Next Article in Journal
Cartilage Collagen Neoepitope C2C Expression in the Articular Cartilage and Its Relation to Joint Tissue Damage in Patients with Knee Osteoarthritis
Previous Article in Journal
Adipose Tissue Segmentation after Lung Slice Localization in Chest CT Images Based on ConvBiGRU and Multi-Module UNet
Previous Article in Special Issue
Obesity-Associated Non-T2 Mechanisms in Obese Asthmatic Individuals
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Unraveling the Complexity of Asthma: Insights from Omics Approaches

by
Esther Herrera-Luis
1 and
Natalia Hernandez-Pacheco
2,3,*
1
Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
2
Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, 11883 Stockholm, Sweden
3
CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(5), 1062; https://doi.org/10.3390/biomedicines12051062
Submission received: 7 May 2024 / Accepted: 10 May 2024 / Published: 11 May 2024
(This article belongs to the Special Issue Recent Advances in Asthma Research in a Multiomics Era)
Asthma is a heterogeneous respiratory disease that represents a substantial social and economic burden [1] and is highly detrimental to the quality of life of both patients and their caregivers [2]. It is the most common chronic disease in children and adolescents [2]. Early therapeutic interventions are critical, as severe symptoms are common at these stages of the life course [3]. Additionally, severe childhood asthma accompanied by a substantial decline in lung function is an important risk factor for the persistence of asthma and the development of other chronic airway diseases during adulthood [4,5,6].
Asthma is a complex condition resulting from numerous interactions between endogenous and exogenous factors [7,8]. To date, most research efforts have focused on unraveling the genetic determinants and environmental factors that play a key role in asthma [9]. Nonetheless, our understanding of the mechanisms underlying this condition and related traits remains incomplete [10], most likely because asthma encompasses a diverse range of pathophysiological mechanisms and variable clinical manifestations [2]. As a result, despite the widespread application of genomics in asthma research [11], we continue to lag far behind in translating its findings into the clinical setting [12]. Moving towards a treatable traits perspective in chronic airway disease could further address the complexity of asthma, as well as patient heterogeneity [13]. Novel opportunities for genetic risk prediction in asthma have also arisen as a result of the recent development of multi-ancestry polygenic risk scores (PRSs) that aggregate the association effect of numerous variants across the genome [14,15]. Nonetheless, other omics remain underrepresented in this field; this is especially true with regard to the concept of combining several data layers through a multiomic strategy [16,17]. These may substantially contribute to the accurate diagnosis of asthma and the development of novel, effective preventive and management strategies, which can be personalized to each patient. Hence, this Special Issue focuses on recent advances in the application of omics approaches to investigate asthma and related traits.
The first work by Martin-Almeida et al. (2023) explores the blood DNA methylome of bronchodilator drug response (BDR) and fractional exhaled nitric oxide (FeNO) in a multi-ethnic pediatric cohort with moderate-to-severe asthma [18]. The authors identify several significant differentially methylated regions (DMRs) enriched in genes involved in aging and immune pathways. Moreover, the CpG cg12835256 (PLA2G12A) reaches genome-wide significance for FeNO and is replicated in nasal epithelial cells. PLA2G12A encodes for the phospholipase A2 Group XIIA, which is implicated in bronchial hyperresponsiveness and eicosanoid synthesis in asthma [19]. These data argue in favor of prior evidence of the potential role of eicosanoids as mediators of airway inflammation in asthma [20].
Secondly, Logoń et al. [21] provide an elegant state-of-the-art summary of the investigation of the microbiome in asthma pathogenesis and treatment. The authors thoroughly describe the importance of the gut and airway microbiota composition in the development and modulation of the immune response, as well as asthma susceptibility and treatment response. Furthermore, metabolites produced by commensal gut bacteria have antibiotic effects against pathogenic microorganisms. Therefore, an altered composition and functionality of the intestinal microbiota caused by environmental exposures across the life course, but most importantly, during infancy and childhood, might be involved in an imbalance in bacterial species and the alteration of the inflammatory response and asthma risk [21]. On the other hand, the bronchial microbiota composition has shown a strong association with a differential response to conventional asthma treatments [22,23]. For instance, enrichment in Haemophilus and bacteria of the Actinobacteria phylum has been associated with an increased response to inhaled corticosteroids (ICS), whereas Proteobacteria have been linked to poorer asthma control [22,23]. Additionally, prebiotic use, probiotics, dietary interventions, and fecal microbiota transplantation have been proposed as promising prophylactic or therapeutic strategies in asthma, though these approaches require further investigation [21].
Thirdly, Shailesh et al. explore (T2) mechanisms in asthma, as well as lung mechanics and function, by mining evidence from human studies and animal models described in the literature [24]. The authors emphasize the intricate interaction between obesity and asthma, highlighting the critical necessity of elucidating the relevant mechanisms, which are currently poorly understood. In particular, the authors describe the role of insulin resistance in triggering airway hyper-responsiveness in asthma. In this context, diabetes medications such as metformin and glucagon-like peptide 1 receptor (GLP-1R) agonists are promising treatments for obesity-associated asthma. Nonetheless, further research into the molecular mechanisms underlying the impact of obesity on asthma severity and randomized clinical trials to assess the efficacy of these drugs are warranted. On the other hand, obesity-related gut microbial dysbiosis and microbiome-derived short-chain fatty acids may influence immune dysregulation in asthma. Indeed, both human studies and animal models have demonstrated that a fiber-rich diet may significantly reduce asthma pathogenesis in obese asthmatic individuals through microbiome regulation. Moreover, the authors highlight the urgent need to investigate the molecular mechanisms underlying obesity-associated non-T2 factors in asthma to prioritize drug targets for potential treatment of obesity-associated asthma [24].
In summary, this Special Issue describes recent advances in asthma research through the application of promising omics approaches, evidencing their high potential for shortening the path toward obtaining a complete picture of the underlying molecular mechanisms of diverse asthma-related traits and the development of accurate diagnostic and management strategies.

Author Contributions

E.H.-L. and N.H.-P. contributed equally to the conceptualization, writing—original draft preparation, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

The Guest Editors would like to acknowledge the valuable contributions of all the authors who participated in this Special Issue.

Conflicts of Interest

The authors declare no conflicts of interest. The funding agencies had no role in the study design, data collection, and analysis, decision to publish, or preparation of the manuscripts. The views expressed are those of the authors and not necessarily those of any funder. E.H.-L. received support, outside of the submitted work, from the National Institute of Health (NIH) National Human Genome Research Institute (NHGRI) (R35HG011944-02). N.H.-P. was supported with a Medium-Term Research Fellowship by the European Academy of Allergy and Clinical Immunology (EAACI) and a Long-Term Research Fellowship by the European Respiratory Society (ERS) (LTRF202101-00861) outside of the submitted work.

References

  1. Ferkol, T.; Schraufnagel, D. The global burden of respiratory disease. Ann. Am. Thorac. Soc. 2014, 11, 404–406. [Google Scholar] [CrossRef] [PubMed]
  2. Global Strategy for Asthma Management and Prevention. Global Initiative for Asthma (GINA). 2023. Available online: http://ginasthma.org/ (accessed on 14 January 2024).
  3. Akdis, C.A.; Agache, I. Global Atlas of Asthma; European Academy of Allergy and Clinical Immunology (EAACI): Zurich, Switzerland, 2014. [Google Scholar]
  4. Svanes, C.; Sunyer, J.; Plana, E.; Dharmage, S.; Heinrich, J.; Jarvis, D.; de Marco, R.; Norback, D.; Raherison, C.; Villani, S.; et al. Early life origins of chronic obstructive pulmonary disease. Thorax 2010, 65, 14–20. [Google Scholar] [CrossRef] [PubMed]
  5. Bisgaard, H.; Nørgaard, S.; Sevelsted, A.; Chawes, B.L.; Stokholm, J.; Mortensen, E.L.; Ulrik, C.S.; Bønnelykke, K. Asthma-like symptoms in young children increase the risk of COPD. J. Allergy Clin. Immunol. 2021, 147, 569–576.e9. [Google Scholar] [CrossRef] [PubMed]
  6. Savran, O.; Bønnelykke, K.; Ulrik, C.S. Relationship between early life asthma and chronic airway disease in adult life—In search for disease trajectories over the life span—The RELATE study based on the Kongsberg cohort. BMC Pulm. Med. 2023, 23, 363. [Google Scholar] [CrossRef] [PubMed]
  7. Nishimura, K.K.; Galanter, J.M.; Roth, L.A.; Oh, S.S.; Thakur, N.; Nguyen, E.A.; Thyne, S.; Farber, H.J.; Serebrisky, D.; Kumar, R.; et al. Early-Life air pollution and asthma risk in minority children the GALA II and SAGE II studies. Am. J. Respir. Crit. Care Med. 2013, 188, 309–318. [Google Scholar] [CrossRef] [PubMed]
  8. Dharmage, S.C.; Perret, J.L.; Custovic, A. Epidemiology of asthma in children and adults. Front. Pediatr. 2019, 7, 246. [Google Scholar] [CrossRef] [PubMed]
  9. Hernandez-Pacheco, N.; Kere, M.; Melén, E. Gene-environment interactions in childhood asthma revisited; expanding the interaction concept. Pediatr. Allergy Immunol. 2022, 33, e13780. [Google Scholar] [CrossRef]
  10. Hernandez-Pacheco, N.; Pino-Yanes, M.; Flores, C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front. Pediatr. 2019, 7, 6. [Google Scholar] [CrossRef] [PubMed]
  11. Espuela-Ortiz, A.; Martin-Gonzalez, E.; Poza-Guedes, P.; González-Pérez, R.; Herrera-Luis, E. Genomics of Treatable Traits in Asthma. Genes 2023, 14, 1824. [Google Scholar] [CrossRef]
  12. Zhang, H.; Klareskog, L.; Matussek, A.; Pfister, S.M.; Benson, M. Translating genomic medicine to the clinic: Challenges and opportunities. Genome Med. 2019, 11, 9. [Google Scholar] [CrossRef]
  13. McDonald, V.M.; Fingleton, J.; Agusti, A.; Hiles, S.A.; Clark, V.L.; Holland, A.E.; Marks, G.B.; Bardin, P.P.; Beasley, R.; Pavord, I.D.; et al. Treatable traits: A new paradigm for 21st century management of chronic airway diseases: Treatable Traits down under International Workshop report. Eur. Respir. J. 2019, 53, 1802058. [Google Scholar] [CrossRef] [PubMed]
  14. Namjou, B.; Lape, M.; Malolepsza, E.; DeVore, S.B.; Weirauch, M.T.; Dikilitas, O.; Jarvik, G.P.; Kiryluk, K.; Kullo, I.J.; Liu, C.; et al. Multiancestral polygenic risk score for pediatric asthma. J. Allergy Clin. Immunol. 2022, 150, 1086–1096. [Google Scholar] [CrossRef] [PubMed]
  15. Tsuo, K.; Zhou, W.; Wang, Y.; Kanai, M.; Namba, S.; Gupta, R.; Majara, L.; Nkambule, L.L.; Morisaki, T.; Okada, Y.; et al. Multi-ancestry meta-analysis of asthma identifies novel associations and highlights the value of increased power and diversity. Cell Genom. 2022, 2, 100212. [Google Scholar] [CrossRef] [PubMed]
  16. Augustine, T.; Al-Aghbar, M.A.; Al-Kowari, M.; Espino-Guarch, M.; van Panhuys, N. Asthma and the Missing Heritability Problem: Necessity for Multiomics Approaches in Determining Accurate Risk Profiles. Front. Immunol. 2022, 13, 822324. [Google Scholar] [CrossRef]
  17. Gautam, Y.; Johansson, E.; Mersha, T.B. Multi-Omics Profiling Approach to Asthma: An Evolving Paradigm. J. Pers. Med. 2022, 12, 66. [Google Scholar] [CrossRef]
  18. Martin-Almeida, M.; Perez-Garcia, J.; Herrera-Luis, E.; Rosa-Baez, C.; Gorenjak, M.; Neerincx, A.H.; Sardón-Prado, O.; Toncheva, A.A.; Harner, S.; Wolff, C.; et al. Epigenome-Wide Association Studies of the Fractional Exhaled Nitric Oxide and Bronchodilator Drug Response in Moderate-to-Severe Pediatric Asthma. Biomedicines 2023, 11, 676. [Google Scholar] [CrossRef]
  19. Hallstrand, T.S.; Chi, E.Y.; Singer, A.G.; Gelb, M.H.; Henderson, W.R. Secreted phospholipase A2 group X overexpression in asthma and bronchial hyperresponsiveness. Am. J. Respir. Crit. Care Med. 2007, 176, 1072–1078. [Google Scholar] [CrossRef] [PubMed]
  20. Sokolowska, M.; Rovati, G.E.; Diamant, Z.; Untersmayr, E.; Schwarze, J.; Lukasik, Z.; Sava, F.; Angelina, A.; Palomares, O.; Akdis, C.A.; et al. Current perspective on eicosanoids in asthma and allergic diseases: EAACI Task Force consensus report, part I. Allergy Eur. J. Allergy Clin. Immunol. 2021, 76, 114–130. [Google Scholar] [CrossRef]
  21. Logoń, K.; Świrkosz, G.; Nowak, M.; Wrześniewska, M.; Szczygieł, A.; Gomułka, K. The Role of the Microbiome in the Pathogenesis and Treatment of Asthma. Biomedicines 2023, 11, 1618. [Google Scholar] [CrossRef]
  22. Durack, J.; Lynch, S.V.; Nariya, S.; Bhakta, N.R.; Beigelman, A.; Castro, M.; Dyer, A.-M.; Israel, E.; Kraft, M.; Martin, R.J.; et al. Features of the bronchial bacterial microbiome associated with atopy, asthma, and responsiveness to inhaled corticosteroid treatment. J. Allergy Clin. Immunol. 2017, 140, 63–75. [Google Scholar] [CrossRef]
  23. Huang, Y.J.; Nariya, S.; Harris, J.M.; Lynch, S.V.; Choy, D.F.; Arron, J.R.; Boushey, H. The airway microbiome in patients with severe asthma: Associations with disease features and severity. J. Allergy Clin. Immunol. 2015, 136, 874–884. [Google Scholar] [CrossRef] [PubMed]
  24. Shailesh, H.; Bhat, A.A.; Janahi, I.A. Obesity-Associated Non-T2 Mechanisms in Obese Asthmatic Individuals. Biomedicines 2023, 11, 2797. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Herrera-Luis, E.; Hernandez-Pacheco, N. Unraveling the Complexity of Asthma: Insights from Omics Approaches. Biomedicines 2024, 12, 1062. https://doi.org/10.3390/biomedicines12051062

AMA Style

Herrera-Luis E, Hernandez-Pacheco N. Unraveling the Complexity of Asthma: Insights from Omics Approaches. Biomedicines. 2024; 12(5):1062. https://doi.org/10.3390/biomedicines12051062

Chicago/Turabian Style

Herrera-Luis, Esther, and Natalia Hernandez-Pacheco. 2024. "Unraveling the Complexity of Asthma: Insights from Omics Approaches" Biomedicines 12, no. 5: 1062. https://doi.org/10.3390/biomedicines12051062

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop