Precision Medicine in Childhood Asthma: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by Javier Perez-Garcia.

Asthma is a complex and multifactorial respiratory disease with a high prevalence in the pediatric population. Variation in treatment response to asthma therapies has been described among patients, and difficult-to-treat asthma carries both high healthcare and socioeconomic burden to the patients and society. Omic studies can be used to discover the molecular mechanisms underlying asthma susceptibility and treatment response, contributing to a better knowledge and definition of asthma pathogenesis and therefore, to the development of precision medicine. This entry aims to summarize the recent findings of omic studies of treatment response in childhood asthma. Between 2018-2019 a total of 13 omic studies has been performed involving genomics, epigenomics, transcriptomics, metabolomics, and the microbiome. These have been focused on the response to three common asthma medications: short-acting beta agonists, inhaled corticosteroids, and leukotriene receptor antagonists. Novel associations of different biomarkers with asthma treatment response have been described. However, stronger evidence and more consistent results are required to implement these molecular biomarkers into clinical practice by establishing the most appropriate therapy for each patient.

  • asthma
  • genomics
  • transcriptomics
  • epigenomics
  • metabolomics
  • microbiome
  • omics
  • inhaled corticosteroid
  • short-acting beta agonist
  • leukotriene receptor antagonist
Please wait, diff process is still running!

References

  1. The Global Asthma Report 2018; Global Asthma Network: Auckland, New Zealand, 2018; ISBN 978-0-473-29125-9\r978-0-473-29126-6.
  2. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention; Global Initiative for Asthma: Fontana, WI, USA, 2019.
  3. Guilbert, T.W.; Bacharier, L.B.; Fitzpatrick, A.M; Severe Asthma in Children. J. Allergy Clin. Immunol. Pract. 2014, 2, 489–500.
  4. Carlos Nunes; Ana Margarida Pereira; Mário Morais-Almeida; Asthma costs and social impact. Asthma Research and Practice 2017, 3, 1, 10.1186/s40733-016-0029-3.
  5. Tyler, S.R.; Bunyavanich, S; Leveraging-Omics for Asthma Endotyping. J. Allergy Clin. Immunol. 2019, 144, 13–23.
  6. Peter M. Visscher; Naomi R. Wray; Qian Zhang; Pamela Sklar; Mark I. McCarthy; Matthew A. Brown; Jian Yang; 10 Years of GWAS Discovery: Biology, Function, and Translation. The American Journal of Human Genetics 2017, 101, 5-22, 10.1016/j.ajhg.2017.06.005.
  7. Petersen, B.S.; Fredrich, B.; Hoeppner, M.P.; Ellinghaus, D.; Franke, A; Opportunities and Challenges of Whole-Genome and-Exome Sequencing. BMC Genet 2017, 18, 14.
  8. Esther Herrera-Luis; Natalia Hernandez-Pacheco; Susanne J. Vijverberg; Carlos Flores; Maria Pino-Yanes; Role of genomics in asthma exacerbations. Current Opinion in Pulmonary Medicine 2019, 25, 101-112, 10.1097/mcp.0000000000000533.
  9. García-Menaya, J.M.; Cordobés-Durán, C.; García-Martín, E.; Agúndez, J.A.G; Pharmacogenetic Factors Affecting Asthma Treatment Response. Potential Implications for Drug Therapy. Front. Pharmacol. 2019, 10, 520.
  10. Natalia Hernandez-Pacheco; Carlos Flores; Sam S. Oh; Esteban G. Burchard; Maria Pino-Yanes; What Ancestry Can Tell Us About the Genetic Origins of Inter-Ethnic Differences in Asthma Expression. Current Allergy and Asthma Reports 2016, 16, 53, 10.1007/s11882-016-0635-4.
  11. Mak, A.C.Y.; White, M.J.; Eckalbar, W.L.; Szpiech, Z.A.; Oh, S.S.; Pino-Yanes, M.; Hu, D.; Goddard, P.; Huntsman, S.; Galanter, J.; et al.et al Faculty Opinions recommendation of Whole-Genome Sequencing of Pharmacogenetic Drug Response in Racially Diverse Children with Asthma.. Am. J. Respir. Crit. Care Med. 2018, 197, 1552–1564, 10.3410/f.732806514.793547058.
  12. Spear, M.L.; Hu, D.; Pino-Yanes, M.; Huntsman, S.; Eng, C.; Levin, A.M.; Ortega, V.E.; White, M.J.; McGarry, M.E.; Thakur, N.; et al.et al A Genome-Wide Association and Admixture Mapping Study of Bronchodilator Drug Response in African Americans with Asthma. Pharmacogenomics J. 2019, 19, 249–259.
  13. Hernandez-Pacheco, N.; Farzan, N.; Francis, B.; Karimi, L.; Repnik, K.; Vijverberg, S.J.; Soares, P.; Schieck, M.; Gorenjak, M.; Forno, E.; et al.et al Genome-wide association study of inhaled corticosteroid response in African-admixed children with asthma. Clin. Exp. Allergy 2019, 49, 789–798.
  14. Hiroshi Kanazawa; Kazuto Hirata; Junichi Yoshikawa; Nitrogen oxides reduce albuterol-induced bronchodilation in patients with bronchial asthma.. Respiration 2002, 69, 490-495, 10.1159/000066466.
  15. Fabio L.M. Ricciardolo; Multiple roles of nitric oxide in the airways. Thorax 2003, 58, 175–182.
  16. Ferreira, M.A.R.; Matheson, M.C.; Duffy, D.L.; Marks, G.B.; Hui, J.; Le Souëf, P.; Danoy, P.; Baltic, S.; Nyholt, D.R.; Jenkins, M.; et al.et al Identification of IL6R and Chromosome 11q13.5 as Risk Loci for Asthma. Lancet 2011, 378, 1006–1014.
  17. Kichaev, G.; Bhatia, G.; Loh, P.R.; Gazal, S.; Burch, K.; Freund, M.K.; Schoech, A.; Pasaniuc, B.; Price, A.L; Leveraging Polygenic Functional Enrichment to Improve GWAS Power. Am. J. Hum. Genet. 2019, 104, 65–75.
  18. Lutz, S.M.; Cho, M.H.; Young, K.; Hersh, C.P.; Castaldi, P.J.; McDonald, M.L.; Regan, E.; Mattheisen, M.; DeMeo, D.L.; Parker, M.; et al.et al A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry.. BMC Genetics 2015, 16, 138, 10.1186/s12863-015-0299-4.
  19. Lee, J.H.; McDonald, M.L.; Cho, M.H.; Wan, E.S.; Castaldi, P.J.; Hunninghake, G.M.; Marchetti, N.; Lynch, D.A.; Crapo, J.D.; Lomas, D.A.; et al.et al DNAH5 is associated with total lung capacity in chronic obstructive pulmonary disease.. Respiratory Research 2014, 15, 97, 10.1186/s12931-014-0097-y.
  20. Ramasamy, A.; Curjuric, I.; Coin, L.J.; Kumar, A.; McArdle, W.L.; Imboden, M.; Leynaert, B.; Kogevinas, M.; Schmid-Grendelmeier, P.; Pekkanen, J.; et al.et al A genome-wide meta-analysis of genetic variants associated with allergic rhinitis and grass sensitization and their interaction with birth order. Journal of Allergy and Clinical Immunology 2011, 128, 996-1005, 10.1016/j.jaci.2011.08.030.
  21. Forno, E.; Celedón, J.C. Epigenomics and Transcriptomics in the Prediction and Diagnosis of Childhood Asthma: Are We There Yet? Front. Pediatr. 2019, 7, 115.
  22. Melissa Fazzari; John M. Greally; Introduction to Epigenomics and Epigenome-Wide Analysis. Advanced Structural Safety Studies 2010, 620, 243-265, 10.1007/978-1-60761-580-4_7.
  23. Mohamad M. Kronfol; Mikhail G. Dozmorov; Rong Huang; Patricia W. Slattum; Joseph L. McClay; The role of epigenomics in personalized medicine. Expert Review of Precision Medicine and Drug Development 2017, 2, 33-45, 10.1080/23808993.2017.1284557.
  24. Alberta Wang; Olena Gruzieva; Weiliang Qiu; Simon Kebede Merid; Juan C. Celedón; Benjamin A. Raby; Cilla Söderhäll; Dawn L. DeMeo; Scott T. Weiss; Erik Melén; et al.Kelan G Tantisira DNA methylation is associated with inhaled corticosteroid response in persistent childhood asthmatics. Clinical Experimental Allergy 2019, 49, 1225-1234, 10.1111/cea.13447.
  25. Alberta L. Wang; Weiliang Qiu; Dawn L. DeMeo; Benjamin A. Raby; Scott Weiss; Kelan G. Tantisira; DNA methylation is associated with improvement in lung function on inhaled corticosteroids in pediatric asthmatics. Pharmacogenetics and Genomics 2019, 29, 65-68, 10.1097/fpc.0000000000000366.
  26. Susanne J. Szabo; Nile G. Jacobson; Anand S. Dighe; Uell Gubler; Kenneth M. Murphy; Developmental commitment to the Th2 lineage by extinction of IL-12 signaling. Immunity 1995, 2, 665-675, 10.1016/1074-7613(95)90011-x.
  27. T Naseer; E M Minshall; D Y Leung; S Laberge; P Ernst; R J Martin; Q Hamid; Expression of IL-12 and IL-13 mRNA in asthma and their modulation in response to steroid therapy. American Journal of Respiratory and Critical Care Medicine 1997, 155, 845-851, 10.1164/ajrccm.155.3.9117015.
  28. A Keane-Myers; M Wysocka; G Trinchieri; M Wills-Karp; Resistance to antigen-induced airway hyperresponsiveness requires endogenous production of IL-12. The Journal of Immunology 1998, 161, 919–926.
  29. R Giordano; A Picu; L Bonelli; F Broglio; F Prodam; S Grottoli; Giampiero Muccioli; E. Ghigo; Emanuela Arvat; The activation of somatostatinergic receptors by either somatostatin-14 or cortistatin-17 often inhibits ACTH hypersecretion in patients with Cushing’s disease. European Journal of Endocrinology 2007, 157, 393-398, 10.1530/eje-07-0147.
  30. Elena Gonzalez-Rey; Marta Pedreño; Virginia Delgado-Maroto; Luciana Souza-Moreira; M. Delgado; Lulling immunity, pain, and stress to sleep with cortistatin. Annals of the New York Academy of Sciences 2015, 1351, 89-98, 10.1111/nyas.12789.
  31. Avery G. Frey; Daniel J. Palenchar; Justin D. Wildemann; Caroline C. Philpott; A Glutaredoxin·BolA Complex Serves as an Iron-Sulfur Cluster Chaperone for the Cytosolic Cluster Assembly Machinery. Journal of Biological Chemistry 2016, 291, 22344-22356, 10.1074/jbc.m116.744946.
  32. Bigler, J.; Boedigheimer, M.; Schofield, J.P.R.; Skipp, P.J.; Corfield, J.; Rowe, A.; Sousa, A.R.; Timour, M.; Twehues, L.; Hu, X.; et al.et al A Severe Asthma Disease Signature from Gene Expression Profiling of Peripheral Blood from U-BIOPRED Cohorts. American Journal of Respiratory and Critical Care Medicine 2017, 195, 1311-1320, 10.1164/rccm.201604-0866OC.
  33. Astle, W.J.; Elding, H.; Jiang, T.; Allen, D.; Ruklisa, D.; Mann, A.L.; Mead, D.; Bouman, H.; Riveros-Mckay, F.; Kostadima, M.A.; et al.et al The Allelic Landscape of Human Blood Cell Trait Variation and Links to Common Complex Disease. Cell 2016, 167, 1415-1429.e19, 10.1016/j.cell.2016.10.042.
  34. Xue Zhang; Jocelyn M. Biagini Myers; Veda K. Yadagiri; Ashley Ulm; Xiaoting Chen; Matthew T. Weirauch; Gurjit K. Khurana Hershey; Hong Ji; Nasal DNA methylation differentiates corticosteroid treatment response in pediatric asthma: A pilot study. PLOS ONE 2017, 12, e0186150, 10.1371/journal.pone.0186150.
  35. Alvin T. Kho; Michael McGeachie; Kip G. Moore; Jody M. Sylvia; Scott Weiss; Kelan G. Tantisira; Circulating microRNAs and prediction of asthma exacerbation in childhood asthma. Respiratory Research 2018, 19, 128, 10.1186/s12931-018-0828-6.
  36. Alvin T. Kho; Sunita Sharma; Joshua S. Davis; Joseph Spina; Dagnie Howard; Kevin McEnroy; Kip Moore; Jody Sylvia; Weiliang Qiu; Scott T. Weiss; Kelan G. Tantisira; Circulating MicroRNAs: Association with Lung Function in Asthma. PLOS ONE 2016, 11, e0157998, 10.1371/journal.pone.0157998.
  37. Yu-Liang Yeh; M.-W. Su; B.-L. Chiang; Y.-H. Yang; C.-H. Tsai; Yungling L. Lee; Genetic profiles of transcriptomic clusters of childhood asthma determine specific severe subtype. Clinical Experimental Allergy 2018, 48, 1164-1172, 10.1111/cea.13175.
  38. Weiliang Qiu; Feng Guo; Kimberly Glass; Guo-Cheng Yuan; John Quackenbush; Xiaobo Zhou; Kelan G. Tantisira; Differential connectivity of gene regulatory networks distinguishes corticosteroid response in asthma. Journal of Allergy and Clinical Immunology 2018, 141, 1250-1258, 10.1016/j.jaci.2017.05.052.
  39. Michael T. Beste; Nicole Pfäffle-Doyle; Emily A. Prentice; Stephanie N. Morris; Uglas A. Lauffenburger; Keith B. Isaacson; Linda G. Griffith; Molecular Network Analysis of Endometriosis Reveals a Role for c-Jun-Regulated Macrophage Activation. Science Translational Medicine 2014, 6, 222ra16, 10.1126/scitranslmed.3007988.
  40. Michael J. McGeachie; George L. Clemmer; Boris Hayete; Heming Xing; Karl Runge; Ann Chen Wu; Xiaofeng Jiang; Quan Lü; Bruce Church; Iya Khalil; Kelan Tantisira; Scott Weiss; Systems biology and in vitro validation identifies family with sequence similarity 129 member A (FAM129A) as an asthma steroid response modulator. Journal of Allergy and Clinical Immunology 2018, 142, 1479-1488.e12, 10.1016/j.jaci.2017.11.059.
  41. Shiqi Tang; Jianwen Wang; Jishi Liu; Yan Huang; Yueyi Zhou; Shikun Yang; Wei Zhang; Minghui Yang; Hao Zhang; Niban protein regulates apoptosis in HK-2 cells via caspase-dependent pathway. Renal Failure 2019, 41, 455-466, 10.1080/0886022x.2019.1619582.
  42. Shintaro Katayama; Katarina Stenberg Hammar; Kaarel Krjutškov; Elisabet Einarsdottir; Gunilla Hedlin; Juha Kere; Cilla Soderhall; Acute wheeze-specific gene module shows correlation with vitamin D and asthma medication. European Respiratory Journal 2019, 55, 1901330, 10.1183/13993003.01330-2019.
  43. Peter Langfelder; Steve Horvath; WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 2008, 9, 559, 10.1186/1471-2105-9-559.
  44. Qiaoshi Lian; Bing Sun; Interferons command Trim22 to fight against viruses. Cellular & Molecular Immunology 2017, 14, 794-796, 10.1038/cmi.2017.76.
  45. Pité, H.; Morais-Almeida, M.; Rocha, S.M. Metabolomics in Asthma: Where Do We Stand? Curr. Opin. Pulm. Med. 2018, 24, 94–103.
  46. Anne H. Neerincx; Susanne J. Vijverberg; Lieuwe D. Bos; Paul Brinkman; Marc P. Van Der Schee; Rianne De Vries; Peter J. Sterk; Anke H. Maitland-Van Der Zee; Breathomics from exhaled volatile organic compounds in pediatric asthma. Pediatric Pulmonology 2017, 52, 1616-1627, 10.1002/ppul.23785.
  47. Su Jung Kim; Su Hee Kim; Ji Hyun Kim; Shin Hwang; Hyun Ju Yoo; Understanding Metabolomics in Biomedical Research. Endocrinology and Metabolism 2016, 31, 7-16, 10.3803/EnM.2016.31.1.7.
  48. Rachel Kelly; Joanne E. Sordillo; Sharon Lutz; Lydiana Avila; Manuel Soto-Quiros; Juan C. Celedón; Michael McGeachie; Amber Dahlin; Kelan G Tantisira; Mengna Huang; Clary B. Clish; Scott Weiss; Jessica Lasky-Su; Ann Chen Wu; Pharmacometabolomics of Bronchodilator Response in Asthma and the Role of Age-Metabolite Interactions.. Metabolites 2019, 9, 179, 10.3390/metabo9090179.
  49. Rachel Kelly; Bo L. Chawes; Kevin Blighe; Yamini V. Virkud; Damien Croteau-Chonka; Michael J. McGeachie; Clary B. Clish; Kevin Bullock; Juan C. Celedón; Scott Weiss; Jessica Lasky-Su; An Integrative Transcriptomic and Metabolomic Study of Lung Function in Children With Asthma. Chest 2018, 154, 335-348, 10.1016/j.chest.2018.05.038.
  50. Tomas Paulenda; Petr Draber; The role of ORMDL proteins, guardians of cellular sphingolipids, in asthma. Allergy 2016, 71, 918-930, 10.1111/all.12877.
  51. Antoaneta Toncheva; Daniel P. Potaczek; Michaela Schedel; Søren W. Gersting; Sven Michel; Natalie Krajnov; Vincent Gaertner; Julian M. Klingbeil; Thomas Illig; Andre Franke; Carla Winkler; Jens M. Hohlfeld; Christian Vogelberg; Andrea Von Berg; Albrecht Bufe; Andrea Heinzmann; Otto Laub; Ernst Rietschel; Burkhard Simma; Jon Genuneit; Ania C. Muntau; Michael Kabesch; Childhood asthma is associated with mutations and gene expression differences ofORMDLgenes that can interact. Allergy 2015, 70, 1288-1299, 10.1111/all.12652.
  52. Jennie G. Ono; Tilla S. Worgall; Stefan Worgall; 17q21 locus and ORMDL3: an increased risk for childhood asthma. Pediatric Research 2014, 75, 165-170, 10.1038/pr.2013.186.
  53. George Weinstock; Genomic approaches to studying the human microbiota. Nature 2012, 489, 250-256, 10.1038/nature11553.
  54. Ranjan, R.; Rani, A.; Metwally, A.; McGee, H.S.; Perkins, D.L; Analysis of the Microbiome: Advantages of Whole Genome Shotgun versus 16S Amplicon Sequencing. Biochem. Biophys. Biochemical and Biophysical Research Communications 2016, 469, 967-977, 10.1016/j.bbrc.2015.12.083.
  55. Yanjiao Zhou; Daniel Jackson; Leonard B. Bacharier; David Mauger; Homer Boushey; Mario Castro; Juliana Durack; Yvonne Huang; Robert F. Lemanske Jr; Gregory A. Storch; et al.George WeinstockKristine WylieRonina CovarAnne M. FitzpatrickWanda PhipatanakulRachel G. RobisonAvraham Beigelman The upper-airway microbiota and loss of asthma control among asthmatic children. Nature Communications 2019, 10, 5714, 10.1038/s41467-019-13698-x.
  56. Juliana Durack; Yvonne J Huang; Snehal Nariya; Laura S. Christian; K. Mark Ansel; Avraham Beigelman; Mario Castro; Anne-Marie Dyer; Elliot Israel; Monica Kraft; Richard J. Martin; David T. Mauger; Sharon R. Rosenberg; Tonya S. King; Steven R. White; Loren C. Denlinger; Fernando Holguin; Stephen C. Lazarus; Njira Lugogo; Stephen P. Peters; Lewis J. Smith; Michael E. Wechsler; Susan V. Lynch; Homer A. Boushey; National Heart, Lung and Blood Institute’s “AsthmaNet”; Bacterial biogeography of adult airways in atopic asthma.. Microbiome 2018, 6, 104, 10.1186/s40168-018-0487-3.
  57. Chung, K.F. Airway Microbial Dysbiosis in Asthmatic Patients: A Target for Prevention and Treatment? J. Allergy Clin. Immunol. 2017, 139, 1071–1081.
  58. Martin Depner; Markus J. Ege; Michael J Cox; Sarah Dwyer; Alan W. Walker; Lena T. Birzele; Jon Genuneit; Elisabeth Horak; Charlotte Braun-Fahrländer; Hanna Danielewicz; Raina M. Maier; Miriam F. Moffatt; William Osmond Cookson; Dick J.J. Heederik; Erika Von Mutius; Antje Legatzki; Miriam F. Moffat; Bacterial microbiota of the upper respiratory tract and childhood asthma. Journal of Allergy and Clinical Immunology 2017, 139, 826-834.e13, 10.1016/j.jaci.2016.05.050.
  59. Kathryn McCauley; Juliana Durack; Ricardo Valladares; Uglas W. Fadrosh; Din L. Lin; Agustin Calatroni; Petra K. Lebeau; Hoang Tran; Kei E. Fujimura; Brandon LaMere; Geil Merana; Kole Lynch; Robyn T. Cohen; Jacqueline Pongracic; Gurjit K. Khurana Hershey; Carolyn M. Kercsmar; Michelle Gill; Andrew H. Liu; Haejin Kim; Meyer Kattan; Stephen J. Teach; Alkis Togias; Homer A. Boushey; James E. Gern; Daniel J. Jackson; Susan V. Lynch; National Institute of Allergy and Infectious Diseases–sponsored Inner-City Asthma Consortium; Distinct nasal airway bacterial microbiotas differentially relate to exacerbation in pediatric patients with asthma. Journal of Allergy and Clinical Immunology 2019, 144, 1187-1197, 10.1016/j.jaci.2019.05.035.
  60. Yanjiao Zhou; Hongyu Gao; Kathie Mihindukulasuriya; Patricio S La Rosa; Kristine M Wylie; Tatiana A. Vishnivetskaya; Mircea L. Podar; Barbara B. Warner; Phillip I. Tarr; David E. Nelson; et al.J. Dennis FortenberryMartin J HollandSarah E BurrWilliam D ShannonErica SodergrenGeorge Weinstock Biogeography of the ecosystems of the healthy human body. Genome Biology 2013, 14, R1, 10.1186/gb-2013-14-1-r1.
More
Video Production Service