Hostname: page-component-77c89778f8-7drxs Total loading time: 0 Render date: 2024-07-17T07:47:42.795Z Has data issue: false hasContentIssue false

Developmental Origins of Health and Disease: Impact of environmental dust exposure in modulating microbiome and its association with non-communicable diseases

Published online by Cambridge University Press:  15 June 2020

Delicia Shu-Qin Ooi
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
Cheryl Pei-Ting Tan
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
Michelle Jia-Yu Tay
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
Siong Gim Ong
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
Elizabeth Huiwen Tham
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore
Kewin Tien Ho Siah
Affiliation:
Division of Gastroenterology & Hepatology, University Medicine Cluster, National University Hospital, Singapore, Singapore Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
Johan Gunnar Eriksson
Affiliation:
Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore University of Helsinki, and Helsinki University Hospital, Helsinki, Finland Folkhälsan Research Center, Helsinki, Finland
Keith M. Godfrey
Affiliation:
NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK Medical Research Council Lifecourse Epidemiology Unit, Southampton, UK
Lynette Pei-Chi Shek
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Khoo Teck Puat-National University Children’s Medical Institute, National University Hospital, National University Health System, Singapore, Singapore Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
Evelyn Xiu-Ling Loo*
Affiliation:
Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
*
Address for correspondence: Evelyn Xiu-Ling Loo, Singapore Institute for Clinical Sciences, Brenner Centre for Molecular Medicine, 30 Medical Drive, Singapore117609, Singapore. E-mail: evelyn_loo@sics.a-star.edu.sg

Abstract

Non-communicable diseases (NCDs) including obesity, diabetes, and allergy are chronic, multi-factorial conditions that are affected by both genetic and environmental factors. Over the last decade, the microbiome has emerged as a possible contributor to the pathogenesis of NCDs. Microbiome profiles were altered in patients with NCDs, and shift in microbial communities was associated with improvement in these health conditions. Since the genetic component of these diseases cannot be altered, the ability to manipulate the microbiome holds great promise for design of novel therapies in the prevention and treatment of NCDs. Together, the Developmental Origins of Health and Disease concept and the microbial hypothesis propose that early life exposure to environmental stimuli will alter the development and composition of the human microbiome, resulting in health consequences. Recent studies indicated that the environment we are exposed to in early life is instrumental in shaping robust immune development, possibly through modulation of the human microbiome (skin, airway, and gut). Despite much research into human microbiome, the origin of their constituent microbiota remains unclear. Dust (also known as particulate matter) is a key determinant of poor air quality in the modern urban environment. It is ubiquitous and serves as a major source and reservoir of microbial communities that modulates the human microbiome, contributing to health and disease. There are evidence that reported significant associations between environmental dust and NCDs. In this review, we will focus on the impact of dust exposure in shaping the human microbiome and its possible contribution to the development of NCDs.

Type
Review
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2020

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Gouda, HN, Charlson, F, Sorsdahl, K, et al. Burden of non-communicable diseases in sub-Saharan Africa, 1990–2017: results from the Global Burden of Disease Study 2017. Lancet Glob Health. 2019; 7 (10), e1375e1387.CrossRefGoogle ScholarPubMed
World Health Organization (WHO). Global Action Plan for the Prevention and Control of NCDs 2013–2020. WHO Website: https://www.who.int/nmh/events/ncd_action_plan/en/ Accessed April 1, 2020.Google Scholar
Ogoina, D, Onyemelukwe, GC. The role of infections in the emergence of non-communicable diseases (NCDs): compelling needs for novel strategies in the developing world. J Infect Public Health. 2009; 2 (1), 1429.CrossRefGoogle ScholarPubMed
Ober, C, Yao, TC. The genetics of asthma and allergic disease: a 21st century perspective. Immunol Rev. 2011; 242 (1), 1030.CrossRefGoogle ScholarPubMed
Temelkova-Kurktschiev, T, Stefanov, T. Lifestyle and genetics in obesity and type 2 diabetes. Exp Clin Endocrinol Diabetes. 2012; 120 (1), 16.Google ScholarPubMed
Kho, ZY, Lal, SK. The human gut microbiome – a potential controller of wellness and disease. Front Microbiol. 2018; 9, 1835.CrossRefGoogle ScholarPubMed
Ley, RE, Turnbaugh, PJ, Klein, S, Gordon, JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006; 444 (7122), 10221023.CrossRefGoogle ScholarPubMed
Sedighi, M, Razavi, S, Navab-Moghadam, F, et al. Comparison of gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microb Pathog. 2017; 111, 362369.CrossRefGoogle ScholarPubMed
Kong, HH, Oh, J, Deming, C, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012; 22 (5), 850859.CrossRefGoogle ScholarPubMed
Myles, IA, Williams, KW, Reckhow, JD, et al. Transplantation of human skin microbiota in models of atopic dermatitis. JCI Insight. 2016; 1 (10).CrossRefGoogle ScholarPubMed
Godfrey, KM, Costello, PM, Lillycrop, KA. The developmental environment, epigenetic biomarkers and long-term health. J Dev Orig Health Dis. 2015; 6 (5), 399406.CrossRefGoogle ScholarPubMed
von Mutius, E. The microbial environment and its influence on asthma prevention in early life. J Allergy Clin Immunol. 2016; 137 (3), 680689.CrossRefGoogle ScholarPubMed
Tamburini, S, Shen, N, Wu, HC, Clemente, JC. The microbiome in early life: implications for health outcomes. Nat Med. 2016; 22 (7), 713.CrossRefGoogle ScholarPubMed
Mueller, NT, Bakacs, E, Combellick, J, Grigoryan, Z, Dominguez-Bello, MG. The infant microbiome development: mom matters. Trends Mol Med. 2015; 21 (2), 109117.CrossRefGoogle ScholarPubMed
Ege, MJ, Mayer, M, Normand, A-C, et al. Exposure to environmental microorganisms and childhood asthma. N Engl J Med. 2011; 364 (8), 701709.CrossRefGoogle ScholarPubMed
O’Connor, GT, Lynch, SV, Bloomberg, GR, et al. Early-life home environment and risk of asthma among inner-city children. J Allergy Clin Immunol. 2018; 141 (4), 14681475.CrossRefGoogle ScholarPubMed
Strachan, DP. Hay fever, hygiene, and household size. BMJ. 1989; 299 (6710), 12591260.CrossRefGoogle ScholarPubMed
Shreiner, A, Huffnagle, GB, Noverr, MC. The “Microflora Hypothesis” of allergic disease. Adv Exp Med Biol. 2008; 635, 113134.CrossRefGoogle ScholarPubMed
Gensollen, T, Blumberg, RS. Correlation between early-life regulation of the immune system by microbiota and allergy development. J Allergy Clin Immunol. 2017; 139 (4), 10841091.CrossRefGoogle ScholarPubMed
Yu, J, Liu, X, Li, Y, et al. Maternal exposure to farming environment protects offspring against allergic diseases by modulating the neonatal TLR-Tregs-Th axis. Clin Trans Allergy. 2018; 8, 34.CrossRefGoogle ScholarPubMed
Douwes, J, Cheng, S, Travier, N, et al. Farm exposure in utero may protect against asthma, hay fever and eczema. Eur Respir J. 2008; 32 (3), 603611.CrossRefGoogle ScholarPubMed
Dogra, S, Sakwinska, O, Soh, SE, et al. Dynamics of infant gut microbiota are influenced by delivery mode and gestational duration and are associated with subsequent adiposity. mBio. 2015; 6 (1), e02419-14.CrossRefGoogle ScholarPubMed
Hagan, T, Cortese, M, Rouphael, N, et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell. 2019; 178 (6), 13131328. e1313.CrossRefGoogle ScholarPubMed
Akagawa, S, Tsuji, S, Onuma, C, et al. Effect of delivery mode and nutrition on gut microbiota in neonates. Ann Nutr Metab. 2019; 74 (2), 132139.CrossRefGoogle ScholarPubMed
Crinnion, W. Particulate matter is a surprisingly common contributor to disease. Integr Med. 2017; 16 (4), 812.Google ScholarPubMed
Inyang, HI, Bae, S. Impacts of dust on environmental systems and human health. J Hazard Mater. 2006; 132 (1), vvi.CrossRefGoogle ScholarPubMed
US Environmental Protection Agency (EPA). Particulate matter (PM) basics. EPA Web site. https://www.epa.gov/pm-pollution/particulate-matter-pm-basics. Updated November 14, 2018. Accessed March 26, 2020.Google Scholar
Hime, NJ, Marks, GB, Cowie, CT. A comparison of the health effects of ambient particulate matter air pollution from five emission sources. Int J Environ Res Public Health. 2018; 15 (6).CrossRefGoogle ScholarPubMed
Apte, K, Salvi, S. Household air pollution and its effects on health. F1000Research. 2016; 5. F1000 Faculty Rev-2593.CrossRefGoogle ScholarPubMed
Kheirbek, I, Haney, J, Douglas, S, Ito, K, Matte, T. The contribution of motor vehicle emissions to ambient fine particulate matter public health impacts in New York City: a health burden assessment. Environ Health. 2016; 15 (1), 89.CrossRefGoogle ScholarPubMed
Shan, Y, Wu, W, Fan, W, Haahtela, T, Zhang, G. House dust microbiome and human health risks. Int Microbiol. 2019; 22 (3), 297304.CrossRefGoogle ScholarPubMed
US Environmental Protection Agency (EPA). Exposure assessment tools by routes. EPA Web site: https://www.epa.gov/expobox/exposure-assessment-tools-routes-ingestion. Updated June 25, 2018. Accessed March 28, 2020.Google Scholar
Liao, J, Yu, H, Xia, W, et al. Exposure to ambient fine particulate matter during pregnancy and gestational weight gain. Environ Int. 2018; 119, 407412.CrossRefGoogle ScholarPubMed
Kim, YJ, Song, IG, Kim, KN, et al. Maternal exposure to particulate matter during pregnancy and adverse birth outcomes in the Republic of Korea. Int J Environ Res Public Health. 2019; 16 (4).Google ScholarPubMed
Deng, Q, Lu, C, Yu, Y, Li, Y, Sundell, J, Norbäck, D. Early life exposure to traffic-related air pollution and allergic rhinitis in preschool children. Respir Med. 2016; 121, 6773.CrossRefGoogle ScholarPubMed
Lynch, SV, Wood, RA, Boushey, H, et al. Effects of early-life exposure to allergens and bacteria on recurrent wheeze and atopy in urban children. J Allergy Clin Immunol. 2014; 134 (3), 593601. e512.CrossRefGoogle ScholarPubMed
Bacharier, LB, Beigelman, A, Calatroni, A, et al. Longitudinal phenotypes of respiratory health in a high-risk urban birth cohort. Am J Respir Crit Care Med. 2019; 199 (1), 7182.CrossRefGoogle Scholar
Loo, EXL, Chew, LJM, Zulkifli, AB, et al. Comparison of microbiota and allergen profile in house dust from homes of allergic and non-allergic subjects – results from the GUSTO study. World Allergy Organ J. 2018; 11 (1), 37.CrossRefGoogle ScholarPubMed
Park, JH, Gold, DR, Spiegelman, DL, Burge, HA, Milton, DK. House dust endotoxin and wheeze in the first year of life. Am J Respir Crit Care Med. 2001; 163 (2), 322328.CrossRefGoogle ScholarPubMed
Songjinda, P, Nakayama, J, Tateyama, A, et al. Differences in developing intestinal microbiota between allergic and non-allergic infants: a pilot study in Japan. Biosci Biotechnol Biochem. 2007; 71 (9), 23382342.CrossRefGoogle ScholarPubMed
Pakarinen, J, Hyvarinen, A, Salkinoja-Salonen, M, et al. Predominance of Gram-positive bacteria in house dust in the low-allergy risk Russian Karelia. Environ Microbiol. 2008; 10 (12), 33173325.CrossRefGoogle ScholarPubMed
Metzger, KB, Tolbert, PE, Klein, M, et al. Ambient air pollution and cardiovascular emergency department visits. Epidemiology. 2004; 15 (1), 4656.CrossRefGoogle ScholarPubMed
Matz, CJ, Egyed, M, Hocking, R, Seenundun, S, Charman, N, Edmonds, N. Human health effects of traffic-related air pollution (TRAP): a scoping review protocol. Sys Rev. 2019; 8 (1), 223.CrossRefGoogle ScholarPubMed
Costa, LG, Cole, TB, Coburn, J, Chang, Y-C, Dao, K, Roqué, PJ. Neurotoxicity of traffic-related air pollution. NeuroToxicology. 2017; 59, 133139.CrossRefGoogle ScholarPubMed
Newman, NC, Ryan, PH, Huang, B, Beck, AF, Sauers, HS, Kahn, RS. Traffic-related air pollution and asthma hospital readmission in children: a longitudinal cohort study. J Pediatr. 2014; 164 (6), 13961402. e1391.CrossRefGoogle ScholarPubMed
Jerrett, M, McConnell, R, Wolch, J, et al. Traffic-related air pollution and obesity formation in children: a longitudinal, multilevel analysis. Environ Health. 2014; 13, 49.CrossRefGoogle ScholarPubMed
Rundle, A, Hoepner, L, Hassoun, A, et al. Association of childhood obesity with maternal exposure to ambient air polycyclic aromatic hydrocarbons during pregnancy. Am J Epidemiol. 2012; 175 (11), 11631172.CrossRefGoogle ScholarPubMed
Toledo-Corral, CM, Alderete, TL, Habre, R, et al. Effects of air pollution exposure on glucose metabolism in Los Angeles minority children. Pediatr Obes. 2018; 13 (1), 5462.CrossRefGoogle ScholarPubMed
Alderete, TL, Habre, R, Toledo-Corral, CM, et al. Longitudinal associations between ambient air pollution with insulin sensitivity, beta-cell function, and adiposity in Los Angeles Latino children. Diabetes. 2017; 66 (7), 17891796.CrossRefGoogle ScholarPubMed
Thiering, E, Cyrys, J, Kratzsch, J, et al. Long-term exposure to traffic-related air pollution and insulin resistance in children: results from the GINIplus and LISAplus birth cohorts. Diabetologia. 2013; 56 (8), 16961704.CrossRefGoogle ScholarPubMed
Weinmayr, G, Hennig, F, Fuks, K, et al. Long-term exposure to fine particulate matter and incidence of type 2 diabetes mellitus in a cohort study: effects of total and traffic-specific air pollution. Environ Health. 2015; 14, 53.CrossRefGoogle Scholar
Prescott, SL, Larcombe, DL, Logan, AC, et al. The skin microbiome: impact of modern environments on skin ecology, barrier integrity, and systemic immune programming. World Allergy Organ J. 2017; 10 (1), 29.CrossRefGoogle ScholarPubMed
Hanski, I, von Hertzen, L, Fyhrquist, N, et al. Environmental biodiversity, human microbiota, and allergy are interrelated. Proc Natl Acad Sci USA. 2012; 109 (21), 83348339.CrossRefGoogle ScholarPubMed
Tang, VH, Chang, BJ, Srinivasan, A, Mathaba, LT, Harnett, GB, Stewart, GA. Skin-associated Bacillus, staphylococcal and micrococcal species from the house dust mite, Dermatophagoides pteronyssinus and bacteriolytic enzymes. Exp Appl Acarol. 2013; 61 (4), 431447.CrossRefGoogle ScholarPubMed
Hanson, B, Zhou, Y, Bautista, EJ, et al. Characterization of the bacterial and fungal microbiome in indoor dust and outdoor air samples: a pilot study. Environ Sci Process Impacts. 2016; 18 (6), 713724.CrossRefGoogle ScholarPubMed
Chiller, K, Selkin, BA, Murakawa, GJ. Skin microflora and bacterial infections of the skin. J Invest Dermatol Symp Proc. 2001; 6 (3), 170174.CrossRefGoogle ScholarPubMed
Lange-Asschenfeldt, B, Marenbach, D, Lang, C, et al. Distribution of bacteria in the epidermal layers and hair follicles of the human skin. Skin Pharmacol Physiol. 2011; 24 (6), 305311.CrossRefGoogle ScholarPubMed
Robinson, CL, Baumann, LM, Romero, K, et al. Effect of urbanisation on asthma, allergy and airways inflammation in a developing country setting. Thorax. 2011; 66 (12), 10511057.CrossRefGoogle Scholar
Rodriguez, A, Vaca, M, Oviedo, G, et al. Urbanisation is associated with prevalence of childhood asthma in diverse, small rural communities in Ecuador. Thorax. 2011; 66 (12), 10431050.CrossRefGoogle ScholarPubMed
Kirjavainen, PV, Karvonen, AM, Adams, RI, et al. Farm-like indoor microbiota in non-farm homes protects children from asthma development. Nat Med. 2019; 25 (7), 10891095.CrossRefGoogle ScholarPubMed
Dyck, R, Karunanayake, C, Pahwa, P, et al. Prevalence, risk factors and co-morbidities of diabetes among adults in rural Saskatchewan: the influence of farm residence and agriculture-related exposures. BMC Public Health. 2013; 13, 7.CrossRefGoogle ScholarPubMed
Karkman, A, Lehtimaki, J, Ruokolainen, L. The ecology of human microbiota: dynamics and diversity in health and disease. Ann N Y Acad Sci. 2017; 1399 (1), 7892.CrossRefGoogle ScholarPubMed
Thimmappaiah Jagadeesh, A, Prakash, PY, Karthik Rao, N, Ramya, V. Culture characterization of the skin microbiome in Type 2 diabetes mellitus: a focus on the role of innate immunity. Diabetes Res Clin Pract. 2017; 134, 17.CrossRefGoogle ScholarPubMed
Gardiner, M, Vicaretti, M, Sparks, J, et al. A longitudinal study of the diabetic skin and wound microbiome. PeerJ. 2017; 5, e3543.CrossRefGoogle ScholarPubMed
Rood, KM, Buhimschi, IA, Jurcisek, JA, et al. Skin microbiota in obese women at risk for surgical site infection after cesarean delivery. Sci Rep. 2018; 8 (1), 8756.CrossRefGoogle ScholarPubMed
Dinwiddie, DL, Denson, JL, Kennedy, JL. Role of the airway microbiome in respiratory infections and asthma in children. Pediatr Allergy Immunol Pulmonol. 2018; 31 (4), 236240.CrossRefGoogle ScholarPubMed
Mariani, J, Favero, C, Spinazze, A, et al. Short-term particulate matter exposure influences nasal microbiota in a population of healthy subjects. Environ Res. 2018; 162, 119126.CrossRefGoogle Scholar
Birzele, LT, Depner, M, Ege, MJ, et al. Environmental and mucosal microbiota and their role in childhood asthma. Allergy. 2017; 72 (1), 109119.CrossRefGoogle ScholarPubMed
Abrahamsson, TR, Jakobsson, HE, Andersson, AF, Björkstén, B, Engstrand, L, Jenmalm, MC. Low diversity of the gut microbiota in infants with atopic eczema. J Allergy Clin Immunol. 2012; 129 (2), 434440. e432.CrossRefGoogle ScholarPubMed
Kraemer, JG, Ramette, A, Aebi, S, Oppliger, A, Hilty, M. Influence of pig farming on the human nasal microbiota: key role of airborne microbial communities. Appl Environ Microbiol. 2018; 84 (6), e02470-17.CrossRefGoogle ScholarPubMed
Shukla, SK, Ye, Z, Sandberg, S, Reyes, I, Fritsche, TR, Keifer, M. The nasal microbiota of dairy farmers is more complex than oral microbiota, reflects occupational exposure, and provides competition for staphylococci. PLoS One. 2017; 12 (8), e0183898.CrossRefGoogle ScholarPubMed
Li, N, He, F, Liao, B, Zhou, Y, Li, B, Ran, P. Exposure to ambient particulate matter alters the microbial composition and induces immune changes in rat lung. Respir Res. 2017; 18 (1), 143.CrossRefGoogle ScholarPubMed
Andersen, ZJ, Loft, S, Ketzel, M, et al. Ambient air pollution triggers wheezing symptoms in infants. Thorax. 2008; 63 (8), 710716.CrossRefGoogle ScholarPubMed
Brauer, M, Hoek, G, Smit, HA, et al. Air pollution and development of asthma, allergy and infections in a birth cohort. Eur Respir J. 2007; 29 (5), 879888.CrossRefGoogle Scholar
Moutsopoulos, NM, Konkel, JE. Tissue-specific immunity at the oral mucosal barrier. Trends Immunol 2018; 39 (4), 276287.CrossRefGoogle ScholarPubMed
Wilson, R, Jones-Otazo, H, Petrovic, S, et al. Revisiting dust and soil ingestion rates based on hand-to-mouth transfer. Human Ecol Risk Assess. 2013; 19 (1), 158188.CrossRefGoogle Scholar
Xue, J, Zartarian, V, Moya, J, et al. A meta-analysis of children’s hand-to-mouth frequency data for estimating nondietary ingestion exposure. J Expo Sci Environ Epidemiol. 2010; 20 (6), 536545.CrossRefGoogle Scholar
Moya, J, Phillips, L. A review of soil and dust ingestion studies for children. J Expo Sci Environ Epidemiol. 2014; 24 (6), 545554.CrossRefGoogle ScholarPubMed
Morman, SA, Plumlee, GS. Dust and human health. In: Knippertz, P, Stuut, J-BW, eds. Mineral Dust: A Key Player in the Earth System. Springer, Dordrecht; 2014, pp. 385409.Google Scholar
Olsen, I, Yamazaki, K. Can oral bacteria affect the microbiome of the gut? J Oral Microbiol. 2019; 11 (1), 1586422.CrossRefGoogle ScholarPubMed
Li, B, Ge, Y, Cheng, L, et al. Oral bacteria colonize and compete with gut microbiota in gnotobiotic mice. Int J Oral Sci. 2019; 11 (1), 10.CrossRefGoogle ScholarPubMed
Yang, Y, Cai, Q, Zheng, W, et al. Oral microbiome and obesity in a large study of low-income and African-American populations. J Oral Microbiol. 2019; 11 (1), 1650597.CrossRefGoogle Scholar
Mervish, NA, Hu, J, Hagan, LA, et al. Associations of the oral microbiota with obesity and menarche in inner city girls. J Child Obes. 2019; 4 (1).Google ScholarPubMed
Long, J, Cai, Q, Steinwandel, M, et al. Association of oral microbiome with type 2 diabetes risk. J Periodontal Res. 2017; 52 (3), 636643.CrossRefGoogle ScholarPubMed
Wang, RR, Xu, YS, Ji, MM, et al. Association of the oral microbiome with the progression of impaired fasting glucose in a Chinese elderly population. J Oral Microbiol. 2019; 11 (1), 1605789.CrossRefGoogle Scholar
Dzidic, M, Abrahamsson, TR, Artacho, A, Collado, MC, Mira, A, Jenmalm, MC. Oral microbiota maturation during the first 7 years of life in relation to allergy development. Allergy. 2018; 73 (10), 20002011.CrossRefGoogle ScholarPubMed
Ley, RE, Peterson, DA, Gordon, JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006; 124 (4), 837848.CrossRefGoogle ScholarPubMed
Kinross, JM, Darzi, AW, Nicholson, JK. Gut microbiome-host interactions in health and disease. Genome Med. 2011; 3 (3), 14.CrossRefGoogle ScholarPubMed
Liu, T, Chen, X, Xu, Y, et al. Gut microbiota partially mediates the effects of fine particulate matter on type 2 diabetes: evidence from a population-based epidemiological study. Environ Int. 2019; 130, 104882.CrossRefGoogle ScholarPubMed
Alderete, TL, Jones, RB, Chen, Z, et al. Exposure to traffic-related air pollution and the composition of the gut microbiota in overweight and obese adolescents. Environ Res. 2018; 161, 472478.CrossRefGoogle ScholarPubMed
Riva, A, Borgo, F, Lassandro, C, et al. Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ Microbiol. 2017; 19 (1), 95105.CrossRefGoogle ScholarPubMed
Konya, T, Koster, B, Maughan, H, et al. Associations between bacterial communities of house dust and infant gut. Environ Res. 2014; 131, 2530.CrossRefGoogle ScholarPubMed
Björkstén, B, Sepp, E, Julge, K, Voor, T, Mikelsaar, M. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol. 2001; 108 (4), 516520.CrossRefGoogle ScholarPubMed
Valkonen, M, Wouters, IM, Täubel, M, et al. Bacterial exposures and associations with atopy and asthma in children. PLoS One. 2015; 10 (6), e0131594.CrossRefGoogle ScholarPubMed
Zheng, H, Liang, H, Wang, Y, et al. Altered gut microbiota composition associated with eczema in infants. PLoS One. 2016; 11 (11), e0166026.CrossRefGoogle ScholarPubMed
Mutlu, EA, Comba, IY, Cho, T, et al. Inhalational exposure to particulate matter air pollution alters the composition of the gut microbiome. Environ Pollut. 2018; 240, 817830.CrossRefGoogle ScholarPubMed
Fujimura, KE, Demoor, T, Rauch, M, et al. House dust exposure mediates gut microbiome Lactobacillus enrichment and airway immune defense against allergens and virus infection. Proc Natl Acad Sci U S A. 2014; 111 (2), 805810.Google Scholar
Kish, L, Hotte, N, Kaplan, GG, et al. Environmental particulate matter induces murine intestinal inflammatory responses and alters the gut microbiome. PLoS One. 2013; 8 (4), e62220.CrossRefGoogle ScholarPubMed
Wang, W, Zhou, J, Chen, M, et al. Exposure to concentrated ambient PM (2.5) alters the composition of gut microbiota in a murine model. Particle Fibre Toxicol. 2018; 15 (1), 17.CrossRefGoogle Scholar
Postler, TS, Ghosh, S. Understanding the Holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab. 2017; 26 (1), 110130.CrossRefGoogle ScholarPubMed
Castaneda, AR, Vogel, CFA, Bein, KJ, Hughes, HK, Smiley-Jewell, S, Pinkerton, KE. Ambient particulate matter enhances the pulmonary allergic immune response to house dust mite in a BALB/c mouse model by augmenting Th2- and Th17-immune responses. Physiol Rep. 2018; 6 (18), e13827.CrossRefGoogle Scholar
Miller, RL, Peden, DB. Environmental effects on immune responses in patients with atopy and asthma. J Allergy Clin Immunol. 2014; 134 (5), 10011008.CrossRefGoogle ScholarPubMed
Pope, CA, Bhatnagar, A, McCracken, JP, Abplanalp, W, Conklin, DJ, O’Toole, T. Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation. Circ Res. 2016; 119 (11), 12041214.CrossRefGoogle ScholarPubMed
Li, W, Dorans, KS, Wilker, EH, et al. Short-term exposure to ambient air pollution and biomarkers of systemic inflammation: the Framingham Heart Study. Arterioscler Thromb Vasc Biol. 2017; 37 (9), 17931800.CrossRefGoogle ScholarPubMed
Salim, SY, Kaplan, GG, Madsen, KL. Air pollution effects on the gut microbiota: a link between exposure and inflammatory disease. Gut Microbes. 2014; 5 (2), 215219.CrossRefGoogle ScholarPubMed
Mutlu, EA, Engen, PA, Soberanes, S, et al. Particulate matter air pollution causes oxidant-mediated increase in gut permeability in mice. Particle Fibre Toxicol. 2011; 8, 19.CrossRefGoogle ScholarPubMed
Valles, Y, Francino, MP. Air pollution, early life microbiome, and development. Curr Environ Health Rep. 2018; 5 (4), 512521.CrossRefGoogle ScholarPubMed
Yan, L, Gong, C, Ying, L, et al. PM2.5 affects establishment of immune tolerance in newborn mice by reducing PD-L1 expression. J Biosci. 2019; 44 (2), 41.CrossRefGoogle ScholarPubMed
Smeekens, JM, Immormino, RM, Balogh, PA, Randell, SH, Kulis, MD, Moran, TP. Indoor dust acts as an adjuvant to promote sensitization to peanut through the airway. Clin Exp Allergy. 2019; 49 (11), 15001511.CrossRefGoogle ScholarPubMed
Ormstad, H. Suspended particulate matter in indoor air: adjuvants and allergen carriers. Toxicology. 2000; 152 (1), 5368.CrossRefGoogle ScholarPubMed
Kim, KE, Cho, D, Park, HJ. Air pollution and skin diseases: adverse effects of airborne particulate matter on various skin diseases. Life Sci. 2016; 152, 126134.CrossRefGoogle ScholarPubMed
Lee, C-W, Lin, Z-C, Hu, SC-S, et al. Urban particulate matter down-regulates filaggrin via COX2 expression/PGE2 production leading to skin barrier dysfunction. Sci Rep. 2016; 6, 27995.CrossRefGoogle ScholarPubMed
Stein, MM, Hrusch, CL, Gozdz, J, et al. Innate immunity and asthma risk in Amish and Hutterite farm children. N Engl J Med. 2016; 375 (5), 411421.CrossRefGoogle ScholarPubMed
Clifton, VL, Moss, TJ, Wooldridge, AL, et al. Development of an experimental model of maternal allergic asthma during pregnancy. J Physiol. 2016; 594 (5), 13111325.CrossRefGoogle ScholarPubMed
Wooldridge, AL, Clifton, VL, Moss, TJM, et al. Maternal allergic asthma during pregnancy alters fetal lung and immune development in sheep: potential mechanisms for programming asthma and allergy. J Physiol. 2019; 597 (16), 42514262.CrossRefGoogle ScholarPubMed
Mendola, P, Mannisto, TI, Leishear, K, Reddy, UM, Chen, Z, Laughon, SK. Neonatal health of infants born to mothers with asthma. J Allergy Clin Immunol. 2014; 133 (1), 8590. e81–e84.CrossRefGoogle ScholarPubMed
Henriquez, OA, Den Beste, K, Hoddeson, EK, Parkos, CA, Nusrat, A, Wise, SK. House dust mite allergen Der p 1 effects on sinonasal epithelial tight junctions. Int Forum Allergy Rhinol. 2013; 3 (8), 630635.CrossRefGoogle ScholarPubMed
Steelant, B, Farre, R, Wawrzyniak, P, et al. Impaired barrier function in patients with house dust mite-induced allergic rhinitis is accompanied by decreased occludin and zonula occludens-1 expression. J Allergy Clin Immunol. 2016; 137 (4), 10431053. e1045.CrossRefGoogle ScholarPubMed
Wan, H, Winton, HL, Soeller, C, et al. Quantitative structural and biochemical analyses of tight junction dynamics following exposure of epithelial cells to house dust mite allergen Der p 1. Clin Exp Allergy. 2000; 30 (5), 685698.CrossRefGoogle ScholarPubMed
Tulic, MK, Vivinus-Nebot, M, Rekima, A, et al. Presence of commensal house dust mite allergen in human gastrointestinal tract: a potential contributor to intestinal barrier dysfunction. Gut. 2016; 65 (5), 757766.CrossRefGoogle ScholarPubMed
Hinz, K. The effect of dust mite extract on esophageal tight junctions in eosinophilic esophagitis. Theses & Dissertations. 2018; 274.Google Scholar
Huang, Q, Zhang, J, Luo, L, et al. Metabolomics reveals disturbed metabolic pathways in human lung epithelial cells exposed to airborne fine particulate matter. Toxicol Res. 2015; 4.Google Scholar
Xu, Y, Wang, W, Zhou, J, et al. Metabolomics analysis of a mouse model for chronic exposure to ambient PM2.5. Environ Pollut. 2019; 247, 953963.CrossRefGoogle ScholarPubMed
Li, H, Cai, J, Chen, R, et al. Particulate matter exposure and stress hormone levels: a randomized, double-blind, crossover trial of air purification. Circulation. 2017; 136 (7), 618627.CrossRefGoogle ScholarPubMed
Kowal, K, Zebrowska, E, Chabowski, A. Altered sphingolipid metabolism is associated with asthma phenotype in house dust mite-allergic patients. Allergy Asthma Immunol Res. 2019; 11 (3), 330342.CrossRefGoogle ScholarPubMed
Kassotis, CD, Hoffman, K, Stapleton, HM. Characterization of adipogenic activity of house dust extracts and semi-volatile indoor contaminants in 3T3-L1 cells. Environ Sci Technol. 2017; 51 (15), 87358745.CrossRefGoogle ScholarPubMed
Turnbaugh, PJ, Ley, RE, Mahowald, MA, Magrini, V, Mardis, ER, Gordon, JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006; 444 (7122), 10271031.CrossRefGoogle ScholarPubMed
Le Chatelier, E, Nielsen, T, Qin, J, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013; 500 (7464), 541546.CrossRefGoogle ScholarPubMed
Nieuwdorp, M, Gilijamse, PW, Pai, N, Kaplan, LM. Role of the microbiome in energy regulation and metabolism. Gastroenterology. 2014; 146 (6), 15251533.CrossRefGoogle ScholarPubMed
Karagulian, F, Belis, C, Dora, C, et al. Contributions to cities’ ambient particulate matter (PM): a systematic review of local source contributions at global level. Atmos Environ. 2015; 120, 475–283.CrossRefGoogle Scholar
Tun, MH, Tun, HM, Mahoney, JJ, et al. Postnatal exposure to household disinfectants, infant gut microbiota and subsequent risk of overweight in children. Can Med Assoc J. 2018; 190 (37), E1097E1107.CrossRefGoogle ScholarPubMed