1
|
Sharma R, Tiwari A, Kho AT, Wang AL, Srivastava U, Piparia S, Desai B, Wong R, Celedón JC, Peters SP, Smith LJ, Irvin CG, Castro M, Weiss ST, Tantisira KG, McGeachie MJ. Circulating microRNAs associated with bronchodilator response in childhood asthma. BMC Pulm Med 2024; 24:553. [PMID: 39497092 PMCID: PMC11536898 DOI: 10.1186/s12890-024-03372-4] [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: 10/06/2023] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Bronchodilator response (BDR) is a measure of improvement in airway smooth muscle tone, inhibition of liquid accumulation and mucus section into the lumen in response to short-acting beta-2 agonists that varies among asthmatic patients. MicroRNAs (miRNAs) are well-known post-translational regulators. Identifying miRNAs associated with BDR could lead to a better understanding of the underlying complex pathophysiology. OBJECTIVE The purpose of this study is to identify circulating miRNAs associated with bronchodilator response in asthma and decipher possible mechanism of bronchodilator response variation. METHODS We used available small RNA sequencing on blood serum from 1,134 asthmatic children aged 6 to 14 years who participated in the Genetics of Asthma in Costa Rica Study (GACRS). We filtered the participants into the highest and lowest bronchodilator response (BDR) quartiles and used DeSeq2 to identify miRNAs with differential expression (DE) in high (N = 277) vs. low (N = 278) BDR group. Replication was carried out in the Leukotriene modifier Or Corticosteroids or Corticosteroid-Salmeterol trial (LOCCS), an adult asthma cohort. The putative target genes of DE miRNAs were identified, and pathway enrichment analysis was performed. RESULTS We identified 10 down-regulated miRNAs having odds ratios (OR) between 0.37 and 0.76 for a doubling of miRNA counts and one up-regulated miRNA (OR = 2.26) between high and low BDR group. These were assessed for replication in the LOCCS cohort, where two miRNAs (miR-200b-3p and miR-1246) were associated. Further, functional annotation of 11 DE miRNAs were performed as well as of two replicated miRs. Target genes of these miRs were enriched in regulation of cholesterol biosynthesis by SREBPs, ESR-mediated signaling, G1/S transition, RHO GTPase cycle, and signaling by TGFB family pathways. CONCLUSION MiRNAs miR-1246 and miR-200b-3p are associated with both childhood and adult asthma BDR. Our findings add to the growing body of evidence that miRNAs play a significant role in the difference of asthma treatment response among patients as it points to genomic regulatory machinery underlying difference in bronchodilator response among patients. TRIAL REGISTRATION LOCCS cohort [ClinicalTrials.gov number NCT00156819, Registration date 20050912], GACRS cohort [ClinicalTrials.gov number NCT00021840].
Collapse
Affiliation(s)
- Rinku Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Anshul Tiwari
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alvin T Kho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Alberta L Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Upasna Srivastava
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
- Department of MEDCSC Neurodevelopment (Child Study Center), Yale University School of Medicine, New Haven, CT, USA
| | - Shraddha Piparia
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Brinda Desai
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Richard Wong
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen P Peters
- Department of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Lewis J Smith
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Charles G Irvin
- Pulmonary and Critical Care Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, KS, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, University of California San Diego and Rady Children's Hospital, San Diego, CA, USA
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Vasileva D, Greenwood CMT, Daley D. A Review of the Epigenetic Clock: Emerging Biomarkers for Asthma and Allergic Disease. Genes (Basel) 2023; 14:1724. [PMID: 37761864 PMCID: PMC10531327 DOI: 10.3390/genes14091724] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
DNA methylation (DNAm) is a dynamic, age-dependent epigenetic modification that can be used to study interactions between genetic and environmental factors. Environmental exposures during critical periods of growth and development may alter DNAm patterns, leading to increased susceptibility to diseases such as asthma and allergies. One method to study the role of DNAm is the epigenetic clock-an algorithm that uses DNAm levels at select age-informative Cytosine-phosphate-Guanine (CpG) dinucleotides to predict epigenetic age (EA). The difference between EA and calendar age (CA) is termed epigenetic age acceleration (EAA) and reveals information about the biological capacity of an individual. Associations between EAA and disease susceptibility have been demonstrated for a variety of age-related conditions and, more recently, phenotypes such as asthma and allergic diseases, which often begin in childhood and progress throughout the lifespan. In this review, we explore different epigenetic clocks and how they have been applied, particularly as related to childhood asthma. We delve into how in utero and early life exposures (e.g., smoking, air pollution, maternal BMI) result in methylation changes. Furthermore, we explore the potential for EAA to be used as a biomarker for asthma and allergic diseases and identify areas for further study.
Collapse
Affiliation(s)
- Denitsa Vasileva
- Centre for Heart Lung Innovation, University of British Columbia and Saint Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada;
| | - Celia M. T. Greenwood
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada;
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC H3A 0G4, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G4, Canada
| | - Denise Daley
- Centre for Heart Lung Innovation, University of British Columbia and Saint Paul’s Hospital, Vancouver, BC V6Z 1Y6, Canada;
- Department of Medicine, Respiratory Division, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
3
|
Sharma R, Tiwari A, Kho AT, Wang AL, Srivastava U, Piparia S, Desai B, Wong R, Celedón JC, Peters SP, Smith LJ, Irvin CG, Castro M, Weiss ST, Tantisira KG, McGeachie MJ. Circulating MicroRNAs associated with Bronchodilator Response in Childhood Asthma. RESEARCH SQUARE 2023:rs.3.rs-3101724. [PMID: 37461659 PMCID: PMC10350209 DOI: 10.21203/rs.3.rs-3101724/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Rationale Bronchodilator response (BDR) is a measure of improvement in airway smooth muscle tone, inhibition of liquid accumulation and mucus section into the lumen in response to short-acting beta-2 agonists that varies among asthmatic patients. MicroRNAs (miRNAs) are well-known post-translational regulators. Identifying miRNAs associated with BDR could lead to a better understanding of the underlying complex pathophysiology. Objective The purpose of this study is to identify circulating miRNAs associated with bronchodilator response in asthma and decipher possible mechanism of bronchodilator response variation. Methods We used available small RNA sequencing on blood serum from 1,134 asthmatic children aged 6 to 14 years who participated in the Genetics of Asthma in Costa Rica Study (GACRS). We filtered the participants into high and low bronchodilator response (BDR) quartiles and used DeSeq2 to identify miRNAs with differential expression (DE) in high (N= 277) vs low (N= 278) BDR group. Replication was carried out in the Leukotriene modifier Or Corticosteroids or Corticosteroid-Salmeterol trial (LOCCS), an adult asthma cohort. The putative target genes of DE miRNAs were identified, and pathway enrichment analysis was performed. Results We identified 10 down-regulated miRNAs having odds ratios (OR) between 0.37 and 0.76 for a doubling of miRNA counts and one up-regulated miRNA (OR=2.26) between high and low BDR group. These were assessed for replication in the LOCCS cohort, where two miRNAs (miR-200b-3p and miR-1246) were associated. Further, functional annotation of 11 DE miRNAs were performed as well as of two replicated miRs. Target genes of these miRs were enriched in regulation of cholesterol biosynthesis by SREBPs, ESR-mediated signaling, G1/S transition, RHO GTPase cycle, and signaling by TGFB family pathways. Conclusion MiRNAs miR-1246 and miR-200b-3p are associated with both childhood and adult asthma BDR. Our findings add to the growing body of evidence that miRNAs play a significant role in the difference of asthma treatment response among patients as it points to genomic regulatory machinery underlying difference in bronchodilator response among patients. Trial registration LOCCS cohort [ClinicalTrials.gov number: NCT00156819], GACRS cohort [ClinicalTrials.gov number: NCT00021840].
Collapse
Affiliation(s)
- Rinku Sharma
- Brigham and Women's Hospital and Harvard Medical School
| | | | - Alvin T Kho
- Brigham and Women's Hospital and Harvard Medical School
| | | | | | | | - Brinda Desai
- University of California San Diego and Rady Children's Hospital
| | - Richard Wong
- University of California San Diego and Rady Children's Hospital
| | - Juan C Celedón
- University of Pittsburgh, UPMC Children's Hospital of Pittsburgh
| | | | | | | | | | - Scott T Weiss
- Brigham and Women's Hospital and Harvard Medical School
| | | | | |
Collapse
|
4
|
Khare M, Piparia S, Tantisira KG. Pharmacogenetics of childhood uncontrolled asthma. Expert Rev Clin Immunol 2023:1-14. [PMID: 37190963 PMCID: PMC10657335 DOI: 10.1080/1744666x.2023.2214363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Asthma is a heterogeneous, multifactorial disease with multiple genetic and environmental risk factors playing a role in pathogenesis and therapeutic response. Understanding of pharmacogenetics can help with matching individualized treatments to specific genotypes of asthma to improve therapeutic outcomes especially in uncontrolled or severe asthma. AREAS COVERED In this review, we outline novel information about biology, pathways, and mechanisms related to interindividual variability in drug response (corticosteroids, bronchodilators, leukotriene modifiers, and biologics) for childhood asthma. We discuss candidate gene, genome-wide association studies and newer omics studies including epigenomics, transcriptomics, proteomics, and metabolomics as well as integrative genomics and systems biology methods related to childhood asthma. The articles were obtained after a series of searches, last updated November 2022, using database PubMed/CINAHL DB. EXPERT OPINION Implementation of pharmacogenetic algorithms can improve therapeutic targeting in children with asthma, particularly with severe or uncontrolled asthma who typically have challenges in clinical management and carry considerable financial burden. Future studies focusing on potential biomarkers both clinical and pharmacogenetic can help formulate a prognostic test for asthma treatment response that would represent true bench to bedside clinical implementation.
Collapse
Affiliation(s)
- Manaswitha Khare
- Division of Pediatric Hospital Medicine, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Division of Pediatric Hospital Medicine, Department of Pediatrics, Rady Children's Hospital of San Diego, San Diego, CA, USA
| | - Shraddha Piparia
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, Rady Children's Hospital of San Diego, San Diego, CA, USA
| |
Collapse
|
5
|
Mabelane T, Masekela R, Dandara C, Hadebe S. Immunogenetics and pharmacogenetics of allergic asthma in Africa. FRONTIERS IN ALLERGY 2023; 4:1165311. [PMID: 37228580 PMCID: PMC10203899 DOI: 10.3389/falgy.2023.1165311] [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: 02/13/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
Asthma is a common chronic condition in children and in an African setting is often highly prevalent in urban areas as compared to rural areas. Asthma is a heritable disease and the genetic risk is often exacerbated by unique localised environmental factors. The Global Initiative for Asthma (GINA) recommendation for the control of asthma includes inhaled corticosteroids (ICS) alone or together with short-acting β2-agonists (SABA) or long-acting β2-agonists (LABA). While these drugs can relieve asthma symptoms, there is evidence of reduced efficacy in people of African ancestry. Whether this is due to immunogenetics, genomic variability in drug metabolising genes (pharmacogenetics) or genetics of asthma-related traits is not well defined. Pharmacogenetic evidence of first-line asthma drugs in people of African ancestry is lacking and is further compounded by the lack of representative genetic association studies in the continent. In this review, we will discuss the paucity of data related to the pharmacogenetics of asthma drugs in people of African ancestry, mainly drawing from African American data. We will further discuss how this gap can be bridged to improve asthma health outcomes in Africa.
Collapse
Affiliation(s)
- Tshegofatso Mabelane
- Department of Medicine, Sefako Makgatho Health Science University, Ga-Rankuwa, South Africa
| | - Refiloe Masekela
- Department of Paediatrics, Nelson Mandela School of Medicine, Inkosi Albert Luthuli Hospital, University of KwaZulu-Natal, Durban, South Africa
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences and Institute of Infectious Diseases Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Platform for Pharmacogenomics Research and Translation, South African Medical Research Council, Cape Town, South Africa
| | - Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
6
|
Stikker BS, Hendriks RW, Stadhouders R. Decoding the genetic and epigenetic basis of asthma. Allergy 2023; 78:940-956. [PMID: 36727912 DOI: 10.1111/all.15666] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/03/2023]
Abstract
Asthma is a complex and heterogeneous chronic inflammatory disease of the airways. Alongside environmental factors, asthma susceptibility is strongly influenced by genetics. Given its high prevalence and our incomplete understanding of the mechanisms underlying disease susceptibility, asthma is frequently studied in genome-wide association studies (GWAS), which have identified thousands of genetic variants associated with asthma development. Virtually all these genetic variants reside in non-coding genomic regions, which has obscured the functional impact of asthma-associated variants and their translation into disease-relevant mechanisms. Recent advances in genomics technology and epigenetics now offer methods to link genetic variants to gene regulatory elements embedded within non-coding regions, which have started to unravel the molecular mechanisms underlying the complex (epi)genetics of asthma. Here, we provide an integrated overview of (epi)genetic variants associated with asthma, focusing on efforts to link these disease associations to biological insight into asthma pathophysiology using state-of-the-art genomics methodology. Finally, we provide a perspective as to how decoding the genetic and epigenetic basis of asthma has the potential to transform clinical management of asthma and to predict the risk of asthma development.
Collapse
Affiliation(s)
- Bernard S Stikker
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Cell Biology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Bai H, Zhang X, Bush WS. Pharmacogenomic and Statistical Analysis. Methods Mol Biol 2023; 2629:305-330. [PMID: 36929083 DOI: 10.1007/978-1-0716-2986-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Genetic variants can alter response to drugs and other therapeutic interventions. The study of this phenomenon, called pharmacogenomics, is similar in many ways to other types of genetic studies but has distinct methodological and statistical considerations. Genetic variants involved in the processing of exogenous compounds exhibit great diversity and complexity, and the phenotypes studied in pharmacogenomics are also more complex than typical genetic studies. In this chapter, we review basic concepts in pharmacogenomic study designs, data generation techniques, statistical analysis approaches, and commonly used methods and briefly discuss the ultimate translation of findings to clinical care.
Collapse
Affiliation(s)
- Haimeng Bai
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Xueyi Zhang
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
8
|
Gereige JD, Xu H, Ortega VE, Cho MH, Liu M, Sakornsakolpat P, Silverman EK, Beaty TH, Miller BE, Bakke P, Gulsvik A, Hersh CP, Morrow JD, Ampleford EJ, Hawkins GA, Bleecker ER, Meyers DA, Peters SP, Celedón JC, Tantisira K, Li J, Dupuis J, O'Connor GT. A genome-wide association study of bronchodilator response in participants of European and African ancestry from six independent cohorts. ERJ Open Res 2022; 8:00484-2021. [PMID: 35769418 PMCID: PMC9234425 DOI: 10.1183/23120541.00484-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 05/08/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Bronchodilator response (BDR) is a measurement of acute bronchodilation in response to short-acting β2-agonists, with a heritability between 10 and 40%. Identifying genetic variants associated with BDR may lead to a better understanding of its complex pathophysiology. Methods We performed a genome-wide association study (GWAS) of BDR in six adult cohorts with participants of European ancestry (EA) and African ancestry (AA) including community cohorts and cohorts ascertained on the basis of obstructive pulmonary disease. Validation analysis was carried out in two paediatric asthma cohorts. Results A total of 10 623 EA and 3597 AA participants were included in the analyses. No single nucleotide polymorphism (SNP) was associated with BDR at the conventional genome-wide significance threshold (p<5×10-8). Performing fine mapping and using a threshold of p<5×10-6 to identify suggestive variants of interest, we identified three SNPs with possible biological relevance: rs35870000 (within FREM1), which may be involved in IgE- and IL5-induced changes in airway smooth muscle cell responsiveness; rs10426116 (within ZNF284), a zinc finger protein, which has been implicated in asthma and BDR previously; and rs4782614 (near ATP2C2), involved in calcium transmembrane transport. Validation in paediatric cohorts yielded no significant SNPs, possibly due to age-genotype interaction effects. Conclusion Ancestry-stratified and ancestry-combined GWAS meta-analyses of over 14 000 participants did not identify genetic variants associated with BDR at the genome-wide significance threshold, although a less stringent threshold identified three variants showing suggestive evidence of association. A common definition and protocol for measuring BDR in research may improve future efforts to identify variants associated with BDR.
Collapse
Affiliation(s)
- Jessica D. Gereige
- Division of Pulmonary, Allergy, Sleep, and Critical Care Medicine, Boston Medical Center, Boston, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Hanfei Xu
- Dept of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Victor E. Ortega
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ming Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Bioinformatics and Computational Biology Program, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Phuwanat Sakornsakolpat
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Terri H. Beaty
- Dept of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Per Bakke
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Amund Gulsvik
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Craig P. Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jarrett D. Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Elizabeth J. Ampleford
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Gregory A. Hawkins
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Eugene R. Bleecker
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Deborah A. Meyers
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Stephen P. Peters
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelan Tantisira
- Division of Pediatric Respiratory Medicine, University of California and Rady Children's Hospital, San Diego, CA, USA
| | - Jiang Li
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Josée Dupuis
- Dept of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - George T. O'Connor
- Division of Pulmonary, Allergy, Sleep, and Critical Care Medicine, Boston Medical Center, Boston, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
9
|
Pharmacogenetics of Bronchodilator Response: Future Directions. Curr Allergy Asthma Rep 2021; 21:47. [PMID: 34958416 DOI: 10.1007/s11882-021-01023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE OF REVIEW Several genome-wide association studies (GWASs) of bronchodilator response (BDR) to albuterol have been published over the past decade. This review describes current knowledge gaps, including pharmacogenetic studies of albuterol response in minority populations, effect modification of pharmacogenetic associations by age, and relevance of BDR phenotype characterization to pharmacogenetic findings. New approaches, such as leveraging additional "omics" data to focus pharmacogenetic interrogation, as well as developing polygenic risk scores in asthma treatment responses, are also discussed. RECENT FINDINGS Recent pharmacogenetic studies of albuterol response in minority populations have identified genetic polymorphisms in loci (DNAH5, NFKB1, PLCB1, ADAMTS3, COX18, and PRKG1), that are associated with BDR. Additional studies are needed to replicate these findings. Modification of the pharmacogenetic associations for SPATS2L and ASB3 polymorphisms by age has also been published. Evidence from metabolomic and epigenomic studies of BDR may point to new pharmacogenetic targets. Lastly, a polygenic risk score for response to albuterol has been developed but requires validation in additional cohorts. In order to expand our knowledge of pharmacogenetics of BDR, additional studies in minority populations are needed. Consideration of effect modification by age and leverage of other "omics" data beyond genomics may also help uncover novel pharmacogenetic loci for use in precision medicine for asthma treatment.
Collapse
|
10
|
Lamb HJ, Hayes BJ, Randhawa IAS, Nguyen LT, Ross EM. Genomic prediction using low-coverage portable Nanopore sequencing. PLoS One 2021; 16:e0261274. [PMID: 34910782 PMCID: PMC8673642 DOI: 10.1371/journal.pone.0261274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/26/2021] [Indexed: 11/18/2022] Open
Abstract
Most traits in livestock, crops and humans are polygenic, that is, a large number of loci contribute to genetic variation. Effects at these loci lie along a continuum ranging from common low-effect to rare high-effect variants that cumulatively contribute to the overall phenotype. Statistical methods to calculate the effect of these loci have been developed and can be used to predict phenotypes in new individuals. In agriculture, these methods are used to select superior individuals using genomic breeding values; in humans these methods are used to quantitatively measure an individual’s disease risk, termed polygenic risk scores. Both fields typically use SNP array genotypes for the analysis. Recently, genotyping-by-sequencing has become popular, due to lower cost and greater genome coverage (including structural variants). Oxford Nanopore Technologies’ (ONT) portable sequencers have the potential to combine the benefits genotyping-by-sequencing with portability and decreased turn-around time. This introduces the potential for in-house clinical genetic disease risk screening in humans or calculating genomic breeding values on-farm in agriculture. Here we demonstrate the potential of the later by calculating genomic breeding values for four traits in cattle using low-coverage ONT sequence data and comparing these breeding values to breeding values calculated from SNP arrays. At sequencing coverages between 2X and 4X the correlation between ONT breeding values and SNP array-based breeding values was > 0.92 when imputation was used and > 0.88 when no imputation was used. With an average sequencing coverage of 0.5x the correlation between the two methods was between 0.85 and 0.92 using imputation, depending on the trait. This suggests that ONT sequencing has potential for in clinic or on-farm genomic prediction, however, further work to validate these findings in a larger population still remains.
Collapse
Affiliation(s)
- Harrison J. Lamb
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
- * E-mail:
| | - Ben J. Hayes
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Imtiaz A. S. Randhawa
- School of Veterinary Science, The University of Queensland, Brisbane, QLD, Australia
| | - Loan T. Nguyen
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| | - Elizabeth M. Ross
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
11
|
Role of Epigenetics in the Pathogenesis, Treatment, Prediction, and Cellular Transformation of Asthma. Mediators Inflamm 2021; 2021:9412929. [PMID: 34566492 PMCID: PMC8457970 DOI: 10.1155/2021/9412929] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Asthma is a mysterious disease with heterogeneity in etiology, pathogenesis, and clinical phenotypes. Although ongoing studies have provided a better understanding of asthma, its natural history, progression, pathogenesis, diversified phenotypes, and even the exact epigenetic linkage between childhood asthma and adult-onset/old age asthma remain elusive in many aspects. Asthma heritability has been established through genetic studies, but genetics is not the only influencing factor in asthma. The increasing incidence and some unsolved queries suggest that there may be other elements related to asthma heredity. Epigenetic mechanisms link genetic and environmental factors with developmental trajectories in asthma. This review provides an overview of asthma epigenetics and its components, including several epigenetic studies on asthma, and discusses the epigenetic linkage between childhood asthma and adult-onset/old age asthma. Studies involving asthma epigenetics present valuable novel approaches to solve issues related to asthma. Asthma epigenetic research guides us towards gene therapy and personalized T cell therapy, directs the discovery of new therapeutic agents, predicts long-term outcomes in severe cases, and is also involved in the cellular transformation of childhood asthma to adult-onset/old age asthma.
Collapse
|
12
|
Muhammad A, Aka IT, Birdwell KA, Gordon AS, Roden DM, Wei WQ, Mosley JD, Van Driest SL. Genome-Wide Approach to Measure Variant-Based Heritability of Drug Outcome Phenotypes. Clin Pharmacol Ther 2021; 110:714-722. [PMID: 34151428 PMCID: PMC8376753 DOI: 10.1002/cpt.2323] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/16/2021] [Indexed: 12/18/2022]
Abstract
Pharmacogenomic studies have successfully identified variants-typically with large effect sizes in drug target and metabolism enzymes-that predict drug outcome phenotypes. However, these variants may account for a limited proportion of phenotype variability attributable to the genome. Using genome-wide common variation, we measured the narrow-sense heritability ( h SNP 2 ) of seven pharmacodynamic and five pharmacokinetic phenotypes across three cardiovascular drugs, two antibiotics, and three immunosuppressants. We used a Bayesian hierarchical mixed model, BayesR, to model the distribution of genome-wide variant effect sizes for each drug phenotype as a mixture of four normal distributions of fixed variance (0, 0.01%, 0.1%, and 1% of the total additive genetic variance). This model allowed us to parse h SNP 2 into bins representing contributions of no-effect, small-effect, moderate-effect, and large-effect variants, respectively. For the 12 phenotypes, a median of 969 (range 235-6,304) unique individuals of European ancestry and a median of 1,201,626 (range 777,427-1,514,275) variants were included in our analyses. The number of variants contributing to h SNP 2 ranged from 2,791 to 5,356 (median 3,347). Estimates for h SNP 2 ranged from 0.05 (angiotensin-converting enzyme inhibitor-induced cough) to 0.59 (gentamicin concentration). Small-effect and moderate-effect variants contributed a majority to h SNP 2 for every phenotype (range 61-95%). We conclude that drug outcome phenotypes are highly polygenic. Thus, larger genome-wide association studies of drug phenotypes are needed both to discover novel variants and to determine how genome-wide approaches may improve clinical prediction of drug outcomes.
Collapse
Affiliation(s)
- Ayesha Muhammad
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Ida T. Aka
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kelly A. Birdwell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Adam S. Gordon
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan D. Mosley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sara L. Van Driest
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
13
|
Fishe JN, Labilloy G, Higley R, Casey D, Ginn A, Baskovich B, Blake KV. Single Nucleotide Polymorphisms (SNPs) in PRKG1 & SPATA13-AS1 are associated with bronchodilator response: a pilot study during acute asthma exacerbations in African American children. Pharmacogenet Genomics 2021; 31:146-154. [PMID: 33851947 PMCID: PMC8373649 DOI: 10.1097/fpc.0000000000000434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Inhaled bronchodilators are the first-line treatment for asthma exacerbations, but individual bronchodilator response (BDR) varies by race and ethnicity. Studies have examined BDR's genetic underpinnings, but many did not include children or were not conducted during an asthma exacerbation. This pilot study tested single-nucleotide polymorphisms' (SNPs') association with pediatric African American BDR during an acute asthma exacerbation. METHODS This was a study of pediatric asthma patients in the age group 2-18 years treated in the emergency department for an asthma exacerbation. We measured BDR before and after inhaled bronchodilator treatments using both the Pediatric Asthma Severity Score (PASS) and asthma severity score. We collected genomic DNA and examined whether 21 candidate SNPs from a review of the literature were associated with BDR using crude odds ratios (OR) and adjusted analysis. RESULTS The final sample population was 53 children, with an average age of 7.2 years. The average initial PASS score (scale of ascending severity from 0 to 6) was 2.5. After adjusting for BMI, age category, gender and smoke exposure, rs912142 was associated with decreased odds of having low BDR (OR, 0.20; 95% confidence interval (CI), 0.02-0.92), and rs7081864 and rs7903366 were associated with decreased odds of having high BDR (OR, 0.097; 95% CI, 0.009-0.62). CONCLUSIONS We found three SNPs significantly associated with pediatric African American BDR that provide information regarding a child's potential response to emergency asthma exacerbation treatment. Once validated in larger studies, such information could guide pharmacogenomic evidence-based emergency asthma treatment to improve patient outcomes.
Collapse
Affiliation(s)
- Jennifer N Fishe
- Department of Emergency Medicine, Division of Research, University of Florida College of Medicine - Jacksonville
- Center for Data Solutions, University of Florida College of Medicine - Jacksonville
| | - Guillaume Labilloy
- Center for Data Solutions, University of Florida College of Medicine - Jacksonville
| | - Rebecca Higley
- Department of Emergency Medicine, Division of Research, University of Florida College of Medicine - Jacksonville
| | - Deirdre Casey
- University of Florida Health Jacksonville, Jacksonville
| | - Amber Ginn
- Department of Pathology, University of Florida College of Medicine - Jacksonville
| | - Brett Baskovich
- Department of Pathology, University of Florida College of Medicine - Jacksonville
| | - Kathryn V Blake
- Nemours Center for Pharmacogenomics and Translational Research, Jacksonville, Florida, USA
| |
Collapse
|
14
|
Sordillo JE, Lutz SM, Jorgenson E, Iribarren C, McGeachie M, Dahlin A, Tantisira K, Kelly R, Lasky-Su J, Sakornsakolpat P, Moll M, Cho MH, Wu AC. A polygenic risk score for asthma in a large racially diverse population. Clin Exp Allergy 2021; 51:1410-1420. [PMID: 34459047 DOI: 10.1111/cea.14007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/02/2021] [Accepted: 08/27/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Polygenic risk scores (PRSs) will have important utility for asthma and other chronic diseases as a tool for predicting disease incidence and subphenotypes. OBJECTIVE We utilized findings from a large multiancestry GWAS of asthma to compute a PRS for asthma with relevance for racially diverse populations. METHODS We derived two PRSs for asthma using a standard approach (based on genome-wide significant variants) and a lasso sum regression approach (allowing all genetic variants to potentially contribute). We used data from the racially diverse Kaiser Permanente GERA cohort (68 638 non-Hispanic Whites, 5874 Hispanics, 6870 Asians and 2760 Blacks). Race was self-reported by questionnaire. RESULTS For the standard PRS, non-Hispanic Whites showed the highest odds ratio for a standard deviation increase in PRS for asthma (OR = 1.16 (95% CI 1.14-1.18)). The standard PRS was also associated with asthma in Hispanic (OR = 1.12 (95% CI 1.05-1.19)) and Asian (OR = 1.10 (95% CI 1.04-1.17)) subjects, with a trend towards increased risk in Blacks (OR = 1.05 (95% CI 0.97-1.15)). We detected an interaction by sex, with men showing a higher risk of asthma with an increase in PRS as compared to women. The lasso sum regression-derived PRS showed stronger associations with asthma in non-Hispanic White subjects (OR = 1.20 (95% CI 1.18-1.23)), Hispanics (OR = 1.17 (95% 1.10-1.26)), Asians (OR = 1.18 (95% CI 1.10-1.27)) and Blacks (OR = 1.10 (95% CI 0.99-1.22)). CONCLUSION Polygenic risk scores across multiple racial/ethnic groups were associated with increased asthma risk, suggesting that PRSs have potential as a tool for predicting disease development.
Collapse
Affiliation(s)
- Joanne E Sordillo
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Massachusetts, USA
| | - Sharon M Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Massachusetts, USA
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Amber Dahlin
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rachel Kelly
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Phuwanat Sakornsakolpat
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew Moll
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ann Chen Wu
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Methods and implementation of a pediatric asthma pharmacogenomic study in the emergency department setting. Pharmacogenet Genomics 2021; 30:201-207. [PMID: 33017130 DOI: 10.1097/fpc.0000000000000414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The emergency department (ED) is a challenging setting to conduct pharmacogenomic studies and integrate that data into fast-paced and potentially life-saving treatment decisions. Therefore, our objective is to present the methods and feasibility of a pilot pharmacogenomic study set in the ED that measured pediatric bronchodilator response (BDR) during acute asthma exacerbations. METHODS This is an exploratory pilot study that collected buccal swabs for DNA and measured BDR during ED encounters for pediatric asthma exacerbations. We evaluated the study's feasibility with a qualitative analysis of ED provider surveys and quantitatively by the proportion of eligible patients enrolled. RESULTS We enrolled 59 out of 90 patients (65%) that were identified and considered eligible during a 5-month period (target enrollment 60 patients over 12 months). The median patient age was 7 years (interquartile range 4-9 years), 61% (N = 36) were male, and 92% (N = 54) were African American. Quality DNA collection was successful for all 59 patients. The ED provider survey response rate was 100%. Most ED providers reported that the study did not impact their workflow (98% of physicians, 88% of nurses, and 90% of respiratory therapists). ED providers did report difficulties with spirometry in the younger age group. CONCLUSIONS Pharmacogenomic studies can be conducted in the ED setting, and enroll a younger patient population with a high proportion of minority participants. By disseminating this study's methods and feasibility analysis, we aim to increase interest in pharmacogenomic studies set in the ED and aimed toward future ED-based pharmacogenomic decision-making.
Collapse
|
16
|
Contreras MG, Keys K, Magaña J, Goddard PC, Risse-Adams O, Zeiger AM, Mak AC, Samedy-Bates LA, Neophytou AM, Lee E, Thakur N, Elhawary JR, Hu D, Huntsman S, Eng C, Hu T, Burchard EG, White MJ. Native American Ancestry and Air Pollution Interact to Impact Bronchodilator Response in Puerto Rican Children with Asthma. Ethn Dis 2021; 31:77-88. [PMID: 33519158 PMCID: PMC7843041 DOI: 10.18865/ed.31.1.77] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective Asthma is the most common chronic disease in children. Short-acting bronchodilator medications are the most commonly prescribed asthma treatment worldwide, regardless of disease severity. Puerto Rican children display the highest asthma morbidity and mortality of any US population. Alarmingly, Puerto Rican children with asthma display poor bronchodilator drug response (BDR). Reduced BDR may explain, in part, the increased asthma morbidity and mortality observed in Puerto Rican children with asthma. Gene-environment interactions may explain a portion of the heritability of BDR. We aimed to identify gene-environment interactions associated with BDR in Puerto Rican children with asthma. Setting Genetic, environmental, and psycho-social data from the Genes-environments and Admixture in Latino Americans (GALA II) case-control study. Participants Our discovery dataset consisted of 658 Puerto Rican children with asthma; our replication dataset consisted of 514 Mexican American children with asthma. Main Outcome Measures We assessed the association of pairwise interaction models with BDR using ViSEN (Visualization of Statistical Epistasis Networks). Results We identified a non-linear interaction between Native American genetic ancestry and air pollution significantly associated with BDR in Puerto Rican children with asthma. This interaction was robust to adjustment for age and sex but was not significantly associated with BDR in our replication population. Conclusions Decreased Native American ancestry coupled with increased air pollution exposure was associated with increased BDR in Puerto Rican children with asthma. Our study acknowledges BDR's phenotypic complexity, and emphasizes the importance of integrating social, environmental, and biological data to further our understanding of complex disease.
Collapse
Affiliation(s)
- María G. Contreras
- Department of Medicine, University of California, San Francisco, CA
- SF BUILD, San Francisco State University, San Francisco, CA
- MARC, San Francisco State University, San Francisco, CA
| | - Kevin Keys
- Department of Medicine, University of California, San Francisco, CA
| | - Joaquin Magaña
- Department of Medicine, University of California, San Francisco, CA
| | - Pagé C. Goddard
- Department of Medicine, University of California, San Francisco, CA
| | - Oona Risse-Adams
- Department of Medicine, University of California, San Francisco, CA
- Lowell Science Research Program, Lowell High School, San Francisco, CA
| | - Andrew M. Zeiger
- Department of Medicine, University of California, San Francisco, CA
- Department of Biology, University of Washington, Seattle, WA
| | - Angel C.Y. Mak
- Department of Medicine, University of California, San Francisco, CA
| | - Lesly-Anne Samedy-Bates
- Department of Medicine, University of California, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA
| | - Andreas M. Neophytou
- Environmental Health Sciences Division, Berkeley School of Public Health, Berkeley, CA
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO
| | - Eunice Lee
- National Institute of Environmental Health Sciences, Cary NC
| | - Neeta Thakur
- Department of Medicine, University of California, San Francisco, CA
| | | | - Donglei Hu
- Department of Medicine, University of California, San Francisco, CA
| | - Scott Huntsman
- Department of Medicine, University of California, San Francisco, CA
| | - Celeste Eng
- Department of Medicine, University of California, San Francisco, CA
| | - Ting Hu
- School of Computing, Queen’s University, Kingston, ON, Canada
| | - Esteban G. Burchard
- Department of Medicine, University of California, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA
| | | |
Collapse
|
17
|
Voorhies K, Sordillo JE, McGeachie M, Ampleford E, Wang AL, Lasky-Su J, Tantisira K, Dahlin A, Kelly RS, Ortega VE, Lutz SM, Wu AC. Age by Single Nucleotide Polymorphism Interactions on Bronchodilator Response in Asthmatics. J Pers Med 2021; 11:jpm11010059. [PMID: 33477890 PMCID: PMC7833432 DOI: 10.3390/jpm11010059] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
An unaddressed and important issue is the role age plays in modulating response to short acting β2-agonists in individuals with asthma. The objective of this study was to identify whether age modifies genetic associations of single nucleotide polymorphisms (SNPs) with bronchodilator response (BDR) to β2-agonists. Using three cohorts with a total of 892 subjects, we ran a genome wide interaction study (GWIS) for each cohort to examine SNP by age interactions with BDR. A fixed effect meta-analysis was used to combine the results. In order to determine if previously identified BDR SNPs had an age interaction, we also examined 16 polymorphisms in candidate genes from two published genome wide association studies (GWAS) of BDR. There were no significant SNP by age interactions on BDR using the genome wide significance level of 5 × 10−8. Using a suggestive significance level of 5 × 10−6, three interactions, including one for a SNP within PRAG1 (rs4840337), were significant and replicated at the significance level of 0.05. Considering candidate genes from two previous GWAS of BDR, three SNPs (rs10476900 (near ADRB2) [p-value = 0.009], rs10827492 (CREM) [p-value = 0.02], and rs72646209 (NCOA3) [p-value = 0.02]) had a marginally significant interaction with age on BDR (p < 0.05). Our results suggest age may be an important modifier of genetic associations for BDR in asthma.
Collapse
Affiliation(s)
- Kirsten Voorhies
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215, USA; (K.V.); (J.E.S.); (S.M.L.)
| | - Joanne E. Sordillo
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215, USA; (K.V.); (J.E.S.); (S.M.L.)
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
| | - Elizabeth Ampleford
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (E.A.); (V.E.O.)
| | - Alberta L. Wang
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
| | - Kelan Tantisira
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
- Division of Pediatric Respiratory Medicine, Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA
| | - Amber Dahlin
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA; (M.M.); (A.L.W.); (J.L.-S.); (K.T.); (A.D.); (R.S.K.)
| | - Victor E. Ortega
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA; (E.A.); (V.E.O.)
| | - Sharon M. Lutz
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215, USA; (K.V.); (J.E.S.); (S.M.L.)
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ann C. Wu
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, MA 02215, USA; (K.V.); (J.E.S.); (S.M.L.)
- Division of General Pediatrics, Department of Pediatrics, Children’s Hospital, Boston, MA 02215, USA
- Correspondence: ; Tel.: +1-(617)-867-4823; Fax: +1-(617)-867-4276
| |
Collapse
|
18
|
Magaña J, Contreras MG, Keys KL, Risse-Adams O, Goddard PC, Zeiger AM, Mak ACY, Elhawary JR, Samedy-Bates LA, Lee E, Thakur N, Hu D, Eng C, Salazar S, Huntsman S, Hu T, Burchard EG, White MJ. An epistatic interaction between pre-natal smoke exposure and socioeconomic status has a significant impact on bronchodilator drug response in African American youth with asthma. BioData Min 2020; 13:7. [PMID: 32636926 PMCID: PMC7333373 DOI: 10.1186/s13040-020-00218-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/23/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Asthma is one of the leading chronic illnesses among children in the United States. Asthma prevalence is higher among African Americans (11.2%) compared to European Americans (7.7%). Bronchodilator medications are part of the first-line therapy, and the rescue medication, for acute asthma symptoms. Bronchodilator drug response (BDR) varies substantially among different racial/ethnic groups. Asthma prevalence in African Americans is only 3.5% higher than that of European Americans, however, asthma mortality among African Americans is four times that of European Americans; variation in BDR may play an important role in explaining this health disparity. To improve our understanding of disparate health outcomes in complex phenotypes such as BDR, it is important to consider interactions between environmental and biological variables. RESULTS We evaluated the impact of pairwise and three-variable interactions between environmental, social, and biological variables on BDR in 233 African American youth with asthma using Visualization of Statistical Epistasis Networks (ViSEN). ViSEN is a non-parametric entropy-based approach able to quantify interaction effects using an information-theory metric known as Information Gain (IG). We performed analyses in the full dataset and in sex-stratified subsets. Our analyses identified several interaction models significantly, and suggestively, associated with BDR. The strongest interaction significantly associated with BDR was a pairwise interaction between pre-natal smoke exposure and socioeconomic status (full dataset IG: 2.78%, p = 0.001; female IG: 7.27%, p = 0.004)). Sex-stratified analyses yielded divergent results for females and males, indicating the presence of sex-specific effects. CONCLUSIONS Our study identified novel interaction effects significantly, and suggestively, associated with BDR in African American children with asthma. Notably, we found that all of the interactions identified by ViSEN were "pure" interaction effects, in that they were not the result of strong main effects on BDR, highlighting the complexity of the network of biological and environmental factors impacting this phenotype. Several associations uncovered by ViSEN would not have been detected using regression-based methods, thus emphasizing the importance of employing statistical methods optimized to detect both additive and non-additive interaction effects when studying complex phenotypes such as BDR. The information gained in this study increases our understanding and appreciation of the complex nature of the interactions between environmental and health-related factors that influence BDR and will be invaluable to biomedical researchers designing future studies.
Collapse
Affiliation(s)
- J. Magaña
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - M. G. Contreras
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Department of Biology, San Francisco State University, San Francisco, CA USA
| | - K. L. Keys
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Berkeley Institute for Data Science, University of California, Berkeley, CA USA
| | - O. Risse-Adams
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Lowell Science Research Program, Lowell High School, San Francisco, CA USA
- Department of Biology, University of California, Santa Cruz, CA USA
| | - P. C. Goddard
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Department of Genetics, Stanford University, Stanford, CA USA
| | - A. M. Zeiger
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA
| | - A. C. Y. Mak
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - J. R. Elhawary
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - L. A. Samedy-Bates
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA USA
| | - E. Lee
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - N. Thakur
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - D. Hu
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - C. Eng
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - S. Salazar
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - S. Huntsman
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| | - T. Hu
- School of Computing, Queen’s University, Kingston, ON Canada
| | - E. G. Burchard
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA USA
| | - M. J. White
- Department of Medicine, University of California, 1550 4th Street, UCSF Rock Hall, Box 2911, San Francisco, CA 94158 USA
| |
Collapse
|
19
|
Early origins of allergy and asthma (ARIES): study protocol for a prospective prenatal birth cohort in Chile. BMC Pediatr 2020; 20:164. [PMID: 32293348 PMCID: PMC7158062 DOI: 10.1186/s12887-020-02077-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Growing evidence shows that atopic dermatitis (AD), food allergy (FA), allergic rhinitis, and asthma are largely determined during the first 1000 days (time elapsed from conception to the 2nd birthday). The ARIES birth cohort aims to determine prenatal and perinatal conditions, as well as genetic and epigenetic factors, that participate in the early setting of immune responses, and the role of these in the later determination of the risk of allergic diseases and asthma in the offspring. Methods We have designed a birth cohort of 250 families with prenatal recruitment (~ 14 weeks). We will genotype relevant allergy/asthma-associated variants in trios and will perform immunophenotyping and evaluation of allergy biomarkers in cord blood. At 1 and 2 years of age we will assess if infants have developed allergic sensitization, AD, FA, as well as biomarkers of asthma including the asthma predictive index. We will also evaluate how maternal conditions modify immune programming through epigenetic modifications and will then depict newborn epigenetic cues of allergy/asthma risk. Next, we will assess composition/diversity of maternal gut, placenta, breastmilk and infant gut microbiome and their association with immunophenotype and biomarkers at birth, and clinical outcomes at age 1 and 2. Finally, we plan to assess how environmental exposures (perinatal outdoor and indoor pollution, allergens and endotoxin) affect the incidence of allergic sensitization, AD, FA, and risk of asthma. Discussion The in-depth study of the ARIES birth cohort shall provide crucial information to understand the rising incidence of allergies and asthma in developing countries, and hopefully provide cues on how to prevent and treat these diseases. Trial registration clinicaltrials.gov NCT04186949, retrospectively registered on December 5, 2019.
Collapse
|
20
|
Zhang E, Levin AM, Williams LK. How does race and ethnicity effect the precision treatment of asthma? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019; 4:337-356. [PMID: 33015363 DOI: 10.1080/23808993.2019.1690396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction Asthma is a common condition that affects large numbers of children and adults, yet the burden of disease is not equally distributed amongst groups. In the United States, African Americans and Puerto Ricans have higher rates of asthma and its complications when compared with European Americans. However, clinical trials and genetic studies have largely focused on the latter group. Areas covered Here we examine what is known regarding differences in asthma treatment response by race-ethnicity. We also review existing genetic studies related to the use of asthma medications, paying special attention to studies that included substantial numbers of non-white population groups. Publicly accessible search engines of the medical literature were queried using combinations of the terms asthma, race, ethnicity, pharmacogenomics, and pharmacogenetics, as well as the names of individual asthma medication classes. The list of articles reviewed was supplemented by bibliographies and expert knowledge. Expert opinion A substantial and coordinated effort is still needed to both identify and validate genetic biomarkers of asthma medication response, as currently there are no clinically actionable genetic markers available for this purpose. The path to identifying such markers in non-white populations is even more formidable, since these groups are underrepresented in existing data.
Collapse
Affiliation(s)
- Ellen Zhang
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Internal Medicine, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
21
|
Ivanova O, Richards LB, Vijverberg SJ, Neerincx AH, Sinha A, Sterk PJ, Maitland‐van der Zee AH. What did we learn from multiple omics studies in asthma? Allergy 2019; 74:2129-2145. [PMID: 31004501 DOI: 10.1111/all.13833] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/25/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022]
Abstract
More than a decade has passed since the finalization of the Human Genome Project. Omics technologies made a huge leap from trendy and very expensive to routinely executed and relatively cheap assays. Simultaneously, we understood that omics is not a panacea for every problem in the area of human health and personalized medicine. Whilst in some areas of research omics showed immediate results, in other fields, including asthma, it only allowed us to identify the incredibly complicated molecular processes. Along with their possibilities, omics technologies also bring many issues connected to sample collection, analyses and interpretation. It is often impossible to separate the intrinsic imperfection of omics from asthma heterogeneity. Still, many insights and directions from applied omics were acquired-presumable phenotypic clusters of patients, plausible biomarkers and potential pathways involved. Omics technologies develop rapidly, bringing improvements also to asthma research. These improvements, together with our growing understanding of asthma subphenotypes and underlying cellular processes, will likely play a role in asthma management strategies.
Collapse
Affiliation(s)
- Olga Ivanova
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Levi B. Richards
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Susanne J. Vijverberg
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Anne H. Neerincx
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Anirban Sinha
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Peter J. Sterk
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
| | - Anke H. Maitland‐van der Zee
- Department of Respiratory Medicine, Amsterdam University Medical Centres (AUMC) University of Amsterdam Amsterdam the Netherlands
- Department of Paediatric Pulmonology Amsterdam UMC/ Emma Children's Hospital Amsterdam the Netherlands
| |
Collapse
|
22
|
Lorenzo-Salazar JM, Flores C. Assessing Asthma Medication Responses in U.S. Minority Children by Whole-Genome Sequencing. Am J Respir Crit Care Med 2019; 197:1513-1514. [PMID: 29578752 DOI: 10.1164/rccm.201803-0457ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | - Carlos Flores
- 1 Instituto Tecnológico y de Energías Renovables Tenerife, Spain.,2 CIBER de Enfermedades Respiratorias Instituto de Salud Carlos III Madrid, Spain and.,3 Research Unit Hospital Universitario Nuestra Señora de Candelaria Tenerife, Spain
| |
Collapse
|
23
|
Cardenas A, Sordillo JE, Rifas-Shiman SL, Chung W, Liang L, Coull BA, Hivert MF, Lai PS, Forno E, Celedón JC, Litonjua AA, Brennan KJ, DeMeo DL, Baccarelli AA, Oken E, Gold DR. The nasal methylome as a biomarker of asthma and airway inflammation in children. Nat Commun 2019; 10:3095. [PMID: 31300640 PMCID: PMC6625976 DOI: 10.1038/s41467-019-11058-3] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 06/19/2019] [Indexed: 12/16/2022] Open
Abstract
The nasal cellular epigenome may serve as biomarker of airway disease and environmental response. Here we collect nasal swabs from the anterior nares of 547 children (mean-age 12.9 y), and measure DNA methylation (DNAm) with the Infinium MethylationEPIC BeadChip. We perform nasal Epigenome-Wide Association analyses (EWAS) of current asthma, allergen sensitization, allergic rhinitis, fractional exhaled nitric oxide (FeNO) and lung function. We find multiple differentially methylated CpGs (FDR < 0.05) and Regions (DMRs; ≥ 5-CpGs and FDR < 0.05) for asthma (285-CpGs), FeNO (8,372-CpGs; 191-DMRs), total IgE (3-CpGs; 3-DMRs), environment IgE (17-CpGs; 4-DMRs), allergic asthma (1,235-CpGs; 7-DMRs) and bronchodilator response (130-CpGs). Discovered DMRs annotated to genes implicated in allergic asthma, Th2 activation and eosinophilia (EPX, IL4, IL13) and genes previously associated with asthma and IgE in EWAS of blood (ACOT7, SLC25A25). Asthma, IgE and FeNO were associated with nasal epigenetic age acceleration. The nasal epigenome is a sensitive biomarker of asthma, allergy and airway inflammation. Epigenetic differences in nasal epithelium have been proposed as a biomarker for lower airway disease and asthma. Here, in epigenome-wide association studies for asthma and other airway traits using nasal swabs, the authors identify differentially methylated CpGs that highlight genes involved in TH2 response.
Collapse
Affiliation(s)
- Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA, 94720, USA. .,Department of Population Medicine, Division of Chronic Disease Research Across the Lifecourse, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA.
| | - Joanne E Sordillo
- Department of Population Medicine, Division of Chronic Disease Research Across the Lifecourse, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA
| | - Sheryl L Rifas-Shiman
- Department of Population Medicine, Division of Chronic Disease Research Across the Lifecourse, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA
| | - Wonil Chung
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Liming Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Marie-France Hivert
- Department of Population Medicine, Division of Chronic Disease Research Across the Lifecourse, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA.,Diabetes Unit, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Peggy S Lai
- Massachusetts General Hospital, Pulmonary/Critical Care, Boston, MA, 02114, USA
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Kasey J Brennan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Dawn L DeMeo
- Department of Medicine, Brigham and Women's Hospital, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, 10032, USA
| | - Emily Oken
- Department of Population Medicine, Division of Chronic Disease Research Across the Lifecourse, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, 02215, USA
| | - Diane R Gold
- Department of Medicine, Brigham and Women's Hospital, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, 02115, USA.,Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| |
Collapse
|
24
|
Na W, Rui-Rui Y, Ming-Kai C, Bin Z, Qing-Li B, Feng-Yun D, Feng-Rong L, Hai-Xin D, Hai-Hua W, Guang-Tao L, Cheng-Qiang J, Yan-Xia J. Establishment of a fluorescent PCR melting curve method for detecting asthma susceptibility using gene SNP typing. J Asthma 2019; 57:850-857. [PMID: 31082286 DOI: 10.1080/02770903.2019.1619084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: To develop a detection method for single nucleotide polymorphisms (SNPs) of bronchial asthma (BA) susceptibility genes (IL-13, IL-33, and GSDMA) based on fluorescence PCR melting curves.Methods: Peripheral blood samples from 33 patients with BA were collected. DNA was extracted, and positive plasmids were constructed. Probes and primers for fluorescence polymerase chain reaction (PCR) were designed according to IL-13, IL-33, and GSDMA sequences, and the SNPs were separately detected by gene sequencing and fluorescence PCR melting curve.Results: The system was successfully divided into 3 SNPs, including IL-13, IL-33, and GSDMA, and a comparison of sequencing methods showed that the results were completely consistent. The lowest detection limit was 1 ng/reaction, the sensitivity and specificity were 100%, and this method had high repeatability (CV = 2.8%).Conclusion: The fluorescence PCR melting curve method is suitable for the rapid and accurate classification of SNPs. The method is economical, simple, and efficient, and is suitable for the screening of the susceptible gene SNPs in a large-scale population of patients with BA.
Collapse
Affiliation(s)
- Wang Na
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Yang Rui-Rui
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, PR China
| | - Chen Ming-Kai
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Zhang Bin
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Bie Qing-Li
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Dong Feng-Yun
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Liang Feng-Rong
- Department of Physical Examination, Medical Examination Center, Jining First People's Hospital, Jining City, Shandong Province, PR China
| | - Dong Hai-Xin
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Wang Hai-Hua
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Li Guang-Tao
- Department of Respiratory Medicine, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China
| | - Jin Cheng-Qiang
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical University, Jining City, Shandong Province, PR China.,Department of Clinical Laboratory Teaching and Research, College of Forensic Medicine and Medical Laboratory, Jining Medical University, Jining, Shandong Province, PR China
| | - Jia Yan-Xia
- Radiology Department, Jining First People's Hospital, Jining City, Shandong Province, PR China.,Department of Medical Imaging Teaching and Research, Basic Medical College, Jining Medical University, Jining City, Shandong Province, PR China
| |
Collapse
|
25
|
Rashkin SR, Chua KC, Ho C, Mulkey F, Jiang C, Mushiroda T, Kubo M, Friedman PN, Rugo HS, McLeod HL, Ratain MJ, Castillos F, Naughton M, Overmoyer B, Toppmeyer D, Witte JS, Owzar K, Kroetz DL. A Pharmacogenetic Prediction Model of Progression-Free Survival in Breast Cancer using Genome-Wide Genotyping Data from CALGB 40502 (Alliance). Clin Pharmacol Ther 2018; 105:738-745. [PMID: 30260474 DOI: 10.1002/cpt.1241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/30/2018] [Indexed: 01/10/2023]
Abstract
Genome-wide genotyping data are increasingly available for pharmacogenetic association studies, but application of these data for development of prediction models is limited. Prediction methods, such as elastic net regularization, have recently been applied to genetic studies but only limitedly to pharmacogenetic outcomes. An elastic net was applied to a pharmacogenetic study of progression-free survival (PFS) of 468 patients with advanced breast cancer in a clinical trial of paclitaxel, nab-paclitaxel, and ixabepilone. A final model included 13 single nucleotide polymorphisms (SNPs) in addition to clinical covariates (prior taxane status, hormone receptor status, disease-free interval, and presence of visceral metastases) with an area under the curve (AUC) integrated over time of 0.81, an increase compared to an AUC of 0.64 for a model with clinical covariates alone. This model may be of value in predicting PFS with microtubule targeting agents and may inform reverse translational studies to understand differential response to these drugs.
Collapse
Affiliation(s)
- Sara R Rashkin
- Department of Biostatistics and Epidemiology, University of California San Francisco, San Francisco, California, USA
| | - Katherina C Chua
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Carol Ho
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Flora Mulkey
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina, USA
| | - Chen Jiang
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina, USA
| | - Tasei Mushiroda
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Paula N Friedman
- Department of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hope S Rugo
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center, Tampa, Florida, USA
| | - Mark J Ratain
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | | - Michael Naughton
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Beth Overmoyer
- Dana-Farber/Partners Cancer Care, Boston, Massachusetts, USA
| | - Deborah Toppmeyer
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - John S Witte
- Department of Biostatistics and Epidemiology, University of California San Francisco, San Francisco, California, USA
| | - Kouros Owzar
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina, USA.,Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
26
|
Mak ACY, White MJ, Eckalbar WL, Szpiech ZA, Oh SS, Pino-Yanes M, Hu D, Goddard P, Huntsman S, Galanter J, Wu AC, Himes BE, Germer S, Vogel JM, Bunting KL, Eng C, Salazar S, Keys KL, Liberto J, Nuckton TJ, Nguyen TA, Torgerson DG, Kwok PY, Levin AM, Celedón JC, Forno E, Hakonarson H, Sleiman PM, Dahlin A, Tantisira KG, Weiss ST, Serebrisky D, Brigino-Buenaventura E, Farber HJ, Meade K, Lenoir MA, Avila PC, Sen S, Thyne SM, Rodriguez-Cintron W, Winkler CA, Moreno-Estrada A, Sandoval K, Rodriguez-Santana JR, Kumar R, Williams LK, Ahituv N, Ziv E, Seibold MA, Darnell RB, Zaitlen N, Hernandez RD. Whole-Genome Sequencing of Pharmacogenetic Drug Response in Racially Diverse Children with Asthma. Am J Respir Crit Care Med 2018; 197:1552-1564. [PMID: 29509491 PMCID: PMC6006403 DOI: 10.1164/rccm.201712-2529oc] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/05/2018] [Indexed: 12/25/2022] Open
Abstract
RATIONALE Albuterol, a bronchodilator medication, is the first-line therapy for asthma worldwide. There are significant racial/ethnic differences in albuterol drug response. OBJECTIVES To identify genetic variants important for bronchodilator drug response (BDR) in racially diverse children. METHODS We performed the first whole-genome sequencing pharmacogenetics study from 1,441 children with asthma from the tails of the BDR distribution to identify genetic association with BDR. MEASUREMENTS AND MAIN RESULTS We identified population-specific and shared genetic variants associated with BDR, including genome-wide significant (P < 3.53 × 10-7) and suggestive (P < 7.06 × 10-6) loci near genes previously associated with lung capacity (DNAH5), immunity (NFKB1 and PLCB1), and β-adrenergic signaling (ADAMTS3 and COX18). Functional analyses of the BDR-associated SNP in NFKB1 revealed potential regulatory function in bronchial smooth muscle cells. The SNP is also an expression quantitative trait locus for a neighboring gene, SLC39A8. The lack of other asthma study populations with BDR and whole-genome sequencing data on minority children makes it impossible to perform replication of our rare variant associations. Minority underrepresentation also poses significant challenges to identify age-matched and population-matched cohorts of sufficient sample size for replication of our common variant findings. CONCLUSIONS The lack of minority data, despite a collaboration of eight universities and 13 individual laboratories, highlights the urgent need for a dedicated national effort to prioritize diversity in research. Our study expands the understanding of pharmacogenetic analyses in racially/ethnically diverse populations and advances the foundation for precision medicine in at-risk and understudied minority populations.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Pino-Yanes
- Research Unit, Hospital Universitario N. S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | - Ann Chen Wu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Precision Medicine Translational Research (PRoMoTeR) Center, Department of Population Medicine, Harvard Medical School and Pilgrim Health Care Institute, Boston, Massachusetts
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics and
| | | | | | | | | | | | | | | | | | | | | | - Pui-Yan Kwok
- Cardiovascular Research Institute
- Institute for Human Genetics, and
| | | | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine, Allergy and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, Allergy and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hakon Hakonarson
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Applied Genomics, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Patrick M. Sleiman
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Applied Genomics, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Amber Dahlin
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kelan G. Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Denise Serebrisky
- Pediatric Pulmonary Division, Jacobi Medical Center, Bronx, New York
| | | | - Harold J. Farber
- Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas
| | - Kelley Meade
- Children’s Hospital and Research Center, Oakland, California
| | | | - Pedro C. Avila
- Department of Medicine, Northwestern University, Chicago, Illinois
| | | | - Shannon M. Thyne
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | | | - Cheryl A. Winkler
- Basic Science Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Frederick National Laboratory, Frederick, Maryland
| | - Andrés Moreno-Estrada
- National Laboratory of Genomics for Biodiversity (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato, Mexico
| | - Karla Sandoval
- National Laboratory of Genomics for Biodiversity (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato, Mexico
| | | | - Rajesh Kumar
- Division of Allergy and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - L. Keoki Williams
- Department of Internal Medicine, and
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Michigan
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences
- Institute for Human Genetics, and
| | | | - Max A. Seibold
- Center for Genes, Environment and Health, Department of Pediatrics, National Jewish Health, Denver, Colorado; and
| | - Robert B. Darnell
- New York Genome Center, New York, New York
- Laboratory of Molecular Neuro-Oncology and
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York
| | | | - Ryan D. Hernandez
- Department of Bioengineering and Therapeutic Sciences
- Cardiovascular Research Institute
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, California
| | - on behalf of the NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium
- Department of Medicine
- Department of Bioengineering and Therapeutic Sciences
- Department of Pediatrics
- Cardiovascular Research Institute
- Institute for Human Genetics, and
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, California
- Research Unit, Hospital Universitario N. S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Precision Medicine Translational Research (PRoMoTeR) Center, Department of Population Medicine, Harvard Medical School and Pilgrim Health Care Institute, Boston, Massachusetts
- Department of Biostatistics, Epidemiology and Informatics and
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- New York Genome Center, New York, New York
- Department of Public Health Sciences
- Department of Internal Medicine, and
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Michigan
- Division of Pediatric Pulmonary Medicine, Allergy and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Center for Applied Genomics, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
- Pediatric Pulmonary Division, Jacobi Medical Center, Bronx, New York
- Department of Allergy and Immunology, Kaiser Permanente Vallejo Medical Center, Vallejo, California
- Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas
- Children’s Hospital and Research Center, Oakland, California
- Bay Area Pediatrics, Oakland, California
- Department of Medicine, Northwestern University, Chicago, Illinois
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Veterans Caribbean Health Care System, San Juan, Puerto Rico
- Basic Science Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Frederick National Laboratory, Frederick, Maryland
- National Laboratory of Genomics for Biodiversity (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato, Mexico
- Centro de Neumologia Pediatrica, San Juan, Puerto Rico
- Division of Allergy and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
- Center for Genes, Environment and Health, Department of Pediatrics, National Jewish Health, Denver, Colorado; and
- Laboratory of Molecular Neuro-Oncology and
- Howard Hughes Medical Institute, The Rockefeller University, New York, New York
| |
Collapse
|
27
|
Xu CJ, Söderhäll C, Bustamante M, Baïz N, Gruzieva O, Gehring U, Mason D, Chatzi L, Basterrechea M, Llop S, Torrent M, Forastiere F, Fantini MP, Carlsen KCL, Haahtela T, Morin A, Kerkhof M, Merid SK, van Rijkom B, Jankipersadsing SA, Bonder MJ, Ballereau S, Vermeulen CJ, Aguirre-Gamboa R, de Jongste JC, Smit HA, Kumar A, Pershagen G, Guerra S, Garcia-Aymerich J, Greco D, Reinius L, McEachan RRC, Azad R, Hovland V, Mowinckel P, Alenius H, Fyhrquist N, Lemonnier N, Pellet J, Auffray C, van der Vlies P, van Diemen CC, Li Y, Wijmenga C, Netea MG, Moffatt MF, Cookson WOCM, Anto JM, Bousquet J, Laatikainen T, Laprise C, Carlsen KH, Gori D, Porta D, Iñiguez C, Bilbao JR, Kogevinas M, Wright J, Brunekreef B, Kere J, Nawijn MC, Annesi-Maesano I, Sunyer J, Melén E, Koppelman GH. DNA methylation in childhood asthma: an epigenome-wide meta-analysis. THE LANCET RESPIRATORY MEDICINE 2018; 6:379-388. [PMID: 29496485 DOI: 10.1016/s2213-2600(18)30052-3] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/09/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND DNA methylation profiles associated with childhood asthma might provide novel insights into disease pathogenesis. We did an epigenome-wide association study to assess methylation profiles associated with childhood asthma. METHODS We did a large-scale epigenome-wide association study (EWAS) within the Mechanisms of the Development of ALLergy (MeDALL) project. We examined epigenome-wide methylation using Illumina Infinium Human Methylation450 BeadChips (450K) in whole blood in 207 children with asthma and 610 controls at age 4-5 years, and 185 children with asthma and 546 controls at age 8 years using a cross-sectional case-control design. After identification of differentially methylated CpG sites in the discovery analysis, we did a validation study in children (4-16 years; 247 cases and 2949 controls) from six additional European cohorts and meta-analysed the results. We next investigated whether replicated CpG sites in cord blood predict later asthma in 1316 children. We subsequently investigated cell-type-specific methylation of the identified CpG sites in eosinophils and respiratory epithelial cells and their related gene-expression signatures. We studied cell-type specificity of the asthma association of the replicated CpG sites in 455 respiratory epithelial cell samples, collected by nasal brushing of 16-year-old children as well as in DNA isolated from blood eosinophils (16 with asthma, eight controls [age 2-56 years]) and compared this with whole-blood DNA samples of 74 individuals with asthma and 93 controls (age 1-79 years). Whole-blood transcriptional profiles associated with replicated CpG sites were annotated using RNA-seq data of subsets of peripheral blood mononuclear cells sorted by fluorescence-activated cell sorting. FINDINGS 27 methylated CpG sites were identified in the discovery analysis. 14 of these CpG sites were replicated and passed genome-wide significance (p<1·14 × 10-7) after meta-analysis. Consistently lower methylation levels were observed at all associated loci across childhood from age 4 to 16 years in participants with asthma, but not in cord blood at birth. All 14 CpG sites were significantly associated with asthma in the second replication study using whole-blood DNA, and were strongly associated with asthma in purified eosinophils. Whole-blood transcriptional signatures associated with these CpG sites indicated increased activation of eosinophils, effector and memory CD8 T cells and natural killer cells, and reduced number of naive T cells. Five of the 14 CpG sites were associated with asthma in respiratory epithelial cells, indicating cross-tissue epigenetic effects. INTERPRETATION Reduced whole-blood DNA methylation at 14 CpG sites acquired after birth was strongly associated with childhood asthma. These CpG sites and their associated transcriptional profiles indicate activation of eosinophils and cytotoxic T cells in childhood asthma. Our findings merit further investigations of the role of epigenetics in a clinical context. FUNDING EU and the Seventh Framework Programme (the MeDALL project).
Collapse
Affiliation(s)
- Cheng-Jian Xu
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; GRIAC research institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cilla Söderhäll
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Mariona Bustamante
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; Centre for Genomic Regulation, the Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Nour Baïz
- Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Sorbonne Université, INSERM, Pierre Louis Institute of Epidemiology and Public Health, Saint-Antoine Medical School, Paris, France
| | - Olena Gruzieva
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ulrike Gehring
- Institute for Risk Assessment Sciences, Utrecht University, the Netherlands
| | - Dan Mason
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, LA, USA; Department of Social Medicine, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece; Department of Genetics and Cell Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Mikel Basterrechea
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Health Research Institute Biodonostia, San Sebastián, Spain; Public Health Department of Gipuzkoa, San Sebastián, Spain
| | - Sabrina Llop
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | | | | | - Maria Pia Fantini
- Department of Biomedical and Neuromotor sciences, University of Bologna, Bologna, Italy
| | - Karin C Lødrup Carlsen
- Department of Paediatrics, Oslo University Hospital, Oslo, Norway; Department of Paediatric and Adolescent Medicine, University of Oslo, Oslo, Norway
| | - Tari Haahtela
- Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Andréanne Morin
- Department of Human Genetics, McGill University and Genome Quebec, Innovation Centre, Montréal, QC, Canada
| | - Marjan Kerkhof
- GRIAC research institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Simon Kebede Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bianca van Rijkom
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Soesma A Jankipersadsing
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marc Jan Bonder
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Stephane Ballereau
- European Institute for Systems Biology and Medicine, Campus Charles Mérieux - Université de Lyon, CIRI CNRS UMR5308, CNRS-ENS-UCBL-ENS, Lyon, France; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Cornelis J Vermeulen
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; GRIAC research institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Raul Aguirre-Gamboa
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Johan C de Jongste
- Department of Pediatrics, Erasmus MC - Sophia Children's Hospital University Medical Center, Rotterdam, The Netherlands
| | - Henriette A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Ashish Kumar
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stefano Guerra
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Judith Garcia-Aymerich
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Dario Greco
- Faculty of Medicine and Life Sciences and Institute of Biosciences and Medical Technology (BioMediTech), University of Tampere, Tampere, Finland
| | - Lovisa Reinius
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Rosemary R C McEachan
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Raf Azad
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Vegard Hovland
- Department of Paediatrics, Oslo University Hospital, Oslo, Norway
| | - Petter Mowinckel
- Department of Paediatrics, Oslo University Hospital, Oslo, Norway
| | - Harri Alenius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Bacteriology and Immunology, Medicum, University of Helsinki, Helsinki, Finland
| | - Nanna Fyhrquist
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Bacteriology and Immunology, Medicum, University of Helsinki, Helsinki, Finland
| | - Nathanaël Lemonnier
- European Institute for Systems Biology and Medicine, Campus Charles Mérieux - Université de Lyon, CIRI CNRS UMR5308, CNRS-ENS-UCBL-ENS, Lyon, France; Institute for Advanced Biosciences, UGA-INSERM U1209-CNRS UMR5309, Site Santé, Allée des Alpes, La Tronche, France
| | - Johann Pellet
- European Institute for Systems Biology and Medicine, Campus Charles Mérieux - Université de Lyon, CIRI CNRS UMR5308, CNRS-ENS-UCBL-ENS, Lyon, France
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, Campus Charles Mérieux - Université de Lyon, CIRI CNRS UMR5308, CNRS-ENS-UCBL-ENS, Lyon, France
| | | | - Pieter van der Vlies
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Research BV, Metslawier, the Netherlands
| | - Cleo C van Diemen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Yang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Josep M Anto
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jean Bousquet
- University Hospital, Montpellier, France; Department of Dermatology, Charité, Berlin, Germany
| | - Tiina Laatikainen
- National Institute for Health and Welfare, Helsinki, Finland; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Catherine Laprise
- Université du Québec à Chicoutimi, Département des sciences fondamentales, Saguenay, QC, Canada; Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, 305 Saint-Vallier, Saguenay, QC, Canada
| | - Kai-Håkon Carlsen
- Department of Paediatrics, Oslo University Hospital, Oslo, Norway; Department of Paediatric and Adolescent Medicine, University of Oslo, Oslo, Norway
| | - Davide Gori
- Department of Biomedical and Neuromotor sciences, University of Bologna, Bologna, Italy
| | - Daniela Porta
- Department of Epidemiology Lazio Regional Health Service, Rome, Italy
| | - Carmen Iñiguez
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Jose Ramon Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, Biocruces Health Research Institute, CIBERDEM, University of the Basque Country UPV-EHU, Leioa-Bizkaia, Spain
| | - Manolis Kogevinas
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Bert Brunekreef
- Institute for Risk Assessment Sciences, Utrecht University, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Martijn C Nawijn
- GRIAC research institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Isabella Annesi-Maesano
- Epidemiology of Allergic and Respiratory Diseases Department (EPAR), Sorbonne Université, INSERM, Pierre Louis Institute of Epidemiology and Public Health, Saint-Antoine Medical School, Paris, France
| | - Jordi Sunyer
- ISGlobal, Centre for Research in Environmental Epidemiology, the Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; IMIM Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Sachs Children's Hospital, Stockholm, Sweden; Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Gerard H Koppelman
- GRIAC research institute Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
28
|
Vijverberg SJH, Farzan N, Slob EMA, Neerincx AH, Maitland-van der Zee AH. Treatment response heterogeneity in asthma: the role of genetic variation. Expert Rev Respir Med 2017; 12:55-65. [PMID: 29115880 DOI: 10.1080/17476348.2018.1403318] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Asthmatic patients show a large heterogeneity in response to asthma medication. Rapidly evolving genotyping technologies have led to the identification of various genetic variants associated with treatment outcomes. Areas covered: This review focuses on the current knowledge of genetic variants influencing treatment response to the most commonly used asthma medicines: short- and long-acting beta-2 agonists (SABA/LABA), inhaled corticosteroids (ICS) and leukotriene modifiers. This review shows that various genetic variants have been identified, but none are currently used to guide asthma treatment. One of the most promising genetic variants is the Arg16 variant in the ADRB2 gene to guide LABA treatment in asthmatic children. Expert commentary: Poor replication of initially promising results and the low fraction of variability accounted for by single genetic variants inhibit pharmacogenetic findings to reach the asthma clinic. Nevertheless, the identification of genetic variation influencing treatment response does provide more insights in the complex processes underlying response and might identify novel targets for treatment. There is a need to report measures of clinical validity, to perform precision-medicine guided trials, as well as to understand how genetic variation interacts with environmental factors. In addition, systems biology approaches might be able to show a more complete picture of these complex interactions.
Collapse
Affiliation(s)
- Susanne J H Vijverberg
- a Department of Respiratory Medicine, Academic Medical Center (AMC) , University of Amsterdam , Amsterdam , The Netherlands
| | - Niloufar Farzan
- a Department of Respiratory Medicine, Academic Medical Center (AMC) , University of Amsterdam , Amsterdam , The Netherlands
| | - Elise M A Slob
- a Department of Respiratory Medicine, Academic Medical Center (AMC) , University of Amsterdam , Amsterdam , The Netherlands
| | - Anne H Neerincx
- a Department of Respiratory Medicine, Academic Medical Center (AMC) , University of Amsterdam , Amsterdam , The Netherlands
| | - Anke H Maitland-van der Zee
- a Department of Respiratory Medicine, Academic Medical Center (AMC) , University of Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
29
|
Kan M, Shumyatcher M, Himes BE. Using omics approaches to understand pulmonary diseases. Respir Res 2017; 18:149. [PMID: 28774304 PMCID: PMC5543452 DOI: 10.1186/s12931-017-0631-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/26/2017] [Indexed: 12/24/2022] Open
Abstract
Omics approaches are high-throughput unbiased technologies that provide snapshots of various aspects of biological systems and include: 1) genomics, the measure of DNA variation; 2) transcriptomics, the measure of RNA expression; 3) epigenomics, the measure of DNA alterations not involving sequence variation that influence RNA expression; 4) proteomics, the measure of protein expression or its chemical modifications; and 5) metabolomics, the measure of metabolite levels. Our understanding of pulmonary diseases has increased as a result of applying these omics approaches to characterize patients, uncover mechanisms underlying drug responsiveness, and identify effects of environmental exposures and interventions. As more tissue- and cell-specific omics data is analyzed and integrated for diverse patients under various conditions, there will be increased identification of key mechanisms that underlie pulmonary biological processes, disease endotypes, and novel therapeutics that are efficacious in select individuals. We provide a synopsis of how omics approaches have advanced our understanding of asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), idiopathic pulmonary fibrosis (IPF), and pulmonary arterial hypertension (PAH), and we highlight ongoing work that will facilitate pulmonary disease precision medicine.
Collapse
Affiliation(s)
- Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| |
Collapse
|
30
|
White MJ, Risse-Adams O, Goddard P, Contreras MG, Adams J, Hu D, Eng C, Oh SS, Davis A, Meade K, Brigino-Buenaventura E, LeNoir MA, Bibbins-Domingo K, Pino-Yanes M, Burchard EG. Novel genetic risk factors for asthma in African American children: Precision Medicine and the SAGE II Study. Immunogenetics 2016; 68:391-400. [PMID: 27142222 DOI: 10.1007/s00251-016-0914-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/25/2016] [Indexed: 01/06/2023]
Abstract
Asthma, an inflammatory disorder of the airways, is the most common chronic disease of children worldwide. There are significant racial/ethnic disparities in asthma prevalence, morbidity, and mortality among US children. This trend is mirrored in obesity, which may share genetic and environmental risk factors with asthma. The majority of asthma biomedical research has been performed in populations of European decent. We sought to identify genetic risk factors for asthma in African American children. We also assessed the generalizability of genetic variants associated with asthma in European and Asian populations to African American children. Our study population consisted of 1227 (812 asthma cases, 415 controls) African American children with genome-wide single nucleotide polymorphism (SNP) data. Logistic regression was used to identify associations between SNP genotype and asthma status. We identified a novel variant in the PTCHD3 gene that is significantly associated with asthma (rs660498, p = 2.2 × 10(-7)) independent of obesity status. Approximately 5 % of previously reported asthma genetic associations identified in European populations replicated in African Americans. Our identification of novel variants associated with asthma in African American children, coupled with our inability to replicate the majority of findings reported in European Americans, underscores the necessity for including diverse populations in biomedical studies of asthma.
Collapse
Affiliation(s)
- Marquitta J White
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA.
| | - O Risse-Adams
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- Lowell Science Research Program, Lowell High School, San Francisco, CA, USA
| | - P Goddard
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - M G Contreras
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- SF BUILD, San Francisco State University, San Francisco, CA, USA
| | - J Adams
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - D Hu
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - C Eng
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - S S Oh
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - A Davis
- Children's Hospital and Research Center Oakland, Oakland, CA, USA
| | - K Meade
- Children's Hospital and Research Center Oakland, Oakland, CA, USA
| | - E Brigino-Buenaventura
- Department of Allergy and Immunology, Kaiser Permanente Vallejo Medical Center, Vallejo, CA, USA
| | | | - K Bibbins-Domingo
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
| | - M Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - E G Burchard
- Department of Medicine, University of California, San Francisco, UCSF Box 2911, San Francisco, CA, 94143-2911, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
31
|
Costa GNO, Dudbridge F, Fiaccone RL, da Silva TM, Conceição JS, Strina A, Figueiredo CA, Magalhães WCS, Rodrigues MR, Gouveia MH, Kehdy FSG, Horimoto ARVR, Horta B, Burchard EG, Pino-Yanes M, Del Rio Navarro B, Romieu I, Hancock DB, London S, Lima-Costa MF, Pereira AC, Tarazona E, Rodrigues LC, Barreto ML. A genome-wide association study of asthma symptoms in Latin American children. BMC Genet 2015; 16:141. [PMID: 26635092 PMCID: PMC4669662 DOI: 10.1186/s12863-015-0296-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/17/2015] [Indexed: 01/14/2023] Open
Abstract
Background Asthma is a chronic disease of the airways and, despite the advances in the knowledge of associated genetic regions in recent years, their mechanisms have yet to be explored. Several genome-wide association studies have been carried out in recent years, but none of these have involved Latin American populations with a high level of miscegenation, as is seen in the Brazilian population. Methods 1246 children were recruited from a longitudinal cohort study in Salvador, Brazil. Asthma symptoms were identified in accordance with an International Study of Asthma and Allergies in Childhood (ISAAC) questionnaire. Following quality control, 1 877 526 autosomal SNPs were tested for association with childhood asthma symptoms by logistic regression using an additive genetic model. We complemented the analysis with an estimate of the phenotypic variance explained by common genetic variants. Replications were investigated in independent Mexican and US Latino samples. Results Two chromosomal regions reached genome-wide significance level for childhood asthma symptoms: the 14q11 region flanking the DAD1 and OXA1L genes (rs1999071, MAF 0.32, OR 1.78, 95 % CI 1.45–2.18, p-value 2.83 × 10−8) and 15q22 region flanking the FOXB1 gene (rs10519031, MAF 0.04, OR 3.0, 95 % CI 2.02–4.49, p-value 6.68 × 10−8 and rs8029377, MAF 0.03, OR 2.49, 95 % CI 1.76–3.53, p-value 2.45 × 10−7). eQTL analysis suggests that rs1999071 regulates the expression of OXA1L gene. However, the original findings were not replicated in the Mexican or US Latino samples. Conclusions We conclude that the 14q11 and 15q22 regions may be associated with asthma symptoms in childhood. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0296-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gustavo N O Costa
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Frank Dudbridge
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | | | - Thiago M da Silva
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | | | - Agostino Strina
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.
| | - Wagner C S Magalhães
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Maira R Rodrigues
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Mateus H Gouveia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Fernanda S G Kehdy
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | - Bernardo Horta
- Programa de Pós Graduação em Epidemiologia, Universidade Federal de Pelotas, Pelotas, Brazil.
| | | | - Maria Pino-Yanes
- Department of Medicine, University of California, San Francisco, USA.
| | - Blanca Del Rio Navarro
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Dana B Hancock
- Behavioral and Urban Health Program, Research Triangle Institute (RTI) International, Research Triangle Park, North Carolina, USA.
| | - Stephanie London
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Alexandre C Pereira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Eduardo Tarazona
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Mauricio L Barreto
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil. .,Centro de Pesquisa Gonçalo Muniz, Fundação Osvaldo Cruz, Salvador, Brazil.
| |
Collapse
|
32
|
Abstract
Asthma is a chronic disease which causes recurrent breathlessness affecting 300 million people worldwide of whom 250,000 die annually. The epigenome is a set of heritable modifications and tags that affect the genome without changing the intrinsic DNA sequence. These marks include DNA methylation, modifications to histone proteins around which DNA is wrapped and expression of noncoding RNA. Alterations in all of these processes have been reported in patients with asthma. In some cases these differences are linked to disease severity and susceptibility and may account for the limited value of genetic studies in asthma. Animal models of asthma suggest that epigenetic modifications and processes are linked to asthma and may be tractable targets for therapeutic intervention.
Collapse
Affiliation(s)
- Peter O Brook
- Imperial College London, National Heart & Lung Institute, Dovehouse Street, London, SW3 6LY, UK
| | - Mark M Perry
- Imperial College London, National Heart & Lung Institute, Dovehouse Street, London, SW3 6LY, UK
| | - Ian M Adcock
- Imperial College London, National Heart & Lung Institute, Dovehouse Street, London, SW3 6LY, UK
| | - Andrew L Durham
- Imperial College London, National Heart & Lung Institute, Dovehouse Street, London, SW3 6LY, UK
| |
Collapse
|
33
|
Maranville JC, Di Rienzo A. Combining genetic and nongenetic biomarkers to realize the promise of pharmacogenomics for inflammatory diseases. Pharmacogenomics 2015; 15:1931-40. [PMID: 25495413 DOI: 10.2217/pgs.14.129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Many drugs used to treat inflammatory diseases are ineffective in a substantial proportion of patients. Identifying patients that are likely to respond to specific therapies would facilitate personalized treatment strategies that could improve outcomes while reducing costs and risks of adverse events. Despite these clear benefits, there are limited examples of predictive biomarkers of drug efficacy currently implemented into clinical practice for inflammatory diseases. We review efforts to identify genetic and nongenetic biomarkers of drug response in these diseases and consider potential benefits from combining multiple sources of biological data into multifeature predictive models.
Collapse
Affiliation(s)
- Joseph C Maranville
- Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
34
|
Rovaris DL, Mota NR, da Silva BS, Girardi P, Victor MM, Grevet EH, Bau CH, Contini V. Should we keep on? Looking into pharmacogenomics of ADHD in adulthood from a different perspective. Pharmacogenomics 2015; 15:1365-81. [PMID: 25155937 DOI: 10.2217/pgs.14.95] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A considerable proportion of adults with attention-deficit/hyperactivity disorder (ADHD) do not respond to the treatment with methylphenidate. This scenario could be due to inherited interindividual differences that may alter pharmacologic treatment response. In this sense, in 2012 we conducted a systematic search on PUBMED-indexed literature for articles containing information about pharmacogenomics of ADHD in adults. Five studies were found on methylphenidate pharmacogenomics and the only significant association was reported by one particular study. However, this single association with the SLC6A3 gene was not replicated in two subsequent reports. In the present review, although we could not find additional pharmacogenomics studies, we discuss these up-to-date findings and suggest new approaches for this field. Additionally, using systeomic-oriented databases, we provide a broad picture of new possible candidate genes as well as potential gene-gene interactions to be investigated in pharmacogenomics of persistent ADHD.
Collapse
Affiliation(s)
- Diego L Rovaris
- Departament of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul (UFRGS), Brazil
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Chhibber A, Kroetz DL, Tantisira KG, McGeachie M, Cheng C, Plenge R, Stahl E, Sadee W, Ritchie MD, Pendergrass SA. Genomic architecture of pharmacological efficacy and adverse events. Pharmacogenomics 2014; 15:2025-48. [PMID: 25521360 PMCID: PMC4308414 DOI: 10.2217/pgs.14.144] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The pharmacokinetic and pharmacodynamic disciplines address pharmacological traits, including efficacy and adverse events. Pharmacogenomics studies have identified pervasive genetic effects on treatment outcomes, resulting in the development of genetic biomarkers for optimization of drug therapy. Pharmacogenomics-based tests are already being applied in clinical decision making. However, despite substantial progress in identifying the genetic etiology of pharmacological response, current biomarker panels still largely rely on single gene tests with a large portion of the genetic effects remaining to be discovered. Future research must account for the combined effects of multiple genetic variants, incorporate pathway-based approaches, explore gene-gene interactions and nonprotein coding functional genetic variants, extend studies across ancestral populations, and prioritize laboratory characterization of molecular mechanisms. Because genetic factors can play a key role in drug response, accurate biomarker tests capturing the main genetic factors determining treatment outcomes have substantial potential for improving individual clinical care.
Collapse
Affiliation(s)
- Aparna Chhibber
- Department of Bioengineering & Therapeutic Sciences, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA,USA
| | - Deanna L Kroetz
- Department of Bioengineering & Therapeutic Sciences, Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA,USA
| | - Kelan G Tantisira
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Michael McGeachie
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Cheng Cheng
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert Plenge
- Division of Rheumatology, Immunology & Allergy, Division of Genetics, Brigham & Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Eli Stahl
- Department of Genetics & Genomic Sciences, Mount Sinai Hospital, New York, NY, USA
| | - Wolfgang Sadee
- Center for Pharmacogenomics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Marylyn D Ritchie
- Department of Biochemistry & Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16801, USA
| | - Sarah A Pendergrass
- Department of Biochemistry & Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16801, USA
| |
Collapse
|
36
|
Ioannidis JPA. To replicate or not to replicate: the case of pharmacogenetic studies: Have pharmacogenomics failed, or do they just need larger-scale evidence and more replication? ACTA ACUST UNITED AC 2014; 6:413-8; discussion 418. [PMID: 23963161 DOI: 10.1161/circgenetics.113.000106] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- John P A Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
Padhukasahasram B, Yang JJ, Levin AM, Yang M, Burchard EG, Kumar R, Kwok PY, Seibold MA, Lanfear DE, Williams LK. Gene-based association identifies SPATA13-AS1 as a pharmacogenomic predictor of inhaled short-acting beta-agonist response in multiple population groups. THE PHARMACOGENOMICS JOURNAL 2014; 14:365-71. [PMID: 24418963 PMCID: PMC4098013 DOI: 10.1038/tpj.2013.49] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/19/2013] [Accepted: 11/26/2013] [Indexed: 01/04/2023]
Abstract
Inhaled short-acting beta-agonist (SABA) medication is commonly used in asthma patients to rapidly reverse airway obstruction and improve acute symptoms. We performed a genome wide association study of SABA medication response using gene-based association tests. A linear mixed model approach was first used for SNP associations, and results were later combined using GATES to generate gene-based associations. Our results identified SPATA13-AS1 as being significantly associated with SABA bronchodilator response in 328 healthy African Americans. In replication, this gene was associated with SABA response among 2 separate groups of African Americans with asthma (n=1,073, p=0.011 and n=1,968, p=0.014), 149 healthy African Americans (p=0.003), and 556 European Americans with asthma (p=0.041). SPATA13-AS1 was also associated with longitudinal SABA medication usage in 2 separate groups of African Americans with asthma (n=658, p=0.047 and n=1,968, p=0.025). Future studies are needed to delineate the precise mechanism by which SPATA13-AS1 may influence SABA response.
Collapse
Affiliation(s)
- B Padhukasahasram
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA
| | - J J Yang
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - A M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - M Yang
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA
| | - E G Burchard
- 1] Department of Medicine, University of California San Francisco, San Francisco, CA, USA [2] Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - R Kumar
- Department of Pediatrics, The Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - P-Y Kwok
- 1] Department of Dermatology, University of California San Francisco, San Francisco, CA, USA [2] Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - M A Seibold
- 1] Integrated Center for Genes, Environment, and Health, National Jewish Health, Denver, CO, Colorado, USA [2] Department of Pediatrics, National Jewish Health, Denver, CO, USA [3] Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Denver, CO, USA
| | - D E Lanfear
- 1] Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA [2] Department of Medicine, Henry Ford Health System, Detroit, MI, USA
| | - L K Williams
- 1] Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, MI, USA [2] Department of Medicine, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|