm J, Söderhäll C, Kere J, Greco D, Scheynius A. DNA Methylation Levels in Mononuclear Leukocytes from the Mother and Her Child Are Associated with IgE Sensitization to Allergens in Early Life. Int J Mol Sci 2021; 22:ijms22020801. [PMID: 33466918 PMCID: PMC7830007 DOI: 10.3390/ijms22020801] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/23/2022] Open
Abstract
DNA methylation changes may predispose becoming IgE-sensitized to allergens. We analyzed whether DNA methylation in peripheral blood mononuclear cells (PBMC) is associated with IgE sensitization at 5 years of age (5Y). DNA methylation was measured in 288 PBMC samples from 74 mother/child pairs from the birth cohort ALADDIN (Assessment of Lifestyle and Allergic Disease During INfancy) using the HumanMethylation450BeadChip (Illumina). PBMCs were obtained from the mothers during pregnancy and from their children in cord blood, at 2 years and 5Y. DNA methylation levels at each time point were compared between children with and without IgE sensitization to allergens at 5Y. For replication, CpG sites associated with IgE sensitization in ALADDIN were evaluated in whole blood DNA of 256 children, 4 years old, from the BAMSE (Swedish abbreviation for Children, Allergy, Milieu, Stockholm, Epidemiology) cohort. We found 34 differentially methylated regions (DMRs) associated with IgE sensitization to airborne allergens and 38 DMRs associated with sensitization to food allergens in children at 5Y (Sidak p ≤ 0.05). Genes associated with airborne sensitization were enriched in the pathway of endocytosis, while genes associated with food sensitization were enriched in focal adhesion, the bacterial invasion of epithelial cells, and leukocyte migration. Furthermore, 25 DMRs in maternal PBMCs were associated with IgE sensitization to airborne allergens in their children at 5Y, which were functionally annotated to the mTOR (mammalian Target of Rapamycin) signaling pathway. This study supports that DNA methylation is associated with IgE sensitization early in life and revealed new candidate genes for atopy. Moreover, our study provides evidence that maternal DNA methylation levels are associated with IgE sensitization in the child supporting early in utero effects on atopy predisposition.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Department of Clinical Science and Education, Karolinska Institutet, and Sachs’ Children and Youth Hospital, Södersjukhuset, SE-118 83 Stockholm, Sweden; (N.A.); (S.K.M.); (E.M.); (J.A.)
- Institute for Immunological Research, University of Cartagena, 130014 Cartagena, Colombia
| | - Giovanni Scala
- Department of Biology, University of Naples Federico II, 80138 Napoli, Italy;
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Institute of Biosciences and Medical Technologies (BioMediTech), Tampere University, 33520 Tampere, Finland
| | - Simon Kebede Merid
- Department of Clinical Science and Education, Karolinska Institutet, and Sachs’ Children and Youth Hospital, Södersjukhuset, SE-118 83 Stockholm, Sweden; (N.A.); (S.K.M.); (E.M.); (J.A.)
| | - Paolo Frumento
- Department of Political Sciences, University of Pisa, 56126 Pisa, Italy;
| | - Sören Bruhn
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (S.B.); (A.A.)
| | - Anna Andersson
- Department of Medicine Solna, Translational Immunology Unit, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (S.B.); (A.A.)
| | - Christoph Ogris
- Stockholm Bioinformatics Center, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, SE-17121 Solna, Sweden; (C.O.); (E.S.)
- Institute of Computational Biology, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | - Matteo Bottai
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (M.B.); (G.P.)
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (M.B.); (G.P.)
| | - Gerard H. Koppelman
- Section of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children’s Hospital, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Groningen Research Institute of Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Erik Melén
- Department of Clinical Science and Education, Karolinska Institutet, and Sachs’ Children and Youth Hospital, Södersjukhuset, SE-118 83 Stockholm, Sweden; (N.A.); (S.K.M.); (E.M.); (J.A.)
- Institute of Environmental Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (M.B.); (G.P.)
| | - Erik Sonnhammer
- Stockholm Bioinformatics Center, Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, SE-17121 Solna, Sweden; (C.O.); (E.S.)
| | - Johan Alm
- Department of Clinical Science and Education, Karolinska Institutet, and Sachs’ Children and Youth Hospital, Södersjukhuset, SE-118 83 Stockholm, Sweden; (N.A.); (S.K.M.); (E.M.); (J.A.)
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (C.S.); (J.K.)
- Department of Women’s and Children’s Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, SE-171 77 Stockholm, Sweden; (C.S.); (J.K.)
- Folkhälsan Research Institute, Stem Cells and Metabolism Research Program, University of Helsinki, 00014 Helsinki, Finland
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Institute of Biosciences and Medical Technologies (BioMediTech), Tampere University, 33520 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, FI-00014 Helsinki, Finland
| | - Annika Scheynius
- Department of Clinical Science and Education, Karolinska Institutet, and Sachs’ Children and Youth Hospital, Södersjukhuset, SE-118 83 Stockholm, Sweden; (N.A.); (S.K.M.); (E.M.); (J.A.)
- Science for Life Laboratory, Karolinska Institutet, SE-171 65 Solna, Sweden
- Correspondence:
| |
Collapse
|
14
|
Zhang YH, Li Z, Zeng T, Chen L, Li H, Huang T, Cai YD. Detecting the Multiomics Signatures of Factor-Specific Inflammatory Effects on Airway Smooth Muscles. Front Genet 2021; 11:599970. [PMID: 33519902 PMCID: PMC7838645 DOI: 10.3389/fgene.2020.599970] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Smooth muscles are a specific muscle subtype that is widely identified in the tissues of internal passageways. This muscle subtype has the capacity for controlled or regulated contraction and relaxation. Airway smooth muscles are a unique type of smooth muscles that constitute the effective, adjustable, and reactive wall that covers most areas of the entire airway from the trachea to lung tissues. Infection with SARS-CoV-2, which caused the world-wide COVID-19 pandemic, involves airway smooth muscles and their surrounding inflammatory environment. Therefore, airway smooth muscles and related inflammatory factors may play an irreplaceable role in the initiation and progression of several severe diseases. Many previous studies have attempted to reveal the potential relationships between interleukins and airway smooth muscle cells only on the omics level, and the continued existence of numerous false-positive optimal genes/transcripts cannot reflect the actual effective biological mechanisms underlying interleukin-based activation effects on airway smooth muscles. Here, on the basis of newly presented machine learning-based computational approaches, we identified specific regulatory factors and a series of rules that contribute to the activation and stimulation of airway smooth muscles by IL-13, IL-17, or the combination of both interleukins on the epigenetic and/or transcriptional levels. The detected discriminative factors (genes) and rules can contribute to the identification of potential regulatory mechanisms linking airway smooth muscle tissues and inflammatory factors and help reveal specific pathological factors for diseases associated with airway smooth muscle inflammation on multiomics levels.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Zhandong Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Tao Zeng
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai, China
| | - Hao Li
- College of Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Tao Huang
- Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
15
|
Abstract
This article will discuss in detail the pathophysiology of asthma from the point of view of lung mechanics. In particular, we will explain how asthma is more than just airflow limitation resulting from airway narrowing but in fact involves multiple consequences of airway narrowing, including ventilation heterogeneity, airway closure, and airway hyperresponsiveness. In addition, the relationship between the airway and surrounding lung parenchyma is thought to be critically important in asthma, especially as related to the response to deep inspiration. Furthermore, dynamic changes in lung mechanics over time may yield important information about asthma stability, as well as potentially provide a window into future disease control. All of these features of mechanical properties of the lung in asthma will be explained by providing evidence from multiple investigative methods, including not only traditional pulmonary function testing but also more sophisticated techniques such as forced oscillation, multiple breath nitrogen washout, and different imaging modalities. Throughout the article, we will link the lung mechanical features of asthma to clinical manifestations of asthma symptoms, severity, and control. © 2020 American Physiological Society. Compr Physiol 10:975-1007, 2020.
Collapse
Affiliation(s)
- David A Kaminsky
- University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - David G Chapman
- University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Zhao L, Shi X, Wang N, Liu C, Wang J. YAP1, targeted by miR-375, enhanced the pro-angiogenesis of airway smooth muscle cells in asthma via STAT3 activation. Cell Cycle 2020; 19:1275-1284. [PMID: 32308108 DOI: 10.1080/15384101.2020.1746874] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
YAP1 was previously reported to regulate the development of multiple tumors, angiogenesis included. Angiogenesis was a specific process of remodeling in asthma. In a recent study, YAP1 was correlated with the progression of asthma. However, the role of YAP1 in airway smooth muscle cell and the asthmatic airway angiogenesis was unclear. In the present study, we used cytokine-stimulated airway smooth muscle cells as asthma cell model in vitro. The results showed a significant up-regulation of YAP1 in asthmatic airway smooth muscle tissue and cytokine-stimulated asthmatic cell model by Western blot. The experimental results of YAP1 loss-of-function combined with STAT3 inhibitor (WP1066) showed that YAP1 knockdown inhibited the expression of VEGF by deactivating STAT3 in cytokine-stimulated ASM cells, which hindered the pro-angiogenesis ability of ASM cells. Besides, by combining prediction and binding site mutation along with luciferase reporter gene experiments, we confirmed direct binding between miR-375 and YAP1. Based on that, the decreased expression level of miR-375 was found to be correlated with the pathogenesis of asthma. Finally, miR-375 was verified to participate in the YAP1-regulated pro-angiogenesis ability of ASM cells. To sum up, we provided the evidence that YAP1 directly binds to miR-375 and takes part in the regulation of the pro-angiogenic ability of ASM cells by activating STAT3 and VEGF signaling.
Collapse
Affiliation(s)
- Long Zhao
- Department of Respiratory and Asthma, Xi'an Children's Hospital , Xi'an, Shanxi, China
| | - Xiaolan Shi
- Department of Respiratory and Asthma, Xi'an Children's Hospital , Xi'an, Shanxi, China
| | - Ning Wang
- Department of Respiratory and Asthma, Xi'an Children's Hospital , Xi'an, Shanxi, China
| | - Cuicui Liu
- Department of Respiratory and Asthma, Xi'an Children's Hospital , Xi'an, Shanxi, China
| | - Jing Wang
- Department of Respiratory and Asthma, Xi'an Children's Hospital , Xi'an, Shanxi, China
| |
Collapse
|
17
|
Anthracopoulos MB, Everard ML. Asthma: A Loss of Post-natal Homeostatic Control of Airways Smooth Muscle With Regression Toward a Pre-natal State. Front Pediatr 2020; 8:95. [PMID: 32373557 PMCID: PMC7176812 DOI: 10.3389/fped.2020.00095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
The defining feature of asthma is loss of normal post-natal homeostatic control of airways smooth muscle (ASM). This is the key feature that distinguishes asthma from all other forms of respiratory disease. Failure to focus on impaired ASM homeostasis largely explains our failure to find a cure and contributes to the widespread excessive morbidity associated with the condition despite the presence of effective therapies. The mechanisms responsible for destabilizing the normal tight control of ASM and hence airways caliber in post-natal life are unknown but it is clear that atopic inflammation is neither necessary nor sufficient. Loss of homeostasis results in excessive ASM contraction which, in those with poor control, is manifest by variations in airflow resistance over short periods of time. During viral exacerbations, the ability to respond to bronchodilators is partially or almost completely lost, resulting in ASM being "locked down" in a contracted state. Corticosteroids appear to restore normal or near normal homeostasis in those with poor control and restore bronchodilator responsiveness during exacerbations. The mechanism of action of corticosteroids is unknown and the assumption that their action is solely due to "anti-inflammatory" effects needs to be challenged. ASM, in evolutionary terms, dates to the earliest land dwelling creatures that required muscle to empty primitive lungs. ASM appears very early in embryonic development and active peristalsis is essential for the formation of the lungs. However, in post-natal life its only role appears to be to maintain airways in a configuration that minimizes resistance to airflow and dead space. In health, significant constriction is actively prevented, presumably through classic negative feedback loops. Disruption of this robust homeostatic control can develop at any age and results in asthma. In order to develop a cure, we need to move from our current focus on immunology and inflammatory pathways to work that will lead to an understanding of the mechanisms that contribute to ASM stability in health and how this is disrupted to cause asthma. This requires a radical change in the focus of most of "asthma research."
Collapse
Affiliation(s)
| | - Mark L. Everard
- Division of Paediatrics & Child Health, Perth Children's Hospital, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
18
|
Dhondalay GKR, Bunning B, Bauer RN, Barnathan ES, Maniscalco C, Baribaud F, Nadeau KC, Andorf S. Transcriptomic and methylomic features in asthmatic and nonasthmatic twins. Allergy 2020; 75:989-992. [PMID: 31758558 DOI: 10.1111/all.14128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Gopal Krishna R. Dhondalay
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | - Bryan Bunning
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | | | | | | | | | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | - Sandra Andorf
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| |
Collapse
|
19
|
Park HW, Weiss ST. Understanding the Molecular Mechanisms of Asthma through Transcriptomics. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:399-411. [PMID: 32141255 PMCID: PMC7061151 DOI: 10.4168/aair.2020.12.3.399] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/01/2020] [Accepted: 01/11/2020] [Indexed: 12/18/2022]
Abstract
The transcriptome represents the complete set of RNA transcripts that are produced by the genome under a specific circumstance or in a specific cell. High-throughput methods, including microarray and bulk RNA sequencing, as well as recent advances in biostatistics based on machine learning approaches provides a quick and effective way of identifying novel genes and pathways related to asthma, which is a heterogeneous disease with diverse pathophysiological mechanisms. In this manuscript, we briefly review how to analyze transcriptome data and then provide a summary of recent transcriptome studies focusing on asthma pathogenesis and asthma drug responses. Studies reviewed here are classified into 2 classes based on the tissues utilized: blood and airway cells.
Collapse
Affiliation(s)
- Heung Woo Park
- The Channing Division of Network Medicine, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Scott T Weiss
- The Channing Division of Network Medicine, Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA.,Partners Center for Personalized Medicine, Partners Health Care, Boston, MA, USA.
| |
Collapse
|
20
|
Bogucka-Kocka A, Zalewski DP, Ruszel KP, Stępniewski A, Gałkowski D, Bogucki J, Komsta Ł, Kołodziej P, Zubilewicz T, Feldo M, Kocki J. Dysregulation of MicroRNA Regulatory Network in Lower Extremities Arterial Disease. Front Genet 2019; 10:1200. [PMID: 31827490 PMCID: PMC6892359 DOI: 10.3389/fgene.2019.01200] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/29/2019] [Indexed: 01/12/2023] Open
Abstract
Atherosclerosis and its comorbidities are the major contributors to the global burden of death worldwide. Lower extremities arterial disease (LEAD) is a common manifestation of atherosclerotic disease of arteries of lower extremities. MicroRNAs belong to epigenetic factors that regulate gene expression and have not yet been extensively studied in LEAD. We aimed to indicate the most promising microRNA and gene expression signatures of LEAD, to identify interactions between microRNA and genes and to describe potential effect of modulated gene expression. High-throughput sequencing was employed to examine microRNAome and transcriptome of peripheral blood mononuclear cells of patients with LEAD, in relation to controls. Statistical significance of microRNAs and genes analysis results was evaluated using DESeq2 and uninformative variable elimination by partial least squares methods. Altered expression of 26 microRNAs (hsa-let-7f-1-3p, hsa-miR-34a-5p, -122-5p, -3591-3p, -34a-3p, -1261, -21-5p, -15a-5p, -548d-5p, -34b-5p, -424-3p, -548aa, -548t-3p, -4423-3p, -196a-5p, -330-3p, -766-3p, -30e-3p, -125b-5p, -1301-3p, -3184-5p, -423-3p, -339-3p, -138-5p, -99a-3p, and -6087) and 14 genes (AK5, CD248, CDS2, FAM129A, FBLN2, GGT1, NOG, NRCAM, PDE7A, RP11-545E17.3, SLC12A2, SLC16A10, SLC4A10, and ZSCAN18) were the most significantly differentially expressed in LEAD group compared to controls. Discriminative value of revealed microRNAs and genes were confirmed by receiver operating characteristic analysis. Dysregulations of 26 microRNAs and 14 genes were used to propose novel biomarkers of LEAD. Regulatory interactions between biomarker microRNAs and genes were studied in silico using R multiMiR package. Functional analysis of genes modulated by proposed biomarker microRNAs was performed using DAVID 6.8 tools and revealed terms closely related to atherosclerosis and, interestingly, the processes involving nervous system. The study provides new insight into microRNA-dependent regulatory mechanisms involved in pathology of LEAD. Proposed microRNA and gene biomarkers of LEAD may provide new diagnostic and therapeutic opportunities.
Collapse
Affiliation(s)
- Anna Bogucka-Kocka
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Daniel P Zalewski
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Karol P Ruszel
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| | - Andrzej Stępniewski
- Ecotech Complex, Analytical and Programme Centre for Advanced Environmentally-Friendly Technologies, University of Marie Curie-Sklodowska, Lublin, Poland
| | - Dariusz Gałkowski
- Department of Pathology and Laboratory Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Jacek Bogucki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| | - Łukasz Komsta
- Chair and Department of Medicinal Chemistry, Medical University of Lublin, Lublin, Poland
| | - Przemysław Kołodziej
- Chair and Department of Biology and Genetics, Medical University of Lublin, Lublin, Poland
| | - Tomasz Zubilewicz
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Marcin Feldo
- Department of Vascular Surgery and Angiology, Medical University of Lublin, Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Chair of Medical Genetics, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
21
|
Peng Y, Zi XX, Tian TF, Lee B, Lum J, Tang SA, Tan KS, Qiu QH, Ye J, Shi L, Guan WJ, Andiappan AK, Wang DY. Whole-transcriptome sequencing reveals heightened inflammation and defective host defence responses in chronic rhinosinusitis with nasal polyps. Eur Respir J 2019; 54:13993003.00732-2019. [PMID: 31439685 DOI: 10.1183/13993003.00732-2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The pathways underlying chronic rhinosinusitis with nasal polyps (CRSwNP) are unclear. We conducted genome-wide gene expression analysis to determine pathways and candidate gene sets associated with CRSwNP. METHODS We performed whole-transcriptome RNA sequencing on 42 polyp (CRSwNP-NP) and 33 paired nonpolyp inferior turbinate (CRSwNP-IT) tissues from patients with CRSwNP and 28 inferior turbinate samples from non-CRS controls (CS-IT). We analysed the differentially expressed genes (DEGs) and the gene sets that were enriched in functional pathways. RESULTS Principal component-informed analysis revealed cilium function and immune regulation as the two main Gene Ontology (GO) categories differentiating CRSwNP patients from controls. We detected 6182 and 1592 DEGs between CRSwNP-NP versus CS-IT and between CRSwNP-NP versus CRSwNP-IT tissues, respectively. Atopy status did not have a major impact on gene expression in various tissues. GO analysis on these DEGs implicated extracellular matrix (ECM) disassembly, O-glycan processing, angiogenesis and host viral response in CRSwNP pathogenesis. Ingenuity Pathway Analysis identified significant enrichment of type 1 interferon signalling and axonal guidance canonical pathways, angiogenesis, and collagen and fibrotic changes in CRSwNP (CRSwNP-NP and CRSwNP-IT) tissues compared with CS-IT. Finally, gene set enrichment analysis implicated sets of genes co-regulated in processes associated with inflammatory response and aberrant cell differentiation in polyp formation. CONCLUSIONS Gene signatures involved in defective host defences (including cilia dysfunction and immune dysregulation), inflammation and abnormal metabolism of ECM are implicated in CRSwNP. Functional validation of these gene expression patterns will open opportunities for CRSwNP therapeutic interventions such as biologics and immunomodulators.
Collapse
Affiliation(s)
- Yang Peng
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Dept of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China.,These authors contributed equally to this work
| | - Xiao-Xue Zi
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Shandong Provincial ENT Hospital Affiliated to Shandong University, Shandong University, Jinan, China.,These authors contributed equally to this work
| | - Teng-Fei Tian
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Dept of Otolaryngology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,These authors contributed equally to this work
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - See Aik Tang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kai Sen Tan
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Qian-Hui Qiu
- Dept of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Dept of Otolaryngology Head and Neck Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jing Ye
- Dept of Otolaryngology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Li Shi
- Shandong Provincial ENT Hospital Affiliated to Shandong University, Shandong University, Jinan, China
| | - Wei-Jie Guan
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - De Yun Wang
- Dept of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
22
|
Diamant Z, Vijverberg S, Alving K, Bakirtas A, Bjermer L, Custovic A, Dahlen S, Gaga M, Gerth van Wijk R, Del Giacco S, Hamelmann E, Heaney LG, Heffler E, Kalayci Ö, Kostikas K, Lutter R, Olin A, Sergejeva S, Simpson A, Sterk PJ, Tufvesson E, Agache I, Seys SF. Toward clinically applicable biomarkers for asthma: An EAACI position paper. Allergy 2019; 74:1835-1851. [PMID: 30953574 DOI: 10.1111/all.13806] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 03/17/2019] [Indexed: 12/13/2022]
Abstract
Inflammation, structural, and functional abnormalities within the airways are key features of asthma. Although these processes are well documented, their expression varies across the heterogeneous spectrum of asthma. Type 2 inflammatory responses are characterized by increased levels of eosinophils, FeNO, and type 2 cytokines in blood and/or airways. Presently, type 2 asthma is the best-defined endotype, typically found in patients with allergic asthma, but surprisingly also in nonallergic patients with (severe) asthma. The etiology of asthma with non-type 2 inflammation is less clear. During the past decade, targeted therapies, including biologicals and small molecules, have been increasingly integrated into treatment strategies of severe asthma. These treatments block specific inflammatory pathways or single mediators. Single or composite biomarkers help to identify patients who will benefit from these treatments. So far, only a few inflammatory biomarkers have been validated for clinical application. The European Academy of Allergy & Clinical Immunology Task Force on Biomarkers in Asthma was initiated to review different biomarker sampling methods and to investigate clinical applicability of new and existing inflammatory biomarkers (point-of-care) to support diagnosis, targeted treatment, and monitoring of severe asthma. Subsequently, we discuss existing and novel targeted therapies for asthma as well as applicable biomarkers.
Collapse
Affiliation(s)
- Zuzana Diamant
- Department of Respiratory Medicine and Allergology Institute for Clinical Science Skane University Hospital Lund Sweden
- Department of Clinical Pharmacy and Pharmacology UMCG and QPS‐NL Groningen The Netherlands
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
| | - Susanne Vijverberg
- Department of Respiratory Medicine Amsterdam UMC University of Amsterdam Amsterdam The Netherlands
| | - Kjell Alving
- Department of Women's and Children's Health Uppsala University Uppsala Sweden
| | - Arzu Bakirtas
- Department of Pediatrics Division of Pediatric Allergy and Asthma Gazi University School of Medicine Ankara Turkey
| | - Leif Bjermer
- Department of Clinical Pharmacy and Pharmacology UMCG and QPS‐NL Groningen The Netherlands
| | - Adnan Custovic
- Section of Paediatrics Department of Medicine Imperial College London London UK
| | - Sven‐Erik Dahlen
- Experimental Asthma and Allergy Research Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | - Mina Gaga
- 7th Respiratory Medicine Department and Asthma Centre Athens Chest Hospital Athens Greece
| | - Roy Gerth van Wijk
- Section of Allergology Department of Internal Medicine Erasmus Medical Center Rotterdam the Netherlands
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - Eckard Hamelmann
- Children's Center Protestant Hospital Bethel Bielefeld Germany
- Allergy Center Ruhr University Bochum Bochum Germany
| | - Liam G. Heaney
- Centre for Experimental Medicine, School of MedicineDentistry and Biomedical Sciences, Queen's University Belfast Belfast UK
| | - Enrico Heffler
- Department of Biomedical Sciences Humanitas University Milan Italy
- Personalized Medicine, Asthma and Allergy Humanitas Research Hospital Milan Italy
| | - Ömer Kalayci
- Division of Pediatric Allergy Faculty of Medicine Hacettepe University Ankara Turkey
| | - Konstantinos Kostikas
- Respiratory Medicine Department University of Ioannina Medical School Ioannina Greece
| | - Rene Lutter
- Department of Respiratory Medicine Amsterdam UMC University of Amsterdam Amsterdam The Netherlands
| | - Anna‐Carin Olin
- Section of Occupational and Environmental Medicine Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | | | - Angela Simpson
- Division of Infection, Immunity and Respiratory Medicine Faculty of Biology, Medicine and Health Manchester Academic Health Sciences Centre University of Manchester and University Hospital of South Manchester NHS Foundation Trust Manchester UK
| | - Peter J. Sterk
- Department of Respiratory Medicine Amsterdam UMC University of Amsterdam Amsterdam The Netherlands
| | - Ellen Tufvesson
- Department of Clinical Pharmacy and Pharmacology UMCG and QPS‐NL Groningen The Netherlands
| | - Ioana Agache
- Department of Allergy and Clinical Immunology Faculty of Medicine Transylvania University Brasov Brasov Romania
| | - Sven F. Seys
- Allergy and Clinical Immunology Research Group Department of Microbiology, Immunology and Transplantation KU Leuven Leuven Belgium
| |
Collapse
|
23
|
Comberiati P, Katial RK, Covar RA. Bronchoprovocation Testing in Asthma: An Update. Immunol Allergy Clin North Am 2019; 38:545-571. [PMID: 30342579 DOI: 10.1016/j.iac.2018.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Bronchial hyperresponsiveness (BHR) is defined as a heightened bronchoconstrictive response to airway stimuli. It complements the cardinal features in asthma, such as variable or reversible airflow limitation and airway inflammation. Although BHR is considered a pathophysiologic hallmark of asthma, it should be acknowledged that this property of the airway is dynamic, because its severity and even presence can vary over time with disease activity, triggers or specific exposure, and with treatment. In addition, it is important to recognize that there is a component that is not reflective of a specific disease entity.
Collapse
Affiliation(s)
- Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, 56126 Pisa, Italy; Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Rohit K Katial
- National Jewish Health, 1400 Jackson Street (J321), Denver, CO 80206, USA
| | - Ronina A Covar
- National Jewish Health, 1400 Jackson Street (J321), Denver, CO 80206, USA.
| |
Collapse
|
24
|
Gelfand EW, Schedel M. Molecular Endotypes Contribute to the Heterogeneity of Asthma. Immunol Allergy Clin North Am 2018; 38:655-665. [PMID: 30342586 DOI: 10.1016/j.iac.2018.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Diagnosis and management of asthma is commonly implemented based on clinical assessment. Although these nonmolecular biomarkers have been useful, limited resolution of the heterogeneity among asthmatic patients and little information regarding the underlying pathobiology of disease in individuals have been provided. Molecular endotying using global transcriptome expression profiling associated with clinical features of asthma has improved our understanding of disease mechanisms, risk assessment of asthma exacerbations, and treatment responses, especially in patients with type 2 inflammation. Further advances in establishing pathobiological subgroups, bioactive pathways, and true disease endotypes hold potential for a more personalized medical approach in asthmatic patients.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA.
| |
Collapse
|
25
|
McGeachie MJ, Clemmer GL, Hayete B, Xing H, Runge K, Wu AC, Jiang X, Lu Q, Church B, Khalil I, Tantisira K, Weiss S. Systems biology and in vitro validation identifies family with sequence similarity 129 member A (FAM129A) as an asthma steroid response modulator. J Allergy Clin Immunol 2018; 142:1479-1488.e12. [PMID: 29410046 DOI: 10.1016/j.jaci.2017.11.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Variation in response to the most commonly used class of asthma controller medication, inhaled corticosteroids, presents a serious challenge in asthma management, particularly for steroid-resistant patients with little or no response to treatment. OBJECTIVE We applied a systems biology approach to primary clinical and genomic data to identify and validate genes that modulate steroid response in asthmatic children. METHODS We selected 104 inhaled corticosteroid-treated asthmatic non-Hispanic white children and determined a steroid responsiveness endophenotype (SRE) using observations of 6 clinical measures over 4 years. We modeled each subject's cellular steroid response using data from a previously published study of immortalized lymphoblastoid cell lines under dexamethasone (DEX) and sham treatment. We integrated SRE with immortalized lymphoblastoid cell line DEX responses and genotypes to build a genome-scale network using the Reverse Engineering, Forward Simulation modeling framework, identifying 7 genes modulating SRE. RESULTS Three of these genes were functionally validated by using a stable nuclear factor κ-light-chain-enhancer of activated B cells luciferase reporter in A549 human lung epithelial cells, IL-1β cytokine stimulation, and DEX treatment. By using small interfering RNA transfection, knockdown of family with sequence similarity 129 member A (FAM129A) produced a reduction in steroid treatment response (P < .001). CONCLUSION With this systems-based approach, we have shown that FAM129A is associated with variation in clinical asthma steroid responsiveness and that FAM129A modulates steroid responsiveness in lung epithelial cells.
Collapse
Affiliation(s)
- Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - George L Clemmer
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass
| | | | - Heming Xing
- Novartis Institute for Biomedical Research, Cambridge, Mass
| | | | - Ann Chen Wu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass; Precision Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Mass
| | - Xiaofeng Jiang
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | | | | | - Kelan Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Scott Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| |
Collapse
|
26
|
Cook DP, Adam RJ, Zarei K, Deonovic B, Stroik MR, Gansemer ND, Meyerholz DK, Au KF, Stoltz DA. CF airway smooth muscle transcriptome reveals a role for PYK2. JCI Insight 2017; 2:95332. [PMID: 28878137 DOI: 10.1172/jci.insight.95332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/27/2017] [Indexed: 12/17/2022] Open
Abstract
Abnormal airway smooth muscle function can contribute to cystic fibrosis (CF) airway disease. We previously found that airway smooth muscle from newborn CF pigs had increased basal tone, an increased bronchodilator response, and abnormal calcium handling. Since CF pigs lack airway infection and inflammation at birth, these findings suggest intrinsic airway smooth muscle dysfunction in CF. In this study, we tested the hypothesis that CFTR loss in airway smooth muscle would produce a distinct set of changes in the airway smooth muscle transcriptome that we could use to develop novel therapeutic targets. Total RNA sequencing of newborn wild-type and CF airway smooth muscle revealed changes in muscle contraction-related genes, ontologies, and pathways. Using connectivity mapping, we identified several small molecules that elicit transcriptional signatures opposite of CF airway smooth muscle, including NVP-TAE684, an inhibitor of proline-rich tyrosine kinase 2 (PYK2). In CF airway smooth muscle tissue, PYK2 phosphorylation was increased and PYK2 inhibition decreased smooth muscle contraction. In vivo NVP-TAE684 treatment of wild-type mice reduced methacholine-induced airway smooth muscle contraction. These findings suggest that studies in the newborn CF pig may provide an important approach to enhance our understanding of airway smooth muscle biology and for discovery of novel airway smooth muscle therapeutics for CF and other diseases of airway hyperreactivity.
Collapse
Affiliation(s)
- Daniel P Cook
- Department of Internal Medicine.,Department of Molecular Physiology and Biophysics, and
| | - Ryan J Adam
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Keyan Zarei
- Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Benjamin Deonovic
- Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | | | | | - David K Meyerholz
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Kin Fai Au
- Department of Internal Medicine.,Department of Biostatistics, College of Public Health, University of Iowa, Iowa City, Iowa, USA
| | - David A Stoltz
- Department of Internal Medicine.,Department of Molecular Physiology and Biophysics, and.,Department of Biomedical Engineering, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
27
|
d'Hooghe JNS, Ten Hacken NHT, Weersink EJM, Sterk PJ, Annema JT, Bonta PI. Emerging understanding of the mechanism of action of Bronchial Thermoplasty in asthma. Pharmacol Ther 2017; 181:101-107. [PMID: 28757156 DOI: 10.1016/j.pharmthera.2017.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bronchial Thermoplasty (BT) is an endoscopic treatment for moderate-to-severe asthma patients who are uncontrolled despite optimal medical therapy. Effectiveness of BT has been demonstrated in several randomized clinical trials. However, the asthma phenotype that benefits most of this treatment is unclear, partly because the mechanism of action is incompletely understood. BT was designed to reduce the amount of airway smooth muscle (ASM), but additional direct and indirect effects on airway pathophysiology are expected. This review will provide an overview of the different components of airway pathophysiology including remodeling, with the ASM as the key player. Current concepts in the understanding of BT clinical effectiveness with a focus on its impact on airway remodeling will be reviewed.
Collapse
Affiliation(s)
- J N S d'Hooghe
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - N H T Ten Hacken
- Department of Respiratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - E J M Weersink
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - P J Sterk
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - J T Annema
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - P I Bonta
- Department of Respiratory Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Thamrin C, Frey U, Kaminsky DA, Reddel HK, Seely AJE, Suki B, Sterk PJ. Systems Biology and Clinical Practice in Respiratory Medicine. The Twain Shall Meet. Am J Respir Crit Care Med 2017; 194:1053-1061. [PMID: 27556336 DOI: 10.1164/rccm.201511-2288pp] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Respiratory diseases are highly complex, being driven by host-environment interactions and manifested by inflammatory, structural, and functional abnormalities that vary over time. Traditional reductionist approaches have contributed vastly to our knowledge of biological systems in health and disease to date; however, they are insufficient to provide an understanding of the behavior of the system as a whole. In this Pulmonary Perspective, we discuss systems biology approaches, especially but not limited to the study of the lung as a complex system. Such integrative approaches take into account the large number of dynamic subunits and their interactions found in biological systems. Borrowing methods from physics and mathematics, it is possible to study the collective behavior of these systems over time and in a multidimensional manner. We first examine the physiological basis for complexity in the respiratory system and its implications for disease. We then expand on the potential applications of systems biology methods to study complex systems, within the context of diagnosis and monitoring of respiratory diseases including asthma, chronic obstructive pulmonary disease (COPD), and critical illness. We summarize the significant advances made in recent years using systems approaches for disease phenotyping, applied to data ranging from the molecular to clinical level, obtained from large-scale asthma and COPD networks. We describe new studies using temporal complexity patterns to characterize asthma and COPD and predict exacerbations as well as predict adverse outcomes in critical care. We highlight new methods that are emerging with this approach and discuss remaining questions that merit greater attention in the field.
Collapse
Affiliation(s)
- Cindy Thamrin
- 1 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Urs Frey
- 2 University Children's Hospital Basel, Basel, Switzerland
| | - David A Kaminsky
- 3 University of Vermont College of Medicine, Burlington, Vermont
| | - Helen K Reddel
- 1 Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J E Seely
- 4 Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Béla Suki
- 5 Department of Biomedical Engineering, Boston University, Boston, Massachusetts; and
| | - Peter J Sterk
- 6 Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Lv J, Sun B, Mai Z, Jiang M, Du J. STAT3 potentiates the ability of airway smooth muscle cells to promote angiogenesis by regulating VEGF signalling. Exp Physiol 2017; 102:598-606. [PMID: 28295786 DOI: 10.1113/ep086136] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/03/2017] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Airway angiogenesis occurs in asthma, and airway smooth muscle (ASM) cells have been reported to be capable of promoting airway angiogenesis. What is the potential mechanism by which ASM cells harvested from patients with asthma are capable of promoting airway angiogenesis? What is the main finding and its importance? Endogenous STAT3 mediated the pro-angiogenic ability of ASM cells by directly activating VEGF signalling. These findings contribute to the understanding of airway angiogenesis in pathology and could represent a possible therapeutic target for asthma. Airway angiogenesis indicates the specific vascular structure remodelling that occurs in asthma. Airway smooth muscle (ASM) cells have been reported to be capable of promoting airway angiogenesis; however, the potential mechanism is not yet fully defined. Herein, we investigated the role of signal transducer and activator of transcription 3 (STAT3) in the progress of airway angiogenesis. Western blot analysis showed that STAT3 activation was aberrantly upregulated in ASM tissues of patients with asthma and ASM cells that were exposed to cytokines to imitate the airway conditions in patients with asthma. Compared with the control group, both the inhibition of STAT3 activation and the silencing of endogenous STAT3 in ASM cells significantly reduced the proliferation, migration and tube-forming ability of human lung microvascular endothelial cells induced by the conditioned medium (CM) of ASM cells. The increased proliferation and migration of human aortic vascular smooth muscle cells were also repressed by inhibition of STAT3 in ASM cells. Besides, the increased activity of VEGF signalling was observed in ASM cells and the CM by RT-PCR and Western blotting assay, whereas this increased activity was reduced by STAT3 silencing. Further studies indicated that STAT3 regulated VEGF activation by directly interacting with the binding site on the 5' region of the VEGF gene. The increase in STAT3-induced pro-angiogenic activity of ASM cells was significantly decreased by administration of VEGF neutralizing antibody. In conclusion, we provided evidence that endogenous STAT3 mediates the pro-angiogenic ability of ASM cells by directly activating VEGF signalling, which could represent a possible therapeutic target for asthma.
Collapse
Affiliation(s)
- Jing Lv
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Baohua Sun
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Zhitao Mai
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Mingming Jiang
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Junfeng Du
- Department of Respiration Medicine, Cangzhou Central Hospital, Cangzhou, 061001, China
| |
Collapse
|
30
|
Chapman DG, Irvin CG. Mechanisms of airway hyper-responsiveness in asthma: the past, present and yet to come. Clin Exp Allergy 2015; 45:706-19. [PMID: 25651937 DOI: 10.1111/cea.12506] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Airway hyper-responsiveness (AHR) has long been considered a cardinal feature of asthma. The development of the measurement of AHR 40 years ago initiated many important contributions to our understanding of asthma and other airway diseases. However, our understanding of AHR in asthma remains complicated by the multitude of potential underlying mechanisms which in reality are likely to have different contributions amongst individual patients. Therefore, the present review will discuss the current state of understanding of the major mechanisms proposed to contribute to AHR and highlight the way in which AHR testing is beginning to highlight distinct abnormalities associated with clinically relevant patient populations. In doing so we aim to provide a foundation by which future research can begin to ascribe certain mechanisms to specific patterns of bronchoconstriction and subsequently match phenotypes of bronchoconstriction with clinical phenotypes. We believe that this approach is not only within our grasp but will lead to improved mechanistic understanding of asthma phenotypes and we hoped to better inform the development of phenotype-targeted therapy.
Collapse
Affiliation(s)
- D G Chapman
- Woolcock Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, NSW, Australia; Department of Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | | |
Collapse
|
31
|
Li X, Ben-Dov IZ, Mauro M, Williams Z. Lowering the quantification limit of the QubitTM RNA HS assay using RNA spike-in. BMC Mol Biol 2015; 16:9. [PMID: 25943882 PMCID: PMC4431604 DOI: 10.1186/s12867-015-0039-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/27/2015] [Indexed: 12/15/2022] Open
Abstract
Background RNA quantification is often a prerequisite for most RNA analyses such as RNA sequencing. However, the relatively low sensitivity and large sample consumption of traditional RNA quantification methods such as UV spectrophotometry and even the much more sensitive fluorescence-based RNA quantification assays, such as the Qubit™ RNA HS Assay, are often inadequate for measuring minute levels of RNA isolated from limited cell and tissue samples and biofluids. Thus, there is a pressing need for a more sensitive method to reliably and robustly detect trace levels of RNA without interference from DNA. Methods To improve the quantification limit of the Qubit™ RNA HS Assay, we spiked-in a known quantity of RNA to achieve the minimum reading required by the assay. Samples containing trace amounts of RNA were then added to the spike-in and measured as a reading increase over RNA spike-in baseline. We determined the accuracy and precision of reading increases between 1 and 20 pg/μL as well as RNA-specificity in this range, and compared to those of RiboGreen®, another sensitive fluorescence-based RNA quantification assay. We then applied Qubit™ Assay with RNA spike-in to quantify plasma RNA samples. Results RNA spike-in improved the quantification limit of the Qubit™ RNA HS Assay 5-fold, from 25 pg/μL down to 5 pg/μL while maintaining high specificity to RNA. This enabled quantification of RNA with original concentration as low as 55.6 pg/μL compared to 250 pg/μL for the standard assay and decreased sample consumption from 5 to 1 ng. Plasma RNA samples that were not measurable by the Qubit™ RNA HS Assay were measurable by our modified method. Conclusions The Qubit™ RNA HS Assay with RNA spike-in is able to quantify RNA with high specificity at 5-fold lower concentration and uses 5-fold less sample quantity than the standard Qubit™ Assay. Electronic supplementary material The online version of this article (doi:10.1186/s12867-015-0039-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xin Li
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, 10461, Bronx, NY, USA. .,Department of Genetics, Albert Einstein College of Medicine, 10461, Bronx, NY, USA.
| | - Iddo Z Ben-Dov
- Nephrology and Hypertension, Hadassah - Hebrew University Medical Center, 91120, Jerusalem, Israel.
| | - Maurizio Mauro
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, 10461, Bronx, NY, USA. .,Department of Genetics, Albert Einstein College of Medicine, 10461, Bronx, NY, USA.
| | - Zev Williams
- Department of Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, 10461, Bronx, NY, USA. .,Department of Genetics, Albert Einstein College of Medicine, 10461, Bronx, NY, USA.
| |
Collapse
|
32
|
Pascoe CD, Swyngedouw NE, Seow CY, Paré PD. Gene expression in asthmatic airway smooth muscle: a mixed bag. Can J Physiol Pharmacol 2014; 93:137-43. [PMID: 25587873 DOI: 10.1139/cjpp-2014-0390] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It has long been known that airway smooth muscle (ASM) contraction contributes significantly to the reversible airflow obstruction that defines asthma. It has also been postulated that phenotypic changes in ASM contribute to the airway hyper-responsiveness (AHR) that is a characteristic feature of asthma. Although there is agreement that the mass of ASM surrounding the airways is significantly increased in asthmatic compared with non-asthmatic airways, it is still uncertain whether there are quantitative or qualitative changes in the level of expression of the genes and proteins involved in the canonical contractile pathway in ASM that could account for AHR. This review will summarize past attempts at quantifying gene expression changes in the ASM of asthmatic lungs as well as non-asthmatic ASM cells stimulated with various inflammatory cytokines. The lack of consistent findings in asthmatic samples coupled with the relative concordance of results from stimulated ASM cells suggests that changes to the contractility of ASM tissues in asthma may be dependent on the presence of an inflammatory environment surrounding the ASM layer. Removal of the ASM from this environment could explain why hypercontractility is rarely seen ex vivo.
Collapse
Affiliation(s)
- Christopher D Pascoe
- a Department of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | | | | | | |
Collapse
|
33
|
Abstract
Genome-wide association studies (GWAS) have been employed in the field of allergic disease, and significant associations have been published for nearly 100 asthma genes/loci. An outcome of GWAS in allergic disease has been the formation of national and international collaborations leading to consortia meta-analyses, and an appreciation for the specificity of genetic associations to sub-phenotypes of allergic disease. Molecular genetics has undergone a technological revolution, leading to next-generation sequencing strategies that are increasingly employed to hone in on the causal variants associated with allergic diseases. Unmet needs include the inclusion of diverse cohorts and strategies for managing big data.
Collapse
Affiliation(s)
- Romina A Ortiz
- Department of Medicine, The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3A.62, Baltimore, MD 21224, USA
| | - Kathleen C Barnes
- Department of Medicine, The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3A.62, Baltimore, MD 21224, USA.
| |
Collapse
|