Skip to main content

OPINION article

Front. Cell. Infect. Microbiol., 07 March 2019
Sec. Microbiome in Health and Disease

Allergy as a Disease of Dysbiosis: Is It Time to Shift the Treatment Paradigm?

  • Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States

Attempts to strike a truce in the “nature-vs. -nurture” debate often end up framing most human disease as “a complex interaction of genes and environment.” This truce fails to distinguish the driver of disease from the risk modifier. Health care providers universally identify tobacco smoke is the root cause (i.e., “driver”) of chronic obstructive pulmonary disease (COPD) despite the known presence of associated monogenetic disorders and risk alleles (“risk modifiers”) (Hardin and Silverman, 2014). However, manuscripts reviewing ailments with less overt environmental causes, such as those covering atopic disease, default to prioritizing this dynamic in the opposite orientation. Allergic disease is frequently referred to as a “complex genetic disease” with “interacting environmental factors” (Weidinger et al., 2018), rather than a complex environmental disease with interacting genetic modifiers. To some extent, a debate about defining disease by gene-environment vs. environment-gene interactions is one of semantics. However, just as language can shape how we define a problem, so too can it influence our perception of its solution. This manuscript will center its discussion on atopic dermatitis (AD), but the commentary is applicable to allergy writ large, as well as diseases ranging from oncology to psychiatry.

Initial twin concordance studies overestimated heritability for AD to be around 75% (Weidinger et al., 2018), failing to recognize that identical twins share their environment more intensely than fraternal twins (Mayhew and Meyre, 2017). Early efforts using genome wide association studies (GWAS) to search for what these initial estimates of heritability suggested would be the causal gene were bolstered by the discovery of monogenic disorders marked by eczematous phenotypes (Lyons et al., 2015). However, many GWAS fail to find a single association; those that do, report allelic frequencies below 10% in their disease population (Bin and Leung, 2016). Of the variants implicated in AD by these individual GWAS trials, only association with FILAGGRIN could be confirmed by meta-analysis aggregation (Paternoster et al., 2011). While the resultant drop in sequence-specific heritability predictions to “ <20%” (Weidinger et al., 2018) does not negate the importance of identified genetic pathways [particularly interleukin (IL-) 4 and IL-13], it does suggest that the majority of patients with AD manifest disease despite genomes that are indistinguishable from healthy controls.

Often, the “missing heritability” is met with calls for better human-genomic assays, such as higher computational power combined with deeper sequencing or evaluation of epigenetic modifications. While greater technology may reveal currently undetectable interactions, it seems as if decades of advances in genomic assessment tools have only changed the working hypothesis from one stating improved genomics will uncover the causal gene, to one stating that improved genomics will uncover the causal gene(s). This hypothesis predominates despite the burden of atopy increasing at a rate more suggestive of being driven by environmental factors (Silverberg, 2017).

In contrast with GWAS results, most microbiome publications have deployed tools of relatively low resolution to provide phyla-level differences in 16 S bacterial ribosomal RNA (Grice and Segre, 2012). For context, assays that could only provide phyla-level analysis of the animal kingdom would be incapable of distinguishing a human from a gold fish. Furthermore, additional variability in the current microbiome literature stems from differing methodical approaches such as sequencing only culturable vs. total bacteria, and/or choice of MLST (multi-locus sequence typing), ribosomal-only genomic, or full metagenomic analysis (Grice and Segre, 2012). Yet, microbiome assessments have found several disease-predictive associations between dysbiosis and atopy. Dysbiosis might be broadly defined as any composition of commensal microbes which deviates from the composition associated with health (Petersen and Round, 2014). Deviations in the microbiome of the nasal passage and lungs differentiates allergic rhinitis and asthma from healthy controls (Pascal et al., 2018). Differences in the gut microbiome associate with food allergy, asthma, and AD (Pascal et al., 2018). Differences in the skin microbiome, especially for Staphylococcus aureus burden, are causally associated with several features of AD pathology (Huang et al., 2017). These associations are even more pronounced if the dysbiosis is seen early in life (Byrd et al., 2017). While current associations between atopy and dysbiosis of the nasal passage, gut, and lungs are limited to bacterial signatures, additional associations between dysbiosis in commensal fungi (the mycobiome) and AD have been identified (Han et al., 2018). Contributions of commensal viruses (the virome) have yet to be elucidated for any allergic disease. However, causal links between asthma and pathologic exposure to viruses (Jartti and Gern, 2017; Kirenga et al., 2018) and parasites (Cruz et al., 2017) have been proposed.

Epidemiologic and clinical trial data further suggest that environmental and therapeutic manipulation of the microbiome can influence atopy. Northern Europeans raised on farms are relatively protected from atopy when compared to their city-dwelling cousins (Cho and Blaser, 2012; Stein et al., 2016; Pascal et al., 2018). Rural-to-urban migration increases atopy risk if the migration occurs pre-birth (maternal migration), during infancy (Rodriguez et al., 2016), or during adulthood (Tham et al., 2018). Animal models supplement epidemiologic data by directly demonstrating that parental diet can exert heritable influence over offspring immunity via the microbiome (Myles, 2014). Although the data is incomplete, there is mounting evidence that stress (either in the patients or their mother) increases the risk of allergic disease (Gilles et al., 2018). Cesarean-section delivery, absence of older siblings, early-life antibiotic use, and many other environmental factors (Kantor and Silverberg, 2017) are all strongly associated with the development of atopy and other immune-mediated diseases (Cho and Blaser, 2012; Pascal et al., 2018). Thus, while all the described environmental influencers are complex, the microbiome may partially explain the “missing heritability” through both direct passage of microbes and alteration of host epigenetics (Sandoval-Motta et al., 2017; Tost, 2018). While use of oral probiotics has shown mixed results for prevention of atopy (Cabana et al., 2017; Pascal et al., 2018), supplemental Lactobacillus rhamnosus enhanced the efficacy of oral immunotherapy for peanuts and milk (Pascal et al., 2018). An early-stage clinical trial using topical application of live Roseomonas mucosa demonstrated improvement in symptoms and associated features of AD (Myles et al., 2018), and topical coagulase negative Staphylococci demonstrated a reduction in the relative abundance of S. aureus (Nakatsuji et al., 2017).

The increased depth of microbial identification offered by metagenomics combined with metabolic pathway analysis may reveal the critical functional pathways of the microbiome that have been suggested by investigations of gnotobiotic mice (Bhattarai and Kashyap, 2016). Elucidating immunologic functions that require microbes will be just as, if not more important, as learning which specific microbes can meet those needs. Of note, improved genetic tools might uncover human variants that lack the ability to support the growth of, or properly respond to, otherwise mutualistic organisms. Therefore, paradoxically, failure to sufficiently investigate the impact of microbes on human health may be causing researchers to overlook the functional consequences of some genetic variants.

The major critique to the position of this manuscript is that current understanding of atopy deems it “obvious” that genetic contributions to allergic disease are low, while the environment is a major influencer. This sentiment is fair, and is supported by excellent recent reviews on the role of the environment in atopy (von Mutius, 2016; Gilles et al., 2018; Wikstén et al., 2018). However, the sentiment that the environment is the most important influencer of allergic disease does not appear to have been translated into the treatment guidelines. The only environmental modifications recommended by the consensus statements for AD treatment form the US (Schneider et al., 2013; Boguniewicz, 2014), Europe (Wollenberg et al., 2018), and Japan (Katayama et al., 2017) are to avoid known allergens such as animals in the home, clothing material, and foods triggers. Similarly, guidelines for rhinitis suggest avoiding those allergens known to be triggering (Dykewicz et al., 2017; Scadding et al., 2017), without any mentions of possible preventive strategies. Furthermore, a recent overview of the drugs in development for AD listed only Jak/Stat inhibitors and anti-cytokine biologics (Weidinger et al., 2018).

Under a new environment-gene paradigm, we would not only expand research into therapeutics that target dysbiosis, we would devote far more resources into examining potential nutritional interventions, stress management, and environmental modifications for the purposes of primary prevention. Current chemical safety tests limited to impacts on mammalian cells would be expanded to examine which environmental stimuli impact the microbiome and how those impacts influence human health. The original hygiene hypothesis stated that exposure to pathogens (or their molecular remnants) confers health by serving as a gauntlet for honing immunity (Strachan et al., 1996). Modern concepts around the hygiene hypothesis acknowledge that a diverse microbial exposure profile, which includes both pathogens and non-pathogens, is required for the development of a healthy immune system (Arrieta et al., 2015). Given the importance of protecting beneficial microbial communities, the potential dysbiotic side effects of pharmaceuticals would be investigated alongside current clinical adverse event assessments. This would be especially true for research into the impacts of the microbiome on early childhood development given that these formative exposures may begin in utero (Pascal et al., 2018).

For overall benefit to human health, the impact heralded by the dawn of the genomic era may one day only be rivaled by the onset of sanitation and vaccination. Mass scale genome searches such as the National Institutes of Health's new All of Us initiative will build upon The Human Genome Project to identify additional genetic targets and improve lives. However, while nature-vs. -nurture may be a false dichotomy, it is not akin to debating the-chicken-or-the-egg. With the mounting evidence stemming from The Human Microbiome Project, our paradigm must shift to incorporate an evidenced-based balance between genetic determinism, environmental influence, and microbial mutualism. Genetics will be the primary driver for some diseases while serving as modifiers for others. At least for allergy, our refusal to think outside of the helix may be limiting recognition of actionable discoveries that could provide large-scale benefit to public health.

Author Contributions

The author confirms being the sole contributor of this work and has approved it for publication.

Conflict of Interest Statement

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The opinions expressed by the author do not reflect those of the US Federal Government, NIH, or US Public Health Service. This work was supported by the Intramural Research Program of The National Institute of Allergy and Infectious Diseases and The National Institutes of Health.

References

Arrieta, M. C., Stiemsma, L. T., Dimitriu, P. A., Thorson, L., Russell, S., Yurist-Doutsch, S., et al. (2015). Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 7:307ra152. doi: 10.1126/scitranslmed.aab2271

PubMed Abstract | CrossRef Full Text

Bhattarai, Y., and Kashyap, P. C. (2016). Germ-free mice model for studying host-microbial interactions. Methods Mol. Biol. 1438, 123–135. doi: 10.1007/978-1-4939-3661-8_8

PubMed Abstract | CrossRef Full Text | Google Scholar

Bin, L., and Leung, D. Y. (2016). Genetic and epigenetic studies of atopic dermatitis. Allergy Asthma Clin. Immunol. 12:52. doi: 10.1186/s13223-016-0158-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Boguniewicz, M. (2014). Atopic dermatitis: the updated practice parameter and beyond. Allergy Asthma Proc. 35, 429–434. doi: 10.2500/aap.2014.35.3798

PubMed Abstract | CrossRef Full Text | Google Scholar

Byrd, A. L., Deming, C., Cassidy, K. B. S., Harrison, O. J., Ng, W. I., Conlan, S., et al. (2017). Staphylococcus aureus and Staphylococcus epidermidis strain diversity underlying pediatric atopic dermatitis. Sci. Transl. Med. 9:eaal4651. doi: 10.1126/scitranslmed.aal4651

PubMed Abstract | CrossRef Full Text | Google Scholar

Cabana, M. D., McKean, M., Caughey, A. B., Fong, L., Lynch, S., Wong, A., et al. (2017). Early probiotic supplementation for eczema and asthma prevention: a randomized controlled trial. Pediatrics 140:e20163000. doi: 10.1542/peds.2016-3000

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, I., and Blaser, M. J. (2012). The human microbiome: at the interface of health and disease. Nat. Rev. Genet. 13, 260–270. doi: 10.1038/nrg3182

PubMed Abstract | CrossRef Full Text | Google Scholar

Cruz, A. A., Cooper, P. J., Figueiredo, C. A., Alcantara-Neves, N. M., Rodrigues, L. C., and Barreto, M. L. (2017). Global issues in allergy and immunology: parasitic infections and allergy. J. Allergy Clin. Immunol. 140, 1217–1228. doi: 10.1016/j.jaci.2017.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Dykewicz, M. S., Wallace, D. V., Baroody, F., Bernstein, J., Craig, T., Finegold, I., et al. (2017). Treatment of seasonal allergic rhinitis: an evidence-based focused 2017 guideline update. Ann. Allergy Asthma Immunol. 119, 489–511.e41. doi: 10.1016/j.anai.2017.08.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Gilles, S., Akdis, C., Lauener, R., Schmid-Grendelmeier, P., Bieber, T., Schäppi, G., et al. (2018). The role of environmental factors in allergy: a critical reappraisal. Exp. Dermatol. 27, 1193–1200. doi: 10.1111/exd.13769

PubMed Abstract | CrossRef Full Text | Google Scholar

Grice, E. A., and Segre, J. A. (2012). The human microbiome: our second genome. Annu. Rev. Genomics Hum. Genet. 13, 151–170. doi: 10.1146/annurev-genom-090711-163814

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, S. H., Cheon, H. I., Hur, M. S., Kim, M. J., Jung, W. H., Lee, Y. W., et al. (2018). Analysis of the skin mycobiome in adult patients with atopic dermatitis. Exp. Dermatol. 27, 366–373. doi: 10.1111/exd.13500

PubMed Abstract | CrossRef Full Text | Google Scholar

Hardin, M., and Silverman, E. K. (2014). Chronic obstructive pulmonary disease genetics: a review of the past and a look into the future. Chronic. Obstr. Pulm. Dis. 1, 33–46. doi: 10.15326/jcopdf.1.1.2014.0120

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y. J., Marsland, B. J., Bunyavanich, S., O'Mahony, L., Leung, D. Y., Muraro, A., et al. (2017). The microbiome in allergic disease: current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma and Immunology and the European Academy of Allergy and Clinical Immunology. J. Allergy Clin. Immunol. 139, 1099–1110. doi: 10.1016/j.jaci.2017.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Jartti, T., and Gern, J. E. (2017). Role of viral infections in the development and exacerbation of asthma in children. J. Allergy Clin. Immunol. 140, 895–906. doi: 10.1016/j.jaci.2017.08.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kantor, R., and Silverberg, J. I. (2017). Environmental risk factors and their role in the management of atopic dermatitis. Expert Rev. Clin. Immunol. 13, 15–26. doi: 10.1080/1744666X.2016.1212660

PubMed Abstract | CrossRef Full Text | Google Scholar

Katayama, I., Aihara, M., Ohya, Y., Saeki, H., Shimojo, N., Shoji, S., et al. (2017). Japanese Society of, Japanese guidelines for atopic dermatitis 2017. Allergol. Int. 66, 230–247. doi: 10.1016/j.alit.2016.12.003

CrossRef Full Text | Google Scholar

Kirenga, B. J., Mugenyi, L., de Jong, C., Lucian Davis, J., Katagira, W. T., et al. (2018). The impact of HIV on the prevalence of asthma in Uganda: a general population survey. Respir. Res. 19:184. doi: 10.1186/s12931-018-0898-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Lyons, J. J., Milner, J. D., and Stone, K. D. (2015). Atopic dermatitis in children: clinical features, pathophysiology, and treatment. Immunol. Allergy Clin. North Am. 35, 161–183. doi: 10.1016/j.iac.2014.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Mayhew, A. J., and Meyre, D. (2017). Assessing the heritability of complex traits in humans: methodological challenges and opportunities. Curr. Genomics 18, 332–340. doi: 10.2174/1389202918666170307161450

PubMed Abstract | CrossRef Full Text | Google Scholar

Myles, I. A. (2014). Fast food fever: reviewing the impacts of the Western diet on immunity. Nutr. J. 13:61. doi: 10.1186/1475-2891-13-61

PubMed Abstract | CrossRef Full Text | Google Scholar

Myles, I. A., Earland, N. J., Anderson, E. D., Moore, I. N., Kieh, M. D., Williams, K. W., et al. (2018). First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis. JCI Insight 3:120608. doi: 10.1172/jci.insight.120608

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakatsuji, T., Chen, T. H., Narala, S., Chun, K. A., Two, A. M., Yun, T., et al. (2017). Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Sci. Transl. Med. 9:eaah4680. doi: 10.1126/scitranslmed.aah4680

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascal, M., Perez-Gordo, M., Caballero, T., Escribese, M. M., Lopez Longo, M. N., Luengo, O., et al. (2018). Microbiome and allergic diseases. Front. Immunol. 9:1584. doi: 10.3389/fimmu.2018.01584

PubMed Abstract | CrossRef Full Text | Google Scholar

Paternoster, L., Standl, M., Chen, C. M., Ramasamy, A., Bonnelykke, K., Duijts, L., et al. (2011). Meta-analysis of genome-wide association studies identifies three new risk loci for atopic dermatitis. Nat. Genet. 44, 187–192. doi: 10.1038/ng.1017

PubMed Abstract | CrossRef Full Text | Google Scholar

Petersen, C., and Round, J. L. (2014). Defining dysbiosis and its influence on host immunity and disease. Cell. Microbiol.16, 1024–1033. doi: 10.1111/cmi.12308

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodriguez, A., Vaca, M. G., Chico, M. E., Rodrigues, L. C., Barreto, M. L., and Cooper, P. J. (2016). Migration and allergic diseases in a rural area of a developing country. J. Allergy Clin. Immunol. 138, 901–903. doi: 10.1016/j.jaci.2016.01.052

PubMed Abstract | CrossRef Full Text | Google Scholar

Sandoval-Motta, S., Aldana, M., Martinez-Romero, E., and Frank, A. (2017). The human microbiome and the missing heritability problem. Front. Genet. 8:80. doi: 10.3389/fgene.2017.00080

PubMed Abstract | CrossRef Full Text | Google Scholar

Scadding, G. K., Kariyawasam, H. H., Scadding, G., Mirakian, R., Buckley, R. J., Dixon, T., et al. (2017). BSACI guideline for the diagnosis and management of allergic and non-allergic rhinitis (Revised Edition 2017; First edition 2007). Clin. Exp. Allergy 47, 856–889. doi: 10.1111/cea.12953

PubMed Abstract | CrossRef Full Text | Google Scholar

Schneider, L., Tilles, S., Lio, P., Boguniewicz, M., Beck, L., LeBovidge, J., et al. (2013). Atopic dermatitis: a practice parameter update 2012. J. Allergy Clin. Immunol. 131, 295–9.e1–27. doi: 10.1016/j.jaci.2012.12.672

PubMed Abstract | CrossRef Full Text | Google Scholar

Silverberg, J. I. (2017). Public health burden and epidemiology of atopic dermatitis. Dermatol. Clin. 35, 283–289. doi: 10.1016/j.det.2017.02.0026

PubMed Abstract | CrossRef Full Text | Google Scholar

Stein, M. M., Hrusch, C. L., Gozdz, J., Igartua, C., Pivniouk, V., Murray, S. E., et al. (2016). Innate immunity and asthma risk in amish and hutterite farm children. N. Engl. J. Med. 375, 411–421. doi: 10.1056/NEJMoa1508749

PubMed Abstract | CrossRef Full Text | Google Scholar

Strachan, D. P., Taylor, E. M., and Carpenter, R. G. (1996). Family structure, neonatal infection, and hay fever in adolescence. Arch. Dis. Child. 74, 422–426. doi: 10.1136/adc.74.5.422

PubMed Abstract | CrossRef Full Text | Google Scholar

Tham, E. H. E., Loo, X. L., Zhu, Y., and Shek, L. P. (2018). Effects of migration on allergic diseases. Int. Arch. Allergy Immunol. 178, 1–13. doi: 10.1159/000494129

PubMed Abstract | CrossRef Full Text | Google Scholar

Tost, J. (2018). A translational perspective on epigenetics in allergic diseases. J. Allergy Clin. Immunol. 142, 715–726. doi: 10.1016/j.jaci.2018.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

von Mutius, E. (2016). The microbial environment and its influence on asthma prevention in early life. J. Allergy Clin. Immunol. 137, 680–689. doi: 10.1016/j.jaci.2015.12.1301

PubMed Abstract | CrossRef Full Text | Google Scholar

Weidinger, S., Beck, L. A., Bieber, T., Kabashima, K., and Irvine, A. D. (2018). Atopic dermatitis. Nat. Rev. Dis. Primers 4:1. doi: 10.1038/s41572-018-0001-z

PubMed Abstract | CrossRef Full Text

Wikstén, J., Toppila-Salmi, S., and Mäkelä, M. (2018). Primary prevention of airway allergy. Curr. Treat Opt. Allergy 5, 347–355. doi: 10.1007/s40521-018-0190-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wollenberg, A., Barbarot, S., Bieber, T., Christen-Zaech, S., Deleuran, M., Fink-Wagner, A., et al. (2018). Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part I. J. Eur. Acad. Dermatol. Venereol. 32, 657–682. doi: 10.1111/jdv.14891

CrossRef Full Text | Google Scholar

Keywords: allergy, dysbiosis, microbiome, eczema, atopic dermatitis

Citation: Myles IA (2019) Allergy as a Disease of Dysbiosis: Is It Time to Shift the Treatment Paradigm? Front. Cell. Infect. Microbiol. 9:50. doi: 10.3389/fcimb.2019.00050

Received: 23 December 2018; Accepted: 18 February 2019;
Published: 07 March 2019.

Edited by:

D. Scott Merrell, Uniformed Services University, United States

Reviewed by:

Kari Christine Nadeau, Stanford University, United States

Copyright © 2019 Myles. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Ian A. Myles, mylesi@niaid.nih.gov

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.