1
|
Walker MT, Bloodworth JC, Kountz TS, McCarty SL, Green JE, Ferrie RP, Campbell JA, Averill SH, Beckman KB, Grammer LC, Eng C, Avila PC, Farber HJ, Rodriguez-Cintron W, Rodriguez-Santana JR, Serebrisky D, Thyne SM, Seibold MA, Burchard EG, Kumar R, Cook-Mills JM. 5-HTP inhibits eosinophilia via intracellular endothelial 5-HTRs; SNPs in 5-HTRs associate with asthmatic lung function. FRONTIERS IN ALLERGY 2024; 5:1385168. [PMID: 38845678 PMCID: PMC11153829 DOI: 10.3389/falgy.2024.1385168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Background Previous research showed that 5-hydroxytryptophan (5HTP), a metabolic precursor of serotonin, reduces allergic lung inflammation by inhibiting eosinophil migration across endothelial monolayers. Objective It is unknown if serotonin receptors are involved in mediating this 5HTP function or if serotonin receptor (HTR) single nucleotide polymorphisms (SNPs) associate with lung function in humans. Methods Serotonin receptor subtypes were assessed by qPCR, western blot, confocal microscopy, pharmacological inhibitors and siRNA knockdown. HTR SNPs were assessed in two cohorts. Results Pharmacological inhibition or siRNA knockdown of the serotonin receptors HTR1A or HTR1B in endothelial cells abrogated the inhibitory effects of 5HTP on eosinophil transendothelial migration. In contrast, eosinophil transendothelial migration was not inhibited by siRNA knockdown of HTR1A or HTR1B in eosinophils. Surprisingly, these HTRs were intracellular in endothelial cells and an extracellular supplementation with serotonin did not inhibit eosinophil transendothelial migration. This is consistent with the inability of serotonin to cross membranes, the lack of selective serotonin reuptake receptors on endothelial cells, and the studies showing minimal impact of selective serotonin reuptake inhibitors on asthma. To extend our HTR studies to humans with asthma, we examined the CHIRAH and GALA cohorts for HTR SNPs that affect HTR function or are associated with behavior disorders. A polygenic index of SNPs in HTRs was associated with lower lung function in asthmatics. Conclusions Serotonin receptors mediate 5HTP inhibition of transendothelial migration and HTR SNPs associate with lower lung function. These results may serve to aid in design of novel interventions for allergic inflammation.
Collapse
Affiliation(s)
- Matthew T. Walker
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jeffrey C. Bloodworth
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Timothy S. Kountz
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha L. McCarty
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jeremy E. Green
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Ryan P. Ferrie
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jackson A. Campbell
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha H. Averill
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Leslie C. Grammer
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Celeste Eng
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Pedro C. Avila
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Harold J. Farber
- Department of Pediatrics, Section of Pulmonology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, United States
| | | | | | - Denise Serebrisky
- Pediatric Pulmonary Division, Jacobi Medical Center, Bronx, NY, United States
| | - Shannon M. Thyne
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Max A. Seibold
- Center for Genes, Environment, and Health and the Department of Pediatrics, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, CO, United States
| | - Esteban G. Burchard
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Rajesh Kumar
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Division of Allergy and Clinical Immunology, Ann and Robert H Lurie Children’s Hospital of Chicago, Chicago, IL, United States
| | - Joan M. Cook-Mills
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
2
|
Nowakowska J, Olechnowicz A, Langwiński W, Koteluk O, Lemańska Ż, Jóźwiak K, Kamiński K, Łosiewski W, Stegmayr J, Wagner D, Alsafadi HN, Lindstedt S, Dziuba M, Bielicka A, Graczyk Z, Szczepankiewicz A. Increased expression of ORMDL3 in allergic asthma: a case control and in vitro study. J Asthma 2023; 60:458-467. [PMID: 35321632 DOI: 10.1080/02770903.2022.2056896] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Asthma is the most frequent chronic disease in children. One of the most replicated genetic findings in childhood asthma is the ORMDL3 gene confirmed in several GWA studies in several pediatric populations. OBJECTIVES The purpose of this study was to analyze ORMDL3 variants and expression in childhood asthma in the Polish population. METHODS In the study we included 416 subject, 223 asthmatic children and 193 healthy control subjects. The analysis of two SNPs (rs3744246 and rs8076131) was performed using genotyping with TaqMan probes. The methylation of the ORMDL3 promoter was examined with Methylation Sensitive HRM (MS-HRM), covering 9 CpG sites. The expression of ORMDL3 was analyzed in PBMCs from pediatric patients diagnosed with allergic asthma and primary human bronchial epithelial cells derived from healthy subjects treated with IL-13, IL-4, or co-treatment with both cytokines to model allergic airway inflammation. RESULTS We found that ORMDL3 expression was increased in allergic asthma both in PBMCs from asthmatic patients as well as in human bronchial epithelial cells stimulated with the current cytokines. We did not observe significant differences between cases and controls either in the genotype distribution of analyzed SNPs (rs3744246 and rs8076131) nor in the level of promoter methylation. CONCLUSIONS Increased ORMDL3 expression is associated with pediatric allergic asthma and upregulated in the airways upon Th2-cytokines stimulation, but further functional studies are required to fully understand its role in this disease.
Collapse
Affiliation(s)
- Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Olechnowicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Oliwia Koteluk
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Żaneta Lemańska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kacper Jóźwiak
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kacper Kamiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Łosiewski
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - John Stegmayr
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Darcy Wagner
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hani N Alsafadi
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Maria Dziuba
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Antonina Bielicka
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Zuzanna Graczyk
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
3
|
Ma S, Yang K, Li Z, Li L, Feng Y, Wang X, Wang J, Zhu Z, Wang Z, Wang J, Zhu Y, Liu L. A retro-inverso modified peptide alleviated ovalbumin-induced asthma model by affecting glycerophospholipid and purine metabolism of immune cells. Pulm Pharmacol Ther 2023; 78:102185. [PMID: 36563740 DOI: 10.1016/j.pupt.2022.102185] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Allergic asthma is a heterogeneous disease involving a variety of inflammatory cells. Immune imbalance or changes in the immune microenvironment are the essential causes that promote inflammation in allergic asthma. Tetraspanin CD81 can be used as a platform for receptor clustering and signal transmission owing to its special transmembrane structure and is known to participate in the physiological processes of cell proliferation, differentiation, adhesion, and migration. Previous studies have shown that CD81-targeting peptidomimetics exhibit anti-allergic lung inflammation. However, due to the low metabolic stability of peptide drugs, their druggability is limited. Here, we aimed to generate a metabolically stable anti-CD81 peptide, evaluate its anti-inflammatory action and establish its mechanism of action. Based on previous reports, we applied retro-inverse peptide modification to obtain a new compound, PD00 (NH2-D-Gly-D-Ser-D-Thr-D-Tyr-D-Thr-D-Gln-D-Gly-COOH), with high metabolic stability. Enhanced ultraperformance liquid chromatography-tandem mass spectrometry was used to investigate the in vitro and in vivo metabolic stabilities of PD00. The affinities of PD00 and CD81 were studied using molecular docking and surface plasmon resonance techniques. An ovalbumin (OVA)-induced asthma model was used to evaluate the effects of PD00 in vivo. Mice were treated with different concentrations of PD00 (175 and 350 μg/kg) for 10 days. Airway hyperresponsiveness (AHR) to acetyl-β-methacholine (Mch), inflammatory cell counts in the bronchoalveolar lavage fluid, and serum OVA-specific IgE levels were detected in the mice at the end of the experiment. Lung tissues were collected for haematoxylin and eosin staining, untargeted metabolomic analysis, and single-cell transcriptome sequencing. PD00 has a high affinity for CD81; therefore, administration of PD00 markedly ameliorated AHR and airway inflammation in mice after OVA sensitisation and exposure. Serum OVA-specific IgE levels decreased considerably. In addition, PD00 treatment increased glycerophospholipid and purine metabolism in immune cells. Collectively, PD00 may regulate the glycerophospholipid and purine metabolism pathways to ameliorate the pathophysiological features of asthma. These findings suggest that PD00 is a potential compound for the treatment of asthma.
Collapse
Affiliation(s)
- Shumei Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China; Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Kuan Yang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
| | - Zhihong Li
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Liang Li
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Yue Feng
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Xiaowei Wang
- Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Jiahui Wang
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Zhengdan Zhu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China; Beijing Institute of Big Data Research, Beijing, PR China
| | - Zhiyong Wang
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Juan Wang
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China
| | - Yizhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China.
| | - Li Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China; Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, PR China; Shanghai Professional and Technical Service Center for Biological Material Drug-Ability Evaluation, Shanghai, PR China.
| |
Collapse
|
4
|
Chiu KL, Chang WS, Tsai CW, Mong MC, Hsia TC, Bau DT. Novel genetic variants in long non-coding RNA MEG3 are associated with the risk of asthma. PeerJ 2023; 11:e14760. [PMID: 36726728 PMCID: PMC9885862 DOI: 10.7717/peerj.14760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/27/2022] [Indexed: 01/28/2023] Open
Abstract
Background Asthma is the most common chronic inflammatory airway disease worldwide. Asthma is a complex disease whose exact etiologic mechanisms remain elusive; however, it is increasingly evident that genetic factors play essential roles in the development of asthma. The purpose of this study is to identify novel genetic susceptibility loci for asthma in Taiwanese. We selected a well-studied long non-coding RNA (lncRNA), MEG3, which is involved in multiple cellular functions and whose expression has been associated with asthma. We hypothesize that genetic variants in MEG3 may influence the risk of asthma. Methods We genotyped four single nucleotide polymorphisms (SNPs) in MEG3, rs7158663, rs3087918, rs11160608, and rs4081134, in 198 patients with asthma and 453 healthy controls and measured serum MEG3 expression level in a subset of controls. Results The variant AG and AA genotypes of MEG3 rs7158663 were significantly over-represented in the patients compared to the controls (P = 0.0024). In logistic regression analyses, compared with the wild-type GG genotype, the heterozygous variant genotype (AG) was associated with a 1.62-fold [95% confidence interval (CI) [1.18-2.32], P = 0.0093] increased risk and the homozygous variant genotype (AA) conferred a 2.68-fold (95% CI [1.52-4.83], P = 0.003) increased risk of asthma. The allelic test showed the A allele was associated with a 1.63-fold increased risk of asthma (95% CI [1.25-2.07], P = 0.0004). The AG plus AA genotypes were also associated with severe symptoms (P = 0.0148). Furthermore, the AG and AA genotype carriers had lower serum MEG3 expression level than the GG genotype carriers, consistent with the reported downregulation of MEG3 in asthma patients. Conclusion MEG3 SNP rs7158663 is a genetic susceptibility locus for asthma in Taiwanese. Individuals carrying the variant genotypes have lower serum MEG3 level and are at increased risks of asthma and severe symptoms.
Collapse
Affiliation(s)
- Kuo-Liang Chiu
- Division of Chest Medicine, Department of Internal Medicine, Taichung Tzu Chi Hospital, Taichung, Taiwan,School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Shin Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Wen Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Mei-Chin Mong
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Te-Chun Hsia
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Da-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
5
|
Breton CV, Landon R, Kahn LG, Enlow MB, Peterson AK, Bastain T, Braun J, Comstock SS, Duarte CS, Hipwell A, Ji H, LaSalle JM, Miller RL, Musci R, Posner J, Schmidt R, Suglia SF, Tung I, Weisenberger D, Zhu Y, Fry R. Exploring the evidence for epigenetic regulation of environmental influences on child health across generations. Commun Biol 2021; 4:769. [PMID: 34158610 PMCID: PMC8219763 DOI: 10.1038/s42003-021-02316-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/03/2021] [Indexed: 02/08/2023] Open
Abstract
Environmental exposures, psychosocial stressors and nutrition are all potentially important influences that may impact health outcomes directly or via interactions with the genome or epigenome over generations. While there have been clear successes in large-scale human genetic studies in recent decades, there is still a substantial amount of missing heritability to be elucidated for complex childhood disorders. Mounting evidence, primarily in animals, suggests environmental exposures may generate or perpetuate altered health outcomes across one or more generations. One putative mechanism for these environmental health effects is via altered epigenetic regulation. This review highlights the current epidemiologic literature and supporting animal studies that describe intergenerational and transgenerational health effects of environmental exposures. Both maternal and paternal exposures and transmission patterns are considered, with attention paid to the attendant ethical, legal and social implications.
Collapse
Affiliation(s)
- Carrie V Breton
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Remy Landon
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Linda G Kahn
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alicia K Peterson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Theresa Bastain
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joseph Braun
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Cristiane S Duarte
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Alison Hipwell
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Ji
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, California National Primate Research Center, University of California, Davis, Davis, CA, USA
| | - Janine M LaSalle
- Department of Medical Microbiology and Immunology, MIND Institute, Genome Center, University of California, Davis, Davis, CA, USA
| | | | - Rashelle Musci
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jonathan Posner
- Department of Psychiatry, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center and New York State Psychiatric Institute, New York, NY, USA
| | - Rebecca Schmidt
- Department of Public Health Sciences, UC Davis School of Medicine, Davis, CA, USA
| | | | - Irene Tung
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Weisenberger
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California and Department of Epidemiology and Biostatistics, University of California, San Francisco, Oakland, CA, USA
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
6
|
Huang J, Hu X, Zheng X, Kuang J, Liu C, Wang X, Tang Y. Effects of STIP1 and GLCCI1 polymorphisms on the risk of childhood asthma and inhaled corticosteroid response in Chinese asthmatic children. BMC Pulm Med 2020; 20:303. [PMID: 33208131 PMCID: PMC7677774 DOI: 10.1186/s12890-020-01332-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Background Asthma is a common chronic lung disease in children. We aimed to determine the associations between stress-induced phosphoprotein 1 (STIP1) and glucocorticoid-induced transcript 1 (GLCCI1) polymorphisms and susceptibility of childhood asthma and inhaled corticosteroid (ICS) response in children. Methods A total of 263 Chinese Han asthmatic children were recruited from the Xiangya Hospital, Central South University. Pulmonary function tests were performed before the treatment and 3 months after the treatment. One hundred fifty non-asthmatic children were recruited. Each participant’s DNA was extracted from the peripheral blood and Method of MassARRAY was used to genotype the single-nucleotide polymorphisms (SNPs). Results STIP1 rs2236647 wild-type homozygote (CC) was associated with increased asthma risk of children (OR = 1.858, 95% CI:1.205–2.864), but not associated with the ICS response. GLCCI1 rs37969, rs37972 and rs37973 polymorphisms were not associated with the risk of childhood asthma. However, rs37969 mutant genotypes (TT/GT) were significantly associated with less improvement in PD20 (p = 0.028). We also found significant associations between rs37969, rs37972 and rs37973 mutant genotypes and less improvement in maximal midexpiratory flow (MMEF) after ICS treatment for 3 months (p = 0.036, p = 0.010 and p = 0.003, respectively). Conclusions STIP1 rs2236647 was associated with asthma risk of children and GLCCI1 rs37969 mutant genotypes were associated with less improvement in airway hyper-responsiveness. GLCCI1 rs37969, rs37972 and rs37973 polymorphisms might be associated with pulmonary function in childhood asthma patients after ICS treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-020-01332-2.
Collapse
Affiliation(s)
- Juan Huang
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pediatric, The First Hospital of Changsha, Changsha, Hunan, China
| | - Xiaolei Hu
- National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangrong Zheng
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jian Kuang
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chentao Liu
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xia Wang
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongjun Tang
- Department of Pediatric, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
The Genomics and Metagenomics of Asthma Severity (GEMAS) Study: Rationale and Design. J Pers Med 2020; 10:jpm10030123. [PMID: 32933076 PMCID: PMC7563269 DOI: 10.3390/jpm10030123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/04/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Asthma exacerbations are a major contributor to the global disease burden, but no significant predictive biomarkers are known. The Genomics and Metagenomics of Asthma Severity (GEMAS) study aims to assess the role of genomics and the microbiome in severe asthma exacerbations. Here, we present the design of GEMAS and the characteristics of patients recruited from March 2018 to March 2020. Different biological samples and demographic and clinical variables were collected from asthma patients recruited by allergy and pulmonary medicine units in several hospitals from Spain. Cases and controls were defined by the presence/absence of severe asthma exacerbations in the past year (oral corticosteroid use, emergency room visits, and/or asthma-related hospitalizations). A total of 137 cases and 120 controls were recruited. After stratifying by recruitment location (i.e., Canary Islands and Basque Country), cases and controls did not differ for most demographic and clinical variables (p > 0.05). However, cases showed a higher proportion of characteristics inherent to asthma exacerbations (impaired lung function, severe disease, uncontrolled asthma, gastroesophageal reflux, and use of asthma medications) compared to controls (p < 0.05). Similar results were found after stratification by recruitment unit. Thereby, asthma patients enrolled in GEMAS are balanced for potential confounders and have clinical characteristics that support the phenotype definition. GEMAS will improve the knowledge of potential biomarkers of asthma exacerbations.
Collapse
|
8
|
Ma X, Wang P, Xu G, Yu F, Ma Y. Integrative genomics analysis of various omics data and networks identify risk genes and variants vulnerable to childhood-onset asthma. BMC Med Genomics 2020; 13:123. [PMID: 32867763 PMCID: PMC7457797 DOI: 10.1186/s12920-020-00768-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Childhood-onset asthma is highly affected by genetic components. In recent years, many genome-wide association studies (GWAS) have reported a large group of genetic variants and susceptible genes associated with asthma-related phenotypes including childhood-onset asthma. However, the regulatory mechanisms of these genetic variants for childhood-onset asthma susceptibility remain largely unknown. METHODS In the current investigation, we conducted a two-stage designed Sherlock-based integrative genomics analysis to explore the cis- and/or trans-regulatory effects of genome-wide SNPs on gene expression as well as childhood-onset asthma risk through incorporating a large-scale GWAS data (N = 314,633) and two independent expression quantitative trait loci (eQTL) datasets (N = 1890). Furthermore, we applied various bioinformatics analyses, including MAGMA gene-based analysis, pathway enrichment analysis, drug/disease-based enrichment analysis, computer-based permutation analysis, PPI network analysis, gene co-expression analysis and differential gene expression analysis, to prioritize susceptible genes associated with childhood-onset asthma. RESULTS Based on comprehensive genomics analyses, we found 31 genes with multiple eSNPs to be convincing candidates for childhood-onset asthma risk; such as, PSMB9 (cis-rs4148882 and cis-rs2071534) and TAP2 (cis-rs9267798, cis-rs4148882, cis-rs241456, and trans-10,447,456). These 31 genes were functionally interacted with each other in our PPI network analysis. Our pathway enrichment analysis showed that numerous KEGG pathways including antigen processing and presentation, type I diabetes mellitus, and asthma were significantly enriched to involve in childhood-onset asthma risk. The co-expression patterns among 31 genes were remarkably altered according to asthma status, and 25 of 31 genes (25/31 = 80.65%) showed significantly or suggestively differential expression between asthma group and control group. CONCLUSIONS We provide strong evidence to highlight 31 candidate genes for childhood-onset asthma risk, and offer a new insight into the genetic pathogenesis of childhood-onset asthma.
Collapse
Affiliation(s)
- Xiuqing Ma
- Department of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100853 China
| | - Peilan Wang
- Outpatient Department, Chinese PLA General Hospital, Beijing, 100853 China
| | - Guobing Xu
- Department of Cardiovascular Medicine, Zhongxiang People’s Hospital, Zhongxiang, 431900 Hubei Province China
| | - Fang Yu
- Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100853 China
| | - Yunlong Ma
- Institute of Biomedical Big Data, School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027 P. R. China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Song W, Zheng S, Li M, Zhang X, Cao R, Ye C, Shao R, Li G, Li J, Liu S, Li H, Li L. Linking endotypes to omics profiles in difficult-to-control asthma using the diagnostic Chinese medicine syndrome differentiation algorithm. J Asthma 2020; 57:532-542. [PMID: 30915875 DOI: 10.1080/02770903.2019.1590589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/27/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
Abstract
Objective: Patients with difficult-to-control asthma have difficulty breathing almost all of the time, even leading to life-threatening asthma attacks. However, only few diagnostic markers for this disease have been identified. We aimed to take advantage of unique Chinese medicine theories for phenotypic classification and to explore molecular signatures in difficult-to-control asthma. Methods: The Chinese medicine syndrome differentiation algorithm (CMSDA) is a syndrome-scoring classification method based on the Chinese medicine overall observation theory. Patients with difficult-to-control asthma were classified into Cold- and Hot-pattern groups according to the CMSDA. DNA methylation and metabolomic profiles were obtained using Infinium Human Methylation 450 BeadChip and gas chromatography-mass spectrometer. Subsequently, an integrated bioinformatics analysis was performed to compare those two patterns and identify Cold/Hot-associated candidates, followed by functional validation studies. Results: A total of 20 patients with difficult-to-control asthma were enrolled in the study. Ten were grouped as Cold and 10 as Hot according to the CMSDA. We identified distinct whole-genome DNA methylation and metabolomic profiles between Cold- and Hot-pattern groups. ALDH3A1 gene exhibited variations in the DNA methylation probe cg10791966, while two metabolic pathways were associated with those two patterns. Conclusions: Our study introduced a novel diagnostic classification approach, the CMSDA, for difficult-to-control asthma. This is an alternative way to categorize diverse syndromes and link endotypes with omics profiles of this disease. ALDH3A1 might be a potential biomarker for precision diagnosis of difficult-to-control asthma.
Collapse
Affiliation(s)
- Wenping Song
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Si Zheng
- Institute of Medical Information (IMI) and Library, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Meng Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xia Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Cao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Cheng Ye
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Guangxi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiao Li
- Institute of Medical Information (IMI) and Library, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Shigang Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Farzan N, Vijverberg SJ, Kabesch M, Sterk PJ, Maitland-van der Zee AH. The use of pharmacogenomics, epigenomics, and transcriptomics to improve childhood asthma management: Where do we stand? Pediatr Pulmonol 2018; 53:836-845. [PMID: 29493882 DOI: 10.1002/ppul.23976] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 02/01/2018] [Indexed: 01/11/2023]
Abstract
Asthma is a complex multifactorial disease and it is the most common chronic disease in children. There is a high variability in response to asthma treatment, even in patients with good adherence to maintenance treatment, and a correct inhalation technique. Distinct underlying disease mechanisms in childhood asthma might be the reason of this heterogeneity. A deeper knowledge of the underlying molecular mechanisms of asthma has led to the recent development of advanced and mechanism-based treatments such as biologicals. However, biologicals are recommended only for patients with specific asthma phenotypes who remain uncontrolled despite high dosages of conventional asthma treatment. One of the main unmet needs in their application is lack of clinically available biomarkers to individualize pediatric asthma management and guide treatment. Pharmacogenomics, epigenomics, and transcriptomics are three omics fields that are rapidly advancing and can provide tools to identify novel asthma mechanisms and biomarkers to guide treatment. Pharmacogenomics focuses on variants in the DNA, epigenomics studies heritable changes that do not involve changes in the DNA sequence but lead to alteration of gene expression, and transcriptomics investigates gene expression by studying the complete set of mRNA transcripts in a cell or a population of cells. Advances in high-throughput technologies and statistical tools together with well-phenotyped patient inclusion and collaborations between different centers will expand our knowledge of underlying molecular mechanisms involved in disease onset and progress. Furthermore, it could help to select and stratify appropriate therapeutic strategies for subgroups of patients and hopefully bring precision medicine to daily practice.
Collapse
Affiliation(s)
- Niloufar Farzan
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne J Vijverberg
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Peter J Sterk
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Sánchez-Martín A, García-Sánchez A, Isidoro-García M. Review on Pharmacogenetics and Pharmacogenomics Applied to the Study of Asthma. Methods Mol Biol 2017; 1434:255-72. [PMID: 27300544 DOI: 10.1007/978-1-4939-3652-6_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nearly one-half of asthmatic patients do not respond to the most common therapies. Evidence suggests that genetic factors may be involved in the heterogeneity in therapeutic response and adverse events to asthma therapies. We focus on the three major classes of asthma medication: β-adrenergic receptor agonist, inhaled corticosteroids, and leukotriene modifiers. Pharmacogenetics and pharmacogenomics studies have identified several candidate genes associated with drug response.In this chapter, the main pharmacogenetic and pharmacogenomic studies in addition to the future perspectives in personalized medicine will be reviewed. The ideal treatment of asthma would be a tailored approach to health care in which adverse effects are minimized and the therapeutic benefit for an individual asthmatic is maximized leading to a more cost-effective care.
Collapse
Affiliation(s)
- Almudena Sánchez-Martín
- Department of Pharmacy, University Hospital of Salamanca, Salamanca, Spain.,Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain
| | - Asunción García-Sánchez
- Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain.,Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - María Isidoro-García
- Salamanca Institute for Biomedical Research (IBSAL), Salamanca, Spain. .,Department of Clinical Biochemistry, University Hospital of Salamanca, Salamanca, Spain. .,Department of Medicine, University of Salamanca, Salamanca, Spain.
| |
Collapse
|
12
|
van Meel ER, den Dekker HT, Elbert NJ, Jansen PW, Moll HA, Reiss IK, de Jongste JC, Jaddoe VWV, Duijts L. A population-based prospective cohort study examining the influence of early-life respiratory tract infections on school-age lung function and asthma. Thorax 2017. [PMID: 29101282 DOI: 10.1136/thoraxjnl‐2017‐210149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND Early-life respiratory tract infections could affect airway obstruction and increase asthma risk in later life. However, results from previous studies are inconsistent. OBJECTIVE We examined the associations of early-life respiratory tract infections with lung function and asthma in school-aged children. METHODS This study among 5197 children born between April 2002 and January 2006 was embedded in a population-based prospective cohort study. Information on physician-attended upper and lower respiratory tract infections until age 6 years (categorised into ≤ 3 and >3-6 years) was obtained by annual questionnaires. Spirometry measures and physician-diagnosed asthma were assessed at age 10 years. RESULTS Upper respiratory tract infections were not associated with adverse respiratory outcomes. Compared with children without lower respiratory tract infections ≤3 years, children with lower respiratory tract infections ≤3 years had a lower FEV1, FVC, FEV1:FVC and forced expiratory flow at 75% of FVC (FEF75) (Z-score (95% CI): ranging from -0.22 (-0.31 to -0.12) to -0.12 (-0.21 to -0.03)) and an increased risk of asthma (OR (95% CI): 1.79 (1.19 to 2.59)). Children with lower respiratory tract infections >3-6 years had an increased risk of asthma (3.53 (2.37 to 5.17)) only. Results were not mediated by antibiotic or paracetamol use and not modified by inhalant allergic sensitisation. Cross-lagged modelling showed that results were not bidirectional and independent of preschool wheezing patterns. CONCLUSION Early-life lower respiratory tract infections ≤3 years are most consistently associated with lower lung function and increased risk of asthma in school-aged children.
Collapse
Affiliation(s)
- Evelien R van Meel
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Herman T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Niels J Elbert
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Pauline W Jansen
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Psychology, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Henriëtte A Moll
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Irwin K Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Johan C de Jongste
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Liesbeth Duijts
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
13
|
van Meel ER, den Dekker HT, Elbert NJ, Jansen PW, Moll HA, Reiss IK, de Jongste JC, Jaddoe VWV, Duijts L. A population-based prospective cohort study examining the influence of early-life respiratory tract infections on school-age lung function and asthma. Thorax 2017; 73:167-173. [PMID: 29101282 DOI: 10.1136/thoraxjnl-2017-210149] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND Early-life respiratory tract infections could affect airway obstruction and increase asthma risk in later life. However, results from previous studies are inconsistent. OBJECTIVE We examined the associations of early-life respiratory tract infections with lung function and asthma in school-aged children. METHODS This study among 5197 children born between April 2002 and January 2006 was embedded in a population-based prospective cohort study. Information on physician-attended upper and lower respiratory tract infections until age 6 years (categorised into ≤ 3 and >3-6 years) was obtained by annual questionnaires. Spirometry measures and physician-diagnosed asthma were assessed at age 10 years. RESULTS Upper respiratory tract infections were not associated with adverse respiratory outcomes. Compared with children without lower respiratory tract infections ≤3 years, children with lower respiratory tract infections ≤3 years had a lower FEV1, FVC, FEV1:FVC and forced expiratory flow at 75% of FVC (FEF75) (Z-score (95% CI): ranging from -0.22 (-0.31 to -0.12) to -0.12 (-0.21 to -0.03)) and an increased risk of asthma (OR (95% CI): 1.79 (1.19 to 2.59)). Children with lower respiratory tract infections >3-6 years had an increased risk of asthma (3.53 (2.37 to 5.17)) only. Results were not mediated by antibiotic or paracetamol use and not modified by inhalant allergic sensitisation. Cross-lagged modelling showed that results were not bidirectional and independent of preschool wheezing patterns. CONCLUSION Early-life lower respiratory tract infections ≤3 years are most consistently associated with lower lung function and increased risk of asthma in school-aged children.
Collapse
Affiliation(s)
- Evelien R van Meel
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Herman T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Niels J Elbert
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Pauline W Jansen
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Institute of Psychology, Erasmus University Rotterdam, Rotterdam, Netherlands
| | - Henriëtte A Moll
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Irwin K Reiss
- Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Johan C de Jongste
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Liesbeth Duijts
- Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.,Division of Neonatology, Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
14
|
Jiang LL, Xie JK, Cui JQ, Wei D, Yin BL, Zhang YN, Chen YH, Han X, Wang Q, Zhang CL. Promoter methylation of yes-associated protein (YAP1) gene in polycystic ovary syndrome. Medicine (Baltimore) 2017; 96:e5768. [PMID: 28079802 PMCID: PMC5266164 DOI: 10.1097/md.0000000000005768] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND DNA methylation modification has been proved to influence the phenotype of polycystic ovary syndrome (PCOS). Genome-wide association studies (GWAS) demonstrate that yes-associated protein (YAP1) genetic sites are associated with PCOS. The study aims to detect the methylation status of YAP1 promoter in ovary granulosa cells (GCs) of PCOS patients and explore novel therapeutic targets for PCOS. METHODS Randomized controlled trial was applied and a total of 72 women were included in the study, including 36 cases of PCOS patients and 36 cases of health controls. Ovary GCs were extracted from in vitro fertilization embryo transfer. Methylation status of YAP1 promoter was detected by bisulfite sequencing PCR (BSP). Protein and mRNA expression of YAP1 were measured by western blotting and real-time quantitate PCR. RESULTS Overall methylation level of YAP1 promoter region from PCOS group was significantly lower than that from control group. CpG sites analysis revealed that 12 sites (-443, -431, -403, -371, -331, -120, -49, -5, +1, +9, +15, +22) were significantly hypomethylated in women with PCOS (P < 0.05). A significant upregulation of YAP1 mRNA and protein expression levels was observed. Testosterone concentration could alleviate the methylation status and demonstrate obvious dose-dependent relation. CONCLUSION Our research achievements manifest that hypomethylation of YAP1 promoter promotes the YAP1 expression, which plays a key role in the pathogenesis and accelerate PCOS.
Collapse
Affiliation(s)
- Li-Le Jiang
- Second Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province
| | - Juan-Ke Xie
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Jin-Quan Cui
- Second Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province
| | - Duo Wei
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Bao-Li Yin
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Ya-Nan Zhang
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Yuan-Hui Chen
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Xiao Han
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Qian Wang
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| | - Cui-Lian Zhang
- Zhengzhou University, Zhengzhou, Henan Province
- Reproductive Medical Center, People's Hospital of Henan Province, Zhengzhou, Henan Province, P.R. China
| |
Collapse
|
15
|
Xiong W, Su Y, Ding J. Empirical likelihood-based robust tests for genetic association analysis with quantitative traits. J Appl Stat 2016. [DOI: 10.1080/02664763.2016.1266469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Wenjun Xiong
- School of Mathematics and Statistics, Guangxi Normal University, Guilin, People's Republic of China
| | - You Su
- School of Mathematics and Statistics, Guangxi Normal University, Guilin, People's Republic of China
| | - Juan Ding
- School of Mathematics and Statistics, Guangxi Normal University, Guilin, People's Republic of China
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
16
|
Guglani L, Joseph CL. Asthma and diet: could food be thy medicine? Indian Pediatr 2015; 52:21-22. [PMID: 25638178 PMCID: PMC4864955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Affiliation(s)
- Lokesh Guglani
- Pediatric Pulmonary Division, Department of Pediatrics, Childrens Hospital of Michigan and Wayne State University School of Medicine; and #Department of Public Health Sciences, Henry Ford Health System; Detroit MI, USA.
| | | |
Collapse
|