Exposure to isocyanates predicts atopic dermatitis prevalence and disrupts therapeutic pathways in commensal bacteria

Jordan Zeldin, Prem Prashant Chaudhary, Jacquelyn Spathies, Manoj Yadav, Brandon N D'Souza, Mohammadali E Alishahedani, Portia Gough, Jobel Matriz, Andrew J Ghio, Yue Li, Ashleigh A Sun, Lawrence F Eichenfield, Eric L Simpson, Ian A Myles, Jordan Zeldin, Prem Prashant Chaudhary, Jacquelyn Spathies, Manoj Yadav, Brandon N D'Souza, Mohammadali E Alishahedani, Portia Gough, Jobel Matriz, Andrew J Ghio, Yue Li, Ashleigh A Sun, Lawrence F Eichenfield, Eric L Simpson, Ian A Myles

Abstract

Atopic dermatitis (AD) is a chronic inflammatory skin condition increasing in industrial nations at a pace that suggests environmental drivers. We hypothesize that the dysbiosis associated with AD may signal microbial adaptations to modern pollutants. Having previously modeled the benefits of health-associated Roseomonas mucosa, we now show that R. mucosa fixes nitrogen in the production of protective glycerolipids and their ceramide by-products. Screening EPA databases against the clinical visit rates identified diisocyanates as the strongest predictor of AD. Diisocyanates disrupted the production of beneficial lipids and therapeutic modeling for isolates of R. mucosa as well as commensal Staphylococcus. Last, while topical R. mucosa failed to meet commercial end points in a placebo-controlled trial, the subgroup who completed the full protocol demonstrated sustained, clinically modest, but statistically significant clinical improvements that differed by study site diisocyanate levels. Therefore, diisocyanates show temporospatial and epidemiological association with AD while also inducing eczematous dysbiosis.

Figures

Fig. 1.. R. mucosa from healthy controls…
Fig. 1.. R. mucosa from healthy controls shows differential carbon and nitrogen metabolism.
(A to J) Three different isolates of Roseomonas from healthy volunteers (RmHV1-3) and three different isolates of Roseomonas from patients with AD (RmAD1-3) were grown in the BioLog system, fold change (FC) for each challenge condition versus control was calculated, the log2FC for RmHV divided by the log2FC for RmAD is shown (A), and then the ratio of log2FC was superimposed onto the urea cycle (arginine biosynthesis) and citrate cycle (alanine, aspartate, and glutamate metabolism; H). (B and C) Individual BioLog components were ranked by significance and analyzed for pathways by MetaboAnalyst. (D) RmHV1-3 and RmAD1-3 were cultured with known amounts of LL-37 for 24 hours; supernatants were collected and assessed for remaining LL-37 by ELISA and the percentage of input LL-37 signal calculated and displayed. (E) Absorbance for cultures of RmHV1-3 or RmAD1-3 in medium with no energy source [minimal medium (MM)], with only carbohydrate energy sources (Nitrogen free), with only amino acid energy sources (Carb free), or with both (Replete). (F) Picture of experimental setup where anaerobic chambers were evacuated before filling with custom gas mixtures. (G) Total change in 13C to 12C ratio for all R. mucosa isolates after 48 hours in culture supplemented with 1% 13CO2. (H) MetaboAnalyst pathway analysis for RmHV1-3 and RmAD1-3 derived from metabolites with significant differences in 13C to 12C ratio after culture in 1% 13CO2 or metabolites differentially affected in RmHV by culture in ambient conditions or in CO2 deprivation (80% N2 and 20% O2). (I and J) Representative image (1.25×) of culture morphology (I) and statistical enumeration (J) of area covered per imaging field by culture of RmHV and RmAD in ambient or CO2 deprivation conditions. Data shown represent two or more independent experiments and are displayed as means ± SEM. For (C) and (H), only −log10P values of >1 were considered significant.
Fig. 2.. Roseomonas isolates differ in nitrogen…
Fig. 2.. Roseomonas isolates differ in nitrogen fixation.
(A) Total change in 15N to 14N ratio for three isolates of Roseomonas from healthy volunteers (RmHV1-3), three isolates of Roseomonas from patients with AD (RmAD1-3), or one isolate of A. brasilense (A. bras) cultured with 15N2 in head space. (B) Flux analysis was performed on metabolites with increase in 15N ratios; metabolites with greatest 15N change were ranked by P value and analyzed by MetaboAnalyst for pathways in lipidomic and metabolic acquisition conditions. (C) Representative images (1.25×) from spiral plated culture on agar for bacteria cultured in ambient air (Amb) or in head space devoid of N2 [79% argon (Ar), 20% oxygen (O2), and 1% carbon dioxide (CO2)]. (D and E) R. mucosa metabolites most affected by N2 deprivation were assessed by MALDI–time-of-flight (TOF) MS and analyzed for pathways affected by MetaboAnalyst. (F) Total intensity values for annotated ceramide containing compounds assessed by MALDI. (G) Mice treated in the MC903 model of AD for 8 days of dermatitis induction, 3 days of treatment with either RmHV cultured in ambient conditions of N2 deprivation (−N2), and then 4 days of observation; change in mouse ear thickness between days 8 and 15 is shown. Reduced thickness indicates reduced swelling and improved outcomes. (H) MALDI-MSI performed on mouse ears; segmentation performed by SCiLS Lab. (I) Pathway analysis derived from metabolites that best distinguish mice ears treated with RmHV1 from ambient conditions versus treatment with RmHV1 cultured in N2 deprivation. (J and K) Distribution of indicated metabolites as indicated by MALDI-MSI. GPL, glycerophospholipids; GPI, glycerophosphatidylinositol; deg., degradation; αLa, α-linolenic acid. Data shown represent two (G to K) or three or more (A to F) independent experiments. *P < 0.05; ns, not significant as determined by ANOVA.
Fig. 3.. Diisocyanates associated with AD diagnoses.
Fig. 3.. Diisocyanates associated with AD diagnoses.
(A) Summary of published environmental factors influencing AD (, , –32, 41, 42). Migration from developing to United States or European Union nations (US/EU); PM, particulate matter; ETS, environmental tobacco smoke; OC, organic carbon; OR, odds ratio; CI, confidence interval. (B) RmHV was cultured in sealed chamber with trace CO, CO + PM, SO2, or gasoline in agar dish lid and compared to pathways modified by N2 deprivation; green indicates that the MetaboAnalyst identified pathway was altered by chemical challenge; dark green indicates significant alteration; black indicates that no growth was evident. (C) Raw values and diagnosis rate for AD by zip code. (D) Cluster analysis for relative risk of having above-average AD diagnosis rate. Regression was performed for 1500+ pollution variables from the EPA TRI, AD diagnosis rates, obesity, deprivation index, and access to pediatricians, allergists, or dermatologist. (E to G) Linear regression using lasso with 1 SE limitation (E), random forest (F), or partial dependence plots (G) for total release values from 2014 to 2019 versus 2019 visit rates. (H) Random forest as in (F) but restricting to only billings by pediatricians. (I) Lasso linear regression with 1 SE for total 2019 pediatric visit rates versus 2014–2019 release values from the RSEI EPA database. (J) Aggregate AD-associated pollutant scores were summated by zip code for each pollutant multiplied by its β correlation value; mean values for each SD section were indicated.
Fig. 4.. TDI disrupts beneficial pathways in…
Fig. 4.. TDI disrupts beneficial pathways in R. mucosa.
(A) Total U.S. production of TDI and MDI since 1940. (B) The 2019 TDI production for nations with available data in linear regression with 95% confidence intervals against reported rates of AD in children under 14 years of age. (C) Detected levels of diisocyanates (DIC) including TDI released during in vitro experiments compared to reported factory release. (D) Representative image (1.25×) of RmHV1-3 cultured with TDI in ambient or N2 deprivation conditions. (E) TDI was measured at serial dilutions so that values at higher dilutions below the lower limit of detection (LLD) could be extrapolated. Percent change in growth and total intensity of annotated ceramides by MALDI at 48 hours are shown. (F) Change in total intensity for annotated ceramides as compared to RmHV1-3 cultured in ambient conditions or RmAD1-3 in ambient conditions (AD) or RmHV under N2 deprivation (−N2), or with TDI, toluene (TOL), or HNCO. (G) MetaboAnalyst indicated affected pathways in RmHV1-3 by culture with TDI or toluene. (H) Lower explosive limit (LEL) detection for RmHV1-3 with and without TDI exposure in ambient or N2 deprivation. (I) Mice treated in the MC903 model of AD before treatment with either diluent or RmHV1 grown with toluene or with TDI; change in mouse ear thickness is shown. Data shown are representative of three or more independent experiments and shown as means ± SEM. *P < 0.05, **P < 0.01 as determined by ANOVA.
Fig. 5.. HNCO disrupts beneficial pathways in…
Fig. 5.. HNCO disrupts beneficial pathways in R. mucosa.
(A) Representative image of RmHV1 cultured with 1 μg of HNCO placed into the lid of the agar dish and cultured for 48 hours in ambient or N2 deprivation conditions. (B) Lower explosive limit (LEL) detection by gas monitoring for RmHV with and without HNCO exposure in ambient or N2 deprivation culture conditions. (C) Change in mouse ear thickness during MC903 model of AD for mice treated with RmHV1 in ambient conditions or exposed to HNCO. (D) MetaboAnalyst pathway analysis for metabolites affected in RmHV1-3 or RmAD1-3 cultured with HNCO in the lid of agar dish. (E) Representative image of RmHV1-3 cultured with diisocyanate-containing glue or corn starch–based diisocyanate-negative glue placed into the lid of the agar dish and cultured for 48 hours. (F) Representative image of RmHV1-3 cultured with diisocyanate-containing glue placed into the lid of the agar dish and cultured for 48 hours in ambient or N2 deprivation conditions. (G and H) MetaboAnalyst pathway analysis for metabolites affected in RmHV1-3 cultured with diisocyanate-containing glue (G) or polyurethane-based glue (H) in the lid of agar dish. (I) Representative image of RmHV1-3 and RmAD1-3 cultured with polyurethane-based glue placed into the lid of the agar dish and cultured for 48 hours. (J) Summary table of RmHV1-3 pathways affected by N2 deprivation compared to pathways affected by indicated challenges along with fold change of total annotated ceramide containing metabolites; green indicates pathway affected per MetaboAnalyst, while dark green indicates significant impact and gray indicates pathway not affected. All images in (A), (E), (F), and (I) were taken at ×1.25 magnification. Data shown are representative of three or more independent experiments and shown as means ± SEM. *P < 0.05 and **P < 0.01 as determined by ANOVA.
Fig. 6.. Clinical results from R. mucosa…
Fig. 6.. Clinical results from R. mucosa treatment were influenced by TDI exposure.
Patients were treated with topical R. mucosa (FB-401) or placebo (sucrose in water) thrice weekly for 16 weeks before a 4-week washout. Two weeks before treatment, topical corticosteroids (TCS) were stopped and a minimally dysbiotic moisturizer regiment was initiated. (A) Example images from participants. (B) ITT analysis for those achieving an IGA of 0 and 1 (C) and percent change in EASI during active treatment. (D) For those completing the washout phase, percent change between week 16 and week 20 as a percentage of enrollment values is shown for EASI, BSA, and pruritis NRS (itch); each dot represents one participant. (E to L) For those completing the entire 20-week trial without topical steroids, raw EASI change (E), the proportion achieving 90% improvement in EASI (EASI90; F), and EASI75 (G). (H to L) Post hoc stratification of the completer population results by study sites’ historical TDI exposure as above the mean (>avg) or below for IGA0-1 (H), raw EASI (I), EASI90 (J), EASI75 (K), and pruritus (L); significance indicated for FB-401 in above or below average TDI zip codes against matched control unless indicated. Data are shown as proportion (B, F to H, J, and K) and means + SD (C, D, I, and L). ns, not significant, +P < 0.1; *P < 0.05; **P < 0.01 as determined by ANOVA (C and D), rANOVA (E, I, and L), or chi-square (B, F to H, J, and K).
Fig. 7.. S. cohnii improves outcomes in…
Fig. 7.. S. cohnii improves outcomes in mouse models of AD via steroid metabolism.
(A) Mice treated in the MC903 model of AD before treatment with either diluent or S. cohnii. (B) Segmented MALDI-MSI performed on mouse ears. (C) MetaboAnalyst pathways affected on mouse ears treated with S. cohnii. (D) Summary table of MetaboAnalyst pathways differentiating isolates of Staphylococcus spp. from healthy volunteers (Staph HV) versus isolates of S. aureus from patients with AD (Staph AD); green indicates that the pathway was affected, dark green indicates significant impact, and gray indicates not affected.
Fig. 8.. TDI and HNCO disrupt beneficial…
Fig. 8.. TDI and HNCO disrupt beneficial pathways in S. cohnii.
(A) Percent change in growth area for RmHV1-3, RmAD1-3, Staph HV1-3, and Staph AD1-3 as quantified from images shown in Figs. 1I, 2C, 4D, 5 (A, E, F, and I), and 8 (B to D and G). Significance is indicated versus Staph AD group. Impacts of NOx on Staph taken from (62). (B to D) Representative image of Staph HV1-3 and Staph AD1-3 cultured with TDI (B), HNCO (C), or polyurethane-containing glue (D). (E) Change in mouse ear thickness in MC903 mouse model after treatment with S. cohnii grown under ambient conditions or with exposure to TDI or HNCO. (F) Intensity levels of LysPG identified by mass/charge ratio and collisional cross section for the three isolates of StaphHV and three isolates from StaphAD. (G and H) Representative images (G) and non-metric multi-dimensional scaling (NMDS) plots (H) for S. cohnii and RmHV1 exposed to TDI with and without pretreatment with lysine (Lys) supplementation. (I) Change in total annotated ceramides with exposure to TDI versus ambient for RmHV with and without Lys supplementation pretreatment. All images were taken at ×1.25 magnification. Data are representative of two or more independent experiments and are displayed as means ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 as determined by ANOVA.

References

    1. A. Paller, J. C. Jaworski, E. L. Simpson, M. Boguniewicz, J. J. Russell, J. K. Block, S. Tofte, J. D. Dunn, S. R. Feldman, A. R. Clark, G. Schwartz, L. F. Eichenfield, Major comorbidities of atopic dermatitis: Beyond allergic disorders. Am. J. Clin. Dermatol. 19, 821–838 (2018).
    1. H. A. Hadi, A. I. Tarmizi, K. A. Khalid, M. Gajdács, A. Aslam, S. Jamshed, The epidemiology and global burden of atopic dermatitis: A narrative review. Life (Basel) 11, 936 (2021).
    1. C. S. Benn, M. Melbye, J. Wohlfahrt, B. Björkstén, P. Aaby, Cohort study of sibling effect, infectious diseases, and risk of atopic dermatitis during first 18 months of life. BMJ 328, 1223 (2004).
    1. W. Karmaus, C. Botezan, Does a higher number of siblings protect against the development of allergy and asthma? A review. J. Epidemiol. Commun. Health 56, 209–217 (2002).
    1. Y. Wang, K. J. Allen, N. H. A. Suaini, R. L. Peters, A.-L. Ponsonby, J. J. Koplin, Asian children living in Australia have a different profile of allergy and anaphylaxis than Australian-born children: A state-wide survey. Clin. Exp. Allergy 48, 1317–1324 (2018).
    1. E. H. Tham, E. X. L. Loo, Y. Zhu, L. P.-C. Shek, Effects of migration on allergic diseases. Int. Arch. Allergy Immunol. 178, 128–140 (2019).
    1. J. A. Odhiambo, H. C. Williams, T. O. Clayton, C. F. Robertson, M. I. Asher; ISAAC Phase Three Study Group , Global variations in prevalence of eczema symptoms in children from ISAAC Phase Three. J. Allergy Clin. Immunol. 124, 1251–1258.e23 (2009).
    1. I. A. Myles, C. R. Castillo, K. D. Barbian, K. Kanakabandi, K. Virtaneva, E. Fitzmeyer, M. Paneru, F. Otaizo-Carrasquero, T. G. Myers, T. E. Markowitz, I. N. Moore, X. Liu, M. Ferrer, Y. Sakamachi, S. Garantziotis, M. Swamydas, M. S. Lionakis, E. D. Anderson, N. J. Earland, S. Ganesan, A. A. Sun, J. R. E. Bergerson, R. A. Silverman, M. Petersen, C. A. Martens, S. K. Datta, Therapeutic responses to Roseomonas mucosa in atopic dermatitis may involve lipid-mediated TNF-related epithelial repair. Sci. Transl. Med. 12, eaaz8631 (2020).
    1. I. A. Myles, N. J. Earland, E. D. Anderson, I. N. Moore, M. D. Kieh, K. W. Williams, A. Saleem, N. M. Fontecilla, P. A. Welch, D. A. Darnell, L. A. Barnhart, A. A. Sun, G. Uzel, S. K. Datta, First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis. JCI Insight 3, e120608 (2018).
    1. L. Li, Z. Han, X. Niu, G. Zhang, Y. Jia, S. Zhang, C. He, Probiotic supplementation for prevention of atopic dermatitis in infants and children: A systematic review and meta-analysis. Am. J. Clin. Dermatol. 20, 367–377 (2019).
    1. T. Nakatsuji, T. R. Hata, Y. Tong, J. Y. Cheng, F. Shafiq, A. M. Butcher, S. S. Salem, S. L. Brinton, A. K. Rudman Spergel, K. Johnson, B. Jepson, A. Calatroni, G. David, M. Ramirez-Gama, P. Taylor, D. Y. M. Leung, R. L. Gallo, Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat. Med. 27, 700–709 (2021).
    1. J. Mashiah, T. Karady, N. Fliss-Isakov, E. Sprecher, D. Slodownik, O. Artzi, L. Samuelov, E. Ellenbogen, A. Godneva, E. Segal, N. Maharshak, Clinical efficacy of fecal microbial transplantation treatment in adults with moderate-to-severe atopic dermatitis. Immun. Inflamm. Dis. 10, e570 (2021).
    1. T. Kobayashi, M. Glatz, K. Horiuchi, H. Kawasaki, H. Akiyama, D. H. Kaplan, H. H. Kong, M. Amagai, K. Nagao, Dysbiosis and Staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity 42, 756–766 (2015).
    1. I. A. Myles, K. W. Williams, J. D. Reckhow, M. L. Jammeh, N. B. Pincus, I. Sastalla, D. Saleem, K. D. Stone, S. K. Datta, Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight 1, e86955 (2016).
    1. I. A. Myles, J. D. Reckhow, K. W. Williams, I. Sastalla, K. M. Frank, S. K. Datta, A method for culturing Gram-negative skin microbiota. BMC Microbiol. 16, 60 (2016).
    1. C. R. Castillo, M. E. Alishahedani, P. Gough, P. P. Chaudhary, M. Yadav, J. Matriz, I. A. Myles, Assessing the effects of common topical exposures on skin bacteria associated with atopic dermatitis. Skin Health Dis. 1, e41 (2021).
    1. J. van Smeden, J. A. Bouwstra, Stratum corneum lipids: Their role for the skin barrier function in healthy subjects and atopic dermatitis patients. Curr. Probl. Dermatol. 49, 8–26 (2016).
    1. A. Shea, M. Wolcott, S. Daefler, D. A. Rozak, Biolog phenotype microarrays. Methods Mol. Biol. 881, 331–373 (2012).
    1. Y. Lai, A. L. Cogen, K. A. Radek, H. J. Park, D. T. Mac Leod, A. Leichtle, A. F. Ryan, A. D. Nardo, R. L. Gallo, Activation of TLR2 by a small molecule produced by Staphylococcus epidermidis increases antimicrobial defense against bacterial skin infections. J. Invest. Dermatol. 130, 2211–2221 (2010).
    1. H. D. Graven, R. F. Keeling, S. C. Piper, P. K. Patra, B. B. Stephens, S. C. Wofsy, L. R. Welp, C. Sweeney, P. P. Tans, J. J. Kelley, B. C. Daube, E. A. Kort, G. W. Santoni, J. D. Bent, Enhanced seasonal exchange of CO2 by northern ecosystems since 1960. Science 341, 1085–1089 (2013).
    1. A. B. Fleischer Jr., Atopic dermatitis: The relationship to temperature and seasonality in the United States. Int. J. Dermatol. 58, 465–471 (2019).
    1. V. Sojo, B. Herschy, A. Whicher, E. Camprubi, N. Lane, The origin of life in alkaline hydrothermal vents. Astrobiology 16, 181–197 (2016).
    1. E. Proksch, pH in nature, humans and skin. J. Dermatol. 45, 1044–1052 (2018).
    1. K. Ahn, The role of air pollutants in atopic dermatitis. J. Allergy Clin. Immunol. 134, 993–999 (2014).
    1. B. Tury, D. Pemberton, R. E. Bailey, Fate and potential environmental effects of methylenediphenyl diisocyanate and toluene diisocyanate released into the atmosphere. J. Air Waste Manag. Assoc. 53, 61–66 (2003).
    1. S. M. Langan, P. Silcocks, H. C. Williams, What causes flares of eczema in children? Br. J. Dermatol. 161, 640–646 (2009).
    1. J. H. Lee, J. Suh, E. H. Kim, J. B. Cho, H. Y. Park, J. Kim, K. Ahn, H. K. Cheong, S.-I. Lee, Surveillance of home environment in children with atopic dermatitis: A questionnaire survey. Asia Pac. Allergy 2, 59–66 (2012).
    1. S.-J. Yi, C. Shon, K.-D. Min, H.-C. Kim, J.-H. Leem, H.-J. Kwon, S. Hong, K. S. Kim, S.-Y. Kim, Association between exposure to traffic-related air pollution and prevalence of allergic diseases in children, Seoul, Korea. Biomed. Res. Int. 2017, 4216107 (2017).
    1. C. Pénard-Morand, C. Raherison, D. Charpin, C. Kopferschmitt, F. Lavaud, D. Caillaud, I. Annesi-Maesano, Long-term exposure to close-proximity air pollution and asthma and allergies in urban children. Eur. Respir. J. 36, 33–40 (2010).
    1. P. Kathuria, J. I. Silverberg, Association of pollution and climate with atopic eczema in US children. Pediatr. Allergy Immunol. 27, 478–485 (2016).
    1. Y. Miyake, K. Tanaka, H. Fujiwara, Y. Mitani, H. Ikemi, S. Sasaki, Y. Ohya, Y. Hirota, Residential proximity to main roads during pregnancy and the risk of allergic disorders in Japanese infants: The Osaka Maternal and Child Health Study. Pediatr. Allergy Immunol. 21, 22–28 (2010).
    1. M. Mubanga, C. Lundholm, B. M. D’Onofrio, M. Stratmann, A. Hedman, C. Almqvist, Association of early life exposure to antibiotics with risk of atopic dermatitis in Sweden. JAMA Netw. Open 4, e215245 (2021).
    1. EPA, in Greenhouse Gas Emissions (EPA, 2021).
    1. J. Tan, C. McKenzie, P. J. Vuillermin, G. Goverse, C. G. Vinuesa, R. E. Mebius, L. Macia, C. R. Mackay, Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 15, 2809–2824 (2016).
    1. D. Johnson, V. Letchumanan, S. Thurairajasingam, L.-H. Lee, A revolutionizing approach to autism spectrum disorder using the microbiome. Nutrients 12, 1983 (2020).
    1. B. Golofast, K. Vales, The connection between microbiome and schizophrenia. Neurosci. Biobehav. Rev. 108, 712–731 (2020).
    1. X. Liang, C. Ou, J. Zhuang, J. Li, F. Zhang, Y. Zhong, Y. Chen, Interplay between skin microbiota dysbiosis and the host immune system in psoriasis: Potential pathogenesis. Front. Immunol. 12, 764384 (2021).
    1. Toluene diisocyanates. IARC Monogr. Eval. Carcinog. Risks Hum. 71, 865–879 (1999).
    1. A. V. Wisnewski, J. Liu, C. A. Redlich, Antigenic changes in human albumin caused by reactivity with the occupational allergen diphenylmethane diisocyanate. Anal. Biochem. 400, 251–258 (2010).
    1. P. Mydel, Z. Wang, M. Brisslert, A. Hellvard, L. E. Dahlberg, S. L. Hazen, M. Bokarewa, Carbamylation-dependent activation of T cells: A novel mechanism in the pathogenesis of autoimmune arthritis. J. Immunol. 184, 6882–6890 (2010).
    1. R. P. Fadadu, B. Grimes, N. P. Jewell, J. Vargo, A. T. Young, K. Abuabara, J. R. Balmes, M. L. Wei, Association of wildfire air pollution and health care use for atopic dermatitis and itch. JAMA Dermatol. 157, 658–666 (2021).
    1. O. Herbarth, G. J. Fritz, M. Rehwagen, M. Richter, S. Röder, U. Schlink, Association between indoor renovation activities and eczema in early childhood. Int. J. Hyg. Environ. Health 209, 241–247 (2006).
    1. J. M. Roberts, P. R. Veres, A. K. Cochran, C. Warneke, I. R. Burling, R. J. Yokelson, B. Lerner, J. B. Gilman, W. C. Kuster, R. Fall, J. de Gouw, Isocyanic acid in the atmosphere and its possible link to smoke-related health effects. Proc. Natl. Acad. Sci. U.S.A. 108, 8966–8971 (2011).
    1. L. Bengtstrom, M. Salden, A. A. Stec, The role of isocyanates in fire toxicity. Fire Sci. Rev. 5, 4 (2016).
    1. M. D. Leslie, M. Ridoli, J. G. Murphy, N. Borduas-Dedekind, Isocyanic acid (HNCO) and its fate in the atmosphere: A review. Environ. Sci. Process. Impacts 21, 793–808 (2019).
    1. K. K. McCleary, K. Barta, L. Butler, K. Capozza, S. Eftekhari, K. Kelley, W.S. Begolka, K. Tullos, and T. Winders. The More Than Skin Deep "Voice of the Patient" Report (2020).
    1. Y. Ito, T. Sasaki, Y. Li, T. Tanoue, Y. Sugiura, A. N. Skelly, W. Suda, Y. Kawashima, N. Okahashi, E. Watanabe, H. Horikawa, A. Shiohama, R. Kurokawa, E. Kawakami, H. Iseki, H. Kawasaki, Y. Iwakura, A. Shiota, L. Yu, J. Hisatsune, H. Koseki, M. Sugai, M. Arita, O. Ohara, T. Matsui, M. Suematsu, M. Hattori, K. Atarashi, M. Amagai, K. Honda, Staphylococcus cohnii is a potentially biotherapeutic skin commensal alleviating skin inflammation. Cell Rep. 35, 109052 (2021).
    1. Y. Zheng, R. L. Hunt, A. E. Villaruz, E. L. Fisher, R. Liu, Q. Liu, G. Y. C. Cheung, M. Li, M. Otto, Commensal Staphylococcus epidermidis contributes to skin barrier homeostasis by generating protective ceramides. Cell Host Microbe 30, 301–313.e9 (2022).
    1. I. A. Myles, I. N. Moore, C. R. Castillo, S. K. Datta, Differing virulence of healthy skin commensals in mouse models of infection. Front. Cell. Infect. Microbiol. 8, 451 (2018).
    1. Y. Oku, K. Kurokawa, N. Ichihashi, K. Sekimizu, Characterization of the Staphylococcus aureus mprF gene, involved in lysinylation of phosphatidylglycerol. Microbiology (Reading) 150, 45–51 (2004).
    1. S. Delanghe, J. R. Delanghe, R. Speeckaert, W. Van Biesen, M. M. Speeckaert, Mechanisms and consequences of carbamoylation. Nat. Rev. Nephrol. 13, 580–593 (2017).
    1. B. C. Martel, P. Lovato, W. Baumer, T. Olivry, Translational animal models of atopic dermatitis for preclinical studies. Yale J. Biol. Med. 90, 389–402 (2017).
    1. D. Hayes-Green, B. P. Love, The Groundwater Approach: Building a Practical Understanding of Structural Racism (The Racial Equity Institute, 2018).
    1. S. A. Smits, J. Leach, E. D. Sonnenburg, C. G. Gonzalez, J. S. Lichtman, G. Reid, R. Knight, A. Manjurano, J. Changalucha, J. E. Elias, M. G. Dominguez-Bello, J. L. Sonnenburg, Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 357, 802–806 (2017).
    1. G. I. Perez Perez, Z. Gao, R. Jourdain, J. Ramirez, F. Gany, C. Clavaud, J. Demaude, L. Breton, M. J. Blaser, Body site is a more determinant factor than human population diversity in the healthy skin microbiome. PLOS ONE 11, e0151990 (2016).
    1. T. Sakai, N. Herrmann, L. Maintz, T. J. Nümm, T. Welchowski, R. A. Claus, M. H. Gräler, T. Bieber, Altered serum phospholipids in atopic dermatitis and association with clinical status. JID Innov. 2, 100092 (2021).
    1. S. Mihara, T. Shibamoto, The role of flavor and fragrance chemicals in TRPA1 (transient receptor potential cation channel, member A1) activity associated with allergies. Allergy Asthma Clin. Immunol. 11, 11 (2015).
    1. F. M. S. Badloe, S. De Vriese, K. Coolens, C. B. Schmidt-Weber, J. Ring, J. Gutermuth, I. K. Krohn, IgE autoantibodies and autoreactive T cells and their role in children and adults with atopic dermatitis. Clin. Transl. Allergy 10, 34 (2020).
    1. H. S. Park, S. K. Lee, Y. M. Lee, S. S. Kim, D. H. Nahm, Longitudinal study of specific antibodies to toluene diisocyanate (TDI)-human serum albumin (HSA) conjugate in patients with TDI-induced asthma. Korean J. Intern. Med. 17, 249–251 (2002).
    1. M. Parker-Lalomio, K. McCann, J. Piorkowski, S. Freels, V. W. Persky, Prenatal exposure to polychlorinated biphenyls and asthma, eczema/hay fever, and frequent ear infections. J. Asthma 55, 1105–1115 (2018).
    1. B. E. Kim, J. Kim, E. Goleva, E. Berdyshev, J. Lee, K. A. Vang, U. H. Lee, S. Y. Han, S. Leung, C. F. Hall, N.-R. Kim, I. Bronova, E. J. Lee, H.-R. Yang, D. Y. Leung, K. Ahn, Particulate matter causes skin barrier dysfunction. JCI Insight 6, e145185 (2021).
    1. X. Janvier, S. Alexandre, A. M. Boukerb, D. Souak, O. Maillot, M. Barreau, F. Gouriou, C. Grillon, M. G. J. Feuilloley, A. Groboillot, Deleterious effects of an air pollutant (NO2) on a selection of commensal skin bacterial strains, potential contributor to dysbiosis? Front. Microbiol. 11, 591839 (2020).
    1. M.-T. Lee, C.-C. Wu, C.-Y. Ou, J.-C. Chang, C.-A. Liu, C.-L. Wang, H. Chuang, H.-C. Kuo, T.-Y. Hsu, C.-P. Chen, K. D. Yang, A prospective birth cohort study of different risk factors for development of allergic diseases in offspring of non-atopic parents. Oncotarget 8, 10858–10870 (2017).
    1. C. S. C. Tan-Lim, N. A. R. Esteban-Ipac, J. B. V. Mantaring III, E. Chan Shih Yen, M. S. T. Recto, O. T. Sison, M. M. Alejandria, Comparative effectiveness of probiotic strains for the treatment of pediatric atopic dermatitis: A systematic review and network meta-analysis. Pediatr. Allergy Immunol. 32, 124–136 (2021).
    1. B. M. Ray, Performance auditing of an in vitro air pollution exposure system. Toxicology 10, 615–624 (1998).
    1. K. Urban, S. Chu, R. L. Giesey, S. Mehrmal, P. Uppal, N. Nedley, G. R. Delost, The global, regional, and national burden of atopic dermatitis in 195 countries and territories: An ecological study from the Global Burden of Disease Study 2017. JAAD Int. 2, 12–18 (2021).
    1. Y. Guo, P. Li, J. Tang, X. Han, X. Zou, G. Xu, Z. Xu, F. Wei, Q. Liu, M. Wang, F. Xiao, W. Zong, C. Shen, J. Li, J. Liu, Y. Luo, J. Chang, N. Sheng, C. Dong, D. Zhang, X. Dai, J. Zhou, C. Meng, H. Niu, X. Shi, X. Zhang, J. Xiang, H. Xu, Q. Ran, Y. Zhou, M. Li, H. Zhang, R. Cheng, X. Gao, H. Wang, H. Gu, L. Ma, Z. Yao, Prevalence of atopic dermatitis in chinese children aged 1-7 ys. Sci. Rep. 6, 29751 (2016).
    1. CDC, Toluene diisocyanate (TDI) and toluenediamine (TDA): Evidence of carcinogenicity (Current Intelligence Bulletin 53, CDC, 1989).
    1. L. Fernandez, Toluene diisocyanate demand worldwide from 2011 to 2020 with a forecast for 2022. Statista (2021).
    1. L. Fernandez, Methylene diphenyl diisocyanate production in the United States from 1990 to 2019. Statista (2021).
    1. Markit, Isocyanates (Process Economics Program Report 1B, 1973).
    1. MerchantResearch, Toluene diisocyanate (TDI): 2021 World market outlook up to 2030 (with COVID-19 impact estimation). Research and Markets, (2021).
    1. W. Yen, How long and how far do adults travel and will adults travel for primary care? (Washington State Health Services Research, 2013).
    1. J. Friedman, T. Hastie, R. Tibshirani, Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010).
    1. M. N. Wright, A. Ziegler, ranger: A fast implementation of random forests for high dimensional data in C++ and R. J. Stat. Softw. 77, 1–17 (2017).
    1. B. M. Greenwell, pdp: An R package for constructing partial dependence plots. R J. 9, 421–436 (2017).

Source: PubMed

3
Subscribe