Air Pollution: Possible Interaction between the Immune and Nervous System?
Abstract
:1. Air Pollution: State of the Art
2. Air Pollution and the Immune System
2.1. Lung and Skin: Route of Exposure
2.2. Air Pollution Pathways in the Immune System
2.3. Air Pollution in Immune System Critical Windows: Pregnancy, Infancy and Childhood
3. Air Pollution and the Nervous System
3.1. Air Pollution Way to the Brain
3.2. Air Pollution in CNS Critical Windows: Neurodevelopment and Neurodegeneration
3.3. Air Pollution Pathways in the CNS
4. Possible Interaction between the Immune and Nervous System?
4.1. Neuro–Immune–Cutaneous–Endocrine (NICE) Network
4.2. Role of Microbiota: The Hygiene Hypothesis
5. Future Directions and Other Considerations
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Kinney, P.L. Climate change, air quality, and human health. Am. J. Prev. Med. 2008, 35, 459–467. [Google Scholar] [CrossRef] [PubMed]
- Brauer, M.; Amann, M.; Burnett, R.T.; Cohen, A.; Dentener, F.; Ezzati, M.; Henderson, S.B.; Krzyzanowski, M.; Martin, R.V.; Van Dingenen, R.; et al. Exposure assessment for estimation of the global burden of disease attributable to outdoor air pollution. Environ. Sci. Technol. 2012, 46, 652–660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, K.H.; Kabir, E.; Kabir, S. A review on the human health impact of airborne particulate matter. Environ. Int. 2015, 74, 136–143. [Google Scholar] [CrossRef]
- Vardoulakis, S.; Giagloglou, E.; Steinle, S.; Davis, A.; Sleeuwenhoek, A.; Galea, K.S.; Dixon, K.; Crawford, J.O. Indoor Exposure to Selected Air Pollutants in the Home Environment: A Systematic Review. Int. J. Environ. Res. Public Health 2020, 17, 8972. [Google Scholar] [CrossRef]
- Mannan, M.; Al-Ghamdi, S.G. Indoor Air Quality in Buildings: A Comprehensive Review on the Factors Influencing Air Pollution in Residential and Commercial Structure. Int. J. Environ. Res. Public Health 2021, 18, 3276. [Google Scholar] [CrossRef]
- WHO. Health Topic—Air Pollution. Available online: https://www.who.int/health-topics/air-pollution#tab=tab_1 (accessed on 1 September 2022).
- Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aslan, A.; Altinoz, B.; Ozsolak, B. The link between urbanization and air pollution in Turkey: Evidence from dynamic autoregressive distributed lag simulations. Environ. Sci. Pollut. Res. Int. 2021, 28, 52370–52380. [Google Scholar] [CrossRef]
- Benziger, C.P.; Roth, G.A.; Moran, A.E. The Global Burden of Disease Study and the Preventable Burden of NCD. Glob. Heart 2016, 11, 393–397. [Google Scholar] [CrossRef]
- Rutkowski, K.; Sowa, P.; Rutkowska-Talipska, J.; Sulkowski, S.; Rutkowski, R. Allergic diseases: The price of civilisational progress. Postepy Dermatol. Alergol. 2014, 31, 77–83. [Google Scholar] [CrossRef] [Green Version]
- WHO. Ambient (Outdoor) Air Quality and Health. 2016. Available online: http://www.who.int/en/news-room/fact-sheets/detail/ambient-(outdoor)-air-quality-and-health (accessed on 1 September 2022).
- Phalen, R.F.; Mendez, L.B.; Oldham, M.J. New developments in aerosol dosimetry. Inhal. Toxicol. 2010, 22 (Suppl. S2), 6–14. [Google Scholar] [CrossRef] [PubMed]
- Kim, B.J.; Lee, S.Y.; Kim, H.B.; Lee, E.; Hong, S.J. Environmental changes, microbiota, and allergic diseases. Allergy Asthma Immunol. Res. 2014, 6, 389–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schraufnagel, D.E.; Balmes, J.R.; Cowl, C.T.; De Matteis, S.; Jung, S.H.; Mortimer, K.; Perez-Padilla, R.; Rice, M.B.; Riojas-Rodriguez, H.; Sood, A.; et al. Air Pollution and Noncommunicable Diseases: A Review by the Forum of International Respiratory Societies’ Environmental Committee, Part 2: Air Pollution and Organ Systems. Chest 2019, 155, 417–426. [Google Scholar] [CrossRef]
- Li, X.; Chen, H.; Li, Y. The effect of air pollution on children’s migration with parents: Evidence from China. Environ. Sci. Pollut. Res. Int. 2020, 27, 12499–12513. [Google Scholar] [CrossRef] [PubMed]
- Brauer, M.; Brook, J.R.; Christidis, T.; Chu, Y.; Crouse, D.L.; Erickson, A.; Hystad, P.; Li, C.; Martin, R.V.; Meng, J.; et al. Mortality-Air Pollution Associations in Low-Exposure Environments (MAPLE): Phase 1. Res. Rep. Health Eff. Inst. 2019, 203, 1–87. [Google Scholar]
- Bert, B.; Maciej, S.; Jie, C.; Zorana, J.A.; Richard, A.; Mariska, B.; Tom, B.; Marie-Christine, B.; Jørgen, B.; Iain, C.; et al. Mortality and Morbidity Effects of Long-Term Exposure to Low-Level PM2.5, BC, NO2, and O3: An Analysis of European Cohorts in the ELAPSE Project. Res. Rep Health Eff. Inst. 2021, 208, 1–127. [Google Scholar]
- Dominici, F.; Schwartz, J.; Di, Q.; Braun, D.; Choirat, C.; Zanobetti, A. Assessing Adverse Health Effects of Long-Term Exposure to Low Levels of Ambient Air Pollution: Phase 1. Res. Rep. Health Eff. Inst. 2019, 200, 1–51. [Google Scholar]
- WHO. Global Air Quality Guideline 2021. Available online: https://apps.who.int/iris/bitstream/handle/10665/345329/9789240034228-eng.pdf?sequence=1&isAllowed=y (accessed on 10 November 2022).
- Nasser, Z.; Salameh, P.; Nasser, W.; Abou Abbas, L.; Elias, E.; Leveque, A. Outdoor particulate matter (PM) and associated cardiovascular diseases in the Middle East. Int. J. Occup. Med. Environ. Health 2015, 28, 641–661. [Google Scholar] [CrossRef]
- Costa, L.G.; Cole, T.B.; Coburn, J.; Chang, Y.C.; Dao, K.; Roque, P. Neurotoxicants are in the air: Convergence of human, animal, and in vitro studies on the effects of air pollution on the brain. BioMed. Res. Int. 2014, 2014, 736385. [Google Scholar] [CrossRef] [Green Version]
- Costa, L.G.; Cole, T.B.; Dao, K.; Chang, Y.C.; Coburn, J.; Garrick, J.M. Effects of air pollution on the nervous system and its possible role in neurodevelopmental and neurodegenerative disorders. Pharm. Ther. 2020, 210, 107523. [Google Scholar] [CrossRef]
- Loomis, D.; Grosse, Y.; Lauby-Secretan, B.; El Ghissassi, F.; Bouvard, V.; Benbrahim-Tallaa, L.; Guha, N.; Baan, R.; Mattock, H.; Straif, K.; et al. The carcinogenicity of outdoor air pollution. Lancet Oncol. 2013, 14, 1262–1263. [Google Scholar] [CrossRef]
- Watson, D.; Fernandez, J.A.; Wittmer, D.; Gravgaerd Pedersen, O. Environmental Pressures from European Consumption and Production. A Study in Integrated Environmental and Economic Analysis; European Environment Agency Technical Report: Copenhagen, Denmark, 2013. [CrossRef]
- World Health Organization; Occupational and Environmental Health Team. WHO Air Quality Guidelines for Particulate Matter, Ozone, Nitrogen Dioxide and Sulfur Dioxide: Global Update 2005: Summary of Risk Assessment; World Health Organization: Geneva, Switzerland, 2006.
- Esposito, S.; Tenconi, R.; Lelii, M.; Preti, V.; Nazzari, E.; Consolo, S.; Patria, M.F. Possible molecular mechanisms linking air pollution and asthma in children. BMC Pulm. Med. 2014, 14, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Real, Á.D.; Santurtún, A.; Teresa Zarrabeitia, M. Epigenetic related changes on air quality. Environ. Res. 2021, 197, 111155. [Google Scholar] [CrossRef] [PubMed]
- European Environment Agency. Air Quality in Europe; EEA Report No 9/2020; European Environment Agency: Copenhagen, Denmark, 2020. [CrossRef]
- Li, F.; An, Z.; Li, H.; Gao, X.; Wang, G.; Wu, W. Involvement of Oxidative Stress and the Epidermal Growth Factor Receptor in Diesel Exhaust Particle-Induced Expression of Inflammatory Mediators in Human Mononuclear Cells. Mediat. Inflamm. 2019, 2019, 3437104. [Google Scholar] [CrossRef] [PubMed]
- Shukla, A.; Bunkar, N.; Kumar, R.; Bhargava, A.; Tiwari, R.; Chaudhury, K.; Goryacheva, I.Y.; Mishra, P.K. Air pollution associated epigenetic modifications: Transgenerational inheritance and underlying molecular mechanisms. Sci. Total Environ. 2019, 656, 760–777. [Google Scholar] [CrossRef]
- Guidelines for Human Exposure Assessment—EPA 2019 (EPA/100/B-1/001). Available online: https://www.epa.gov/risk/guidelines-human-exposure-assessment (accessed on 1 September 2022).
- Shahadin, M.S.; Ab Mutalib, N.S.; Latif, M.T.; Greene, C.M.; Hassan, T. Challenges and future direction of molecular research in air pollution-related lung cancers. Lung Cancer 2018, 118, 69–75. [Google Scholar] [CrossRef]
- Paulin, L.; Hansel, N. Particulate air pollution and impaired lung function. F1000Research 2016, 5, 201. [Google Scholar] [CrossRef]
- Leikauf, G.D.; Kim, S.H.; Jang, A.S. Mechanisms of ultrafine particle-induced respiratory health effects. Exp. Mol. Med. 2020, 52, 329–337. [Google Scholar] [CrossRef]
- Donaldson, K.; Seaton, A. A short history of the toxicology of inhaled particles. Part. Fibre Toxicol. 2012, 9, 13. [Google Scholar] [CrossRef] [Green Version]
- Nemmar, A.; Hoet, P.H.; Vanquickenborne, B.; Dinsdale, D.; Thomeer, M.; Hoylaerts, M.F.; Vanbilloen, H.; Mortelmans, L.; Nemery, B. Passage of inhaled particles into the blood circulation in humans. Circulation 2002, 105, 411–414. [Google Scholar] [CrossRef] [Green Version]
- Mills, N.L.; Amin, N.; Robinson, S.D.; Anand, A.; Davies, J.; Patel, D.; de la Fuente, J.M.; Cassee, F.R.; Boon, N.A.; Macnee, W.; et al. Do inhaled carbon nanoparticles translocate directly into the circulation in humans? Am. J. Respir. Crit Care Med. 2006, 173, 426–431. [Google Scholar] [CrossRef] [Green Version]
- IARC. IARC Monographs on the evaluation of carcinogenic risk to humans. In Diesel and Gasoline Engine Exhaust and Some Nitroarenes; IARC: Lyon, France, 2013; Volume 105, pp. 1–714. [Google Scholar]
- Nguyen, A.V.; Soulika, A.M. The Dynamics of the Skin’s Immune System. Int. J. Mol. Sci. 2019, 20, 1811. [Google Scholar] [CrossRef] [PubMed]
- Fogarty, A.W. What have studies of non-industrialized countries told us about the cause of allergic disease? Clin. Exp. Allergy 2015, 45, 87–93. [Google Scholar] [CrossRef] [PubMed]
- Dijkhoff, I.M.; Drasler, B.; Karakocak, B.B.; Petri-Fink, A.; Valacchi, G.; Eeman, M.; Rothen-Rutishauser, B. Impact of airborne particulate matter on skin: A systematic review from epidemiology to in vitro studies. Part. Fibre Toxicol. 2020, 17, 35. [Google Scholar] [CrossRef]
- Ahn, K. The role of air pollutants in atopic dermatitis. J. Allergy Clin. Immunol. 2014, 134, 993–999. [Google Scholar] [CrossRef] [PubMed]
- Kurutas, E.B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: Current state. Nutr. J. 2016, 15, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bartra, J.; Mullol, J.; del Cuvillo, A.; Dávila, I.; Ferrer, M.; Jáuregui, I.; Montoro, J.; Sastre, J.; Valero, A. Air pollution and allergens. J. Investig. Allergol. Clin. Immunol. 2007, 17 (Suppl. S2), 3–8. [Google Scholar] [PubMed]
- Muranaka, M.; Suzuki, S.; Koizumi, K.; Takafuji, S.; Miyamoto, T.; Ikemori, R.; Tokiwa, H. Adjuvant activity of diesel-exhaust particulates for the production of IgE antibody in mice. J. Allergy Clin. Immunol. 1986, 77, 616–623. [Google Scholar] [CrossRef]
- Yanagisawa, R.; Takano, H.; Inoue, K.I.; Ichinose, T.; Sadakane, K.; Yoshino, S.; Yamaki, K.; Yoshikawa, T.; Hayakawa, K. Components of diesel exhaust particles differentially affect Th1/Th2 response in a murine model of allergic airway inflammation. Clin. Exp. Allergy 2006, 36, 386–395. [Google Scholar] [CrossRef]
- Kobayashi, T. Exposure to diesel exhaust aggravates nasal allergic reaction in guinea pigs. Am. J. Res. Crit Care Med. 2000, 162, 352–356. [Google Scholar] [CrossRef]
- Devalia, J.L.; Bayram, H.; Abdelaziz, M.M.; Sapsford, R.J.; Davies, R.J. Differences between cytokine release from bronchial epithelial cells of asthmatic patients and non-asthmatic subjects: Effect of exposure to diesel exhaust particles. Int. Arch. Allergy Immunol. 2019, 118, 437–439. [Google Scholar] [CrossRef]
- Pierdominici, M.; Maselli, A.; Cecchetti, S.; Tinari, A.; Mastrofrancesco, A.; Alfè, M.; Gargiulo, V.; Beatrice, C.; Di Blasio, G.; Carpinelli, G.; et al. Diesel exhaust particle exposure in vitro impacts T lymphocyte phenotype and function. Part. Fibre Toxicol. 2014, 11, 74. [Google Scholar] [CrossRef] [PubMed]
- Müller, L.; Chehrazi, C.V.; Henderson, M.W.; Noah, T.L.; Jaspers, I. Diesel exhaust particles modify natural killer cell function and cytokine release. Part. Fibre Toxicol. 2013, 10, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marano, F.; Boland, S.; Bonvallot, V.; Baulig, A.; Baeza-Squiban, A. Human airway epithelial cells in culture for studying the molecular mechanisms of the inflammatory response triggered by diesel exhaust particles. Cell Biol. Toxicol. 2002, 18, 315–320. [Google Scholar] [CrossRef] [PubMed]
- Amara, N.; Bachoual, R.; Desmard, M.; Golda, S.; Guichard, C.; Lanone, S.; Aubier, M.; Ogier-Denis, E.; Boczkowski, J. Diesel exhaust particles induce matrix metalloprotease-1 in human lung epithelial cells via a NADP(H) oxidase/NOX4 redox-dependent mechanism. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2007, 293, L170–L181. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Alam, J.; Venkatesan, M.I.; Eiguren-Fernandez, A.; Schmitz, D.; Di Stefano, E.; Slaughter, N.; Killeen, E.; Wang, X.; Huang, A.; et al. Nrf2 is a key transcription factor that regulates antioxidant defense in macrophages and epithelial cells: Protecting against the proinflammatory and oxidizing effects of diesel exhaust chemicals. J. Immunol. 2004, 173, 3467–3481. [Google Scholar] [CrossRef] [Green Version]
- Pourazar, J.; Mudway, I.S.; Samet, J.M.; Helleday, R.; Blomberg, A.; Wilson, S.J.; Frew, A.J.; Kelly, F.J.; Sandström, T. Diesel exhaust activates redox-sensitive transcription factors and kinases in human airways. Am. J. Physiol.-Lung Cell Mol. Physiol. 2005, 289, L724–L730. [Google Scholar] [CrossRef]
- Tal, T.L.; Simmons, S.O.; Silbajoris, R.; Dailey, L.; Cho, S.H.; Ramabhadran, R.; Linak, W.; Reed, W.; Bromberg, P.A.; Samet, J.M. Differential transcriptional regulation of IL-8 expression by human airway epithelial cells exposed to diesel exhaust particles. Toxicol. Appl. Pharmacol. 2010, 243, 46–54. [Google Scholar] [CrossRef]
- Jantzen, K.; Roursgaard, M.; Desler, C.; Loft, S.; Rasmussen, L.J.; Møller, P. Oxidative damage to DNA by diesel exhaust particle exposure in co-cultures of human lung epithelial cells and macrophages. Mutagenesis 2012, 27, 693–701. [Google Scholar] [CrossRef] [Green Version]
- de Oliveira, A.A.F.; de Oliveira, T.F.; Dias, M.F.; Medeiros, M.H.G.; Di Mascio, P.; Veras, M.; Lemos, M.; Marcourakis, T.; Saldiva, P.H.N.; Loureiro, A.P.M. Genotoxic and epigenotoxic effects in mice exposed to concentrated ambient fine particulate matter (PM2.5) from São Paulo city, Brazil. Part. Fibre Toxicol. 2018, 15, 40. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.W.; Vo, T.T.T.; Wu, C.Z.; Chi, M.C.; Lin, C.M.; Fang, M.L.; Lee, I.T. The Inducible Role of Ambient Particulate Matter in Cancer Progression via Oxidative Stress-Mediated Reactive Oxygen Species Pathways: A Recent Perception. Cancers 2020, 12, 2505. [Google Scholar] [CrossRef]
- Wang, J.; Huang, J.; Wang, L.; Chen, C.; Yang, D.; Jin, M.; Bai, C.; Song, Y. Urban particulate matter triggers lung inflammation via the ROS-MAPK-NF-κB signaling pathway. J. Thorac. Dis. 2017, 9, 4398–4412. [Google Scholar] [CrossRef] [PubMed]
- Al Hanai, A.H.; Antkiewicz, D.S.; Hemming, J.D.C.; Shafer, M.M.; Lai, A.M.; Arhami, M.; Hosseini, V.; Schauer, J.J. Seasonal variations in the oxidative stress and inflammatory potential of PM2.5 in Tehran using an alveolar macrophage model; The role of chemical composition and sources. Environ. Int. 2019, 123, 417–427. [Google Scholar] [CrossRef] [PubMed]
- Ishii, H.; Hayashi, S.; Hogg, J.C.; Fujii, T.; Goto, Y.; Sakamoto, N.; Mukae, H.; Vincent, R.; Van Eeden, S.F. Alveolar macrophage-epithelial cell interaction following exposure to atmospheric particles induces the release of mediators involved in monocyte mobilization and recruitment. Respir. Res. 2005, 6, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de FC Lichtenfels, A.J.; van der Plaat, D.A.; de Jong, K.; van Diemen, C.C.; Postma, D.S.; Nedeljkovic, I.; Van Duijn, C.M.; Amin, N.; la Bastide-van Gemert, S.; de Vries, M.; et al. Long-term Air Pollution Exposure, Genome-wide DNA Methylation and Lung Function in the LifeLines Cohort Study. Environ. Health Perspect. 2018, 126, 027004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clifford, R.L.; Jones, M.J.; MacIsaac, J.L.; McEwen, L.M.; Goodman, S.J.; Mostafavi, S.; Kobor, M.S.; Carlsten, C. Inhalation of diesel exhaust and allergen alters human bronchial epithelium DNA methylation. J. Allergy Clin. Immunol. 2017, 139, 112–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pai, V.V.; Shukla, P.; Kikkeri, N.N. Antioxidants in dermatology. Indian Derm. Online J. 2014, 5, 210–214. [Google Scholar] [CrossRef]
- Chaiprasongsuk, A.; Lohakul, J.; Soontrapa, K.; Sampattavanich, S.; Akarasereenont, P.; Panich, U. Activation of Nrf2 Reduces UVA-Mediated MMP-1 Upregulation via MAPK/AP-1 Signaling Cascades: The Photoprotective Effects of Sulforaphane and Hispidulin. J. Pharmacol. Exp. Ther. 2017, 360, 388–398. [Google Scholar] [CrossRef] [Green Version]
- D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248. [Google Scholar] [CrossRef] [Green Version]
- Prasad, R.; Katiyar, S.K. Crosstalk among UV-Induced Inflammatory Mediators, DNA Damage and Epigenetic Regulators Facilitates Suppression of the Immune System. Photochem. Photobiol. 2017, 93, 930–936. [Google Scholar] [CrossRef] [Green Version]
- Ryu, Y.S.; Kang, K.A.; Piao, M.J.; Ahn, M.J.; Yi, J.M.; Bossis, G.; Hyun, Y.M.; Park, C.O.; Hyun, J.W. Particulate matter-induced senescence of skin keratinocytes involves oxidative stress-dependent epigenetic modifications. Exp. Mol. Med. 2019, 51, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Tiotiu, A.I.; Novakova, P.; Nedeva, D.; Chong-Neto, H.J.; Novakova, S.; Steiropoulos, P.; Kowal, K. Impact of Air Pollution on Asthma Outcomes. Int. J. Environ. Res. Public Health 2020, 17, 6212. [Google Scholar] [CrossRef]
- To, T.; Zhu, J.; Stieb, D.; Gray, N.; Fong, I.; Pinault, L.; Jerrett, M.; Robichaud, A.; Ménard, R.; van Donkelaar, A.; et al. Early life exposure to air pollution and incidence of childhood asthma, allergic rhinitis and eczema. Eur. Respir. J. 2020, 55, 1900913. [Google Scholar] [CrossRef]
- Bieber, T. Atopic dermatitis. N. Engl. J. Med. 2008, 358, 1483–1494. [Google Scholar] [CrossRef]
- Centers for Disease Control and Prevention (US); National Center for Chronic Disease Prevention and Health Promotion (US); Office on Smoking and Health (US). How Tobacco Smoke Causes Disease: The Biology and Behavioral Basis for Smoking-Attributable Disease: A Report of the Surgeon General; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2010.
- Morita, A. Tobacco smoke causes premature skin aging. J. Derm. Sci. 2007, 48, 169–175. [Google Scholar] [CrossRef] [PubMed]
- Lipa, K.; Zając, N.; Owczarek, W.; Ciechanowicz, P.; Szymaǹska, E.; Walecka, I. Does smoking affect your skin? Postepy Dermatol. Alergol. 2021, 38, 371–376. [Google Scholar] [CrossRef] [PubMed]
- Kantor, R.; Kim, A.; Thyssen, J.P.; Silverberg, J.I. Association of atopic dermatitis with smoking: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2016, 75, 1119–1125.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, B.L.; Norhaizan, M.E.; Liew, W.P.; Sulaiman Rahman, H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef] [Green Version]
- Valacchi, G.; Sticozzi, C.; Belmonte, G.; Cervellati, F.; Demaude, J.; Chen, N.; Krol, Y.; Oresajo, C. Vitamin C Compound Mixtures Prevent Ozone-Induced Oxidative Damage in Human Keratinocytes as Initial Assessment of Pollution Protection. PLoS ONE 2015, 10, e0131097. [Google Scholar] [CrossRef]
- Lodovici, M.; Bigagli, E. Oxidative stress and air pollution exposure. J. Toxicol. 2011, 2011, 487074. [Google Scholar] [CrossRef]
- Yang, I.V.; Schwartz, D.A. Epigenetic mechanisms and the development of asthma. J. Allergy Clin. Immunol. 2012, 130, 1243–1255. [Google Scholar] [CrossRef] [Green Version]
- Smith, K.R.; Bruce, N.; Balakrishnan, K.; Adair-Rohani, H.; Balmes, J.; Chafe, Z.; Dherani, M.; Hosgood, H.D.; Mehta, S.; Pope, D. Millions dead: How do we know and what does it mean? Methods used in the comparative risk assessment of household air pollution. Annu. Rev. Public Health 2014, 35, 185–206. [Google Scholar] [CrossRef] [PubMed]
- Lacasaña, M.; Esplugues, A.; Ballester, F. Exposure to ambient air pollution and prenatal and early childhood health effects. Eur J. Epidemiol. 2005, 20, 183–199. [Google Scholar] [CrossRef] [PubMed]
- Cao, J.; Xu, X.; Hylkema, M.N.; Zeng, E.Y.; Sly, P.D.; Suk, W.A.; Bergman, Å.; Huo, X. Early-life Exposure to Widespread Environmental Toxicants and Health Risk: A Focus on the Immune and Respiratory Systems. Ann. Glob. Health 2016, 82, 119–131. [Google Scholar] [CrossRef] [Green Version]
- Gascon, M.; Sunyer, J. Air pollution and respiratory health in childhood. Arch. Bronconeu. Mol. 2015, 51, 371–372. [Google Scholar] [CrossRef] [Green Version]
- Johnson, N.M.; Hoffmann, A.R.; Behlen, J.C.; Lau, C.; Pendleton, D.; Harvey, N.; Shore, R.; Li, Y.; Chen, J.; Tian, Y.; et al. Air pollution and children’s health-a review of adverse effects associated with prenatal exposure from fine to ultrafine particulate matter. Environ. Health Prev. Med. 2021, 26, 72. [Google Scholar] [CrossRef]
- Bauer, R.N.; Diaz-Sanchez, D.; Jaspers, I. Effects of air pollutants on innate immunity: The role of Toll-like receptors and nucleotide-binding oligomerization domain-like receptors. J. Allergy Clin. Immunol. 2012, 129, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Becker, S.; Fenton, M.J.; Soukup, J.M. Involvement of microbial components and toll-like receptors 2 and 4 in cytokine responses to air pollution particles. Am. J. Respir. Cell Mol. Biol. 2002, 27, 611–618. [Google Scholar] [CrossRef]
- Kollmann, T.R.; Levy, O.; Montgomery, R.R.; Goriely, S. Innate immune function by Toll-like receptors: Distinct responses in newborns and the elderly. Immunity 2012, 37, 771–783. [Google Scholar] [CrossRef] [Green Version]
- Sram, R.J. Impact of Air Pollution on the Health of the Population in Parts of the Czech Republic. Int. J. Environ. Res. Public Health 2020, 17, 6454. [Google Scholar] [CrossRef]
- Peden, D.B. Prenatal exposure to particulate matter air pollution: A preventable risk for childhood asthma. J. Allergy Clin. Immunol. 2021, 148, 716–718. [Google Scholar] [CrossRef] [PubMed]
- Deng, Q.; Lu, C.; Li, Y.; Sundell, J.; Norbak, D. Exposure to outdoor air pollution during trimesters of pregnancy and childhood asthma, allergic rhinitis, and eczema. Environ. Res. 2016, 150, 119–127. [Google Scholar] [CrossRef] [PubMed]
- Lavigne, É.; Bélair, M.A.; Rodriguez Duque, D.; Do, M.T.; Stieb, D.M.; Hystad, P.; van Donkelaar, A.; Martin, R.V.; Crouse, D.L.; Crighton, E.; et al. Effect modification of perinatal exposure to air pollution and childhood asthma incidence. Eur. Respir. J. 2018, 51, 1701884. [Google Scholar] [CrossRef] [Green Version]
- Fu, L.; Yang, X.; Liu, X.; Yu, G.; Wang, Z. Prenatal O3 exposure increases the severity of OVA-induced asthma in offspring. Ecotoxicol. Environ. Saf. 2020, 188, 109867. [Google Scholar] [CrossRef] [PubMed]
- Kannan, S.; Misra, D.P.; Dvonch, J.T.; Krishnakumar, A. Exposures to airborne particulate matter and adverse perinatal outcomes: A biologically plausible mechanistic framework for exploring potential effect modification by nutrition. Environ. Health Perspect. 2006, 114, 1636–1642. [Google Scholar] [CrossRef] [PubMed]
- Dietert, R.R.; Etzel, R.A.; Chen, D.; Halonen, M.; Holladay, S.D.; Jarabek, A.M.; Landreth, K.; Peden, D.B.; Pinkerton, K.; Smialowicz, R.J.; et al. Workshop to identify critical windows of exposure for children’s health: Immune and respiratory systems work group summary. Environ. Health Perspect. 2000, 108 (Suppl. S3), 483–490. [Google Scholar] [CrossRef] [Green Version]
- Hew, K.M.; Walker, A.I.; Kohli, A.; Garcia, M.; Syed, A.; McDonald-Hyman, C.; Noth, E.M.; Mann, J.K.; Pratt, B.; Balmes, J.; et al. Childhood exposure to ambient polycyclic aromatic hydrocarbons is linked to epigenetic modifications and impaired systemic immunity in T cells. Clin. Exp. Allergy 2015, 45, 238–248. [Google Scholar] [CrossRef] [Green Version]
- 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, 67–73. [Google Scholar] [CrossRef] [Green Version]
- Bowatte, G.; Lodge, C.; Lowe, A.J.; Erbas, B.; Perret, J.; Abramson, M.J.; Matheson, M.; Dharmage, S.C. The influence of childhood traffic-related air pollution exposure on asthma, allergy and sensitization: A systematic review and a meta-analysis of birth cohort studies. Allergy 2015, 70, 245–256. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Wei, C.C.; Wan, L.; Lin, C.L.; Tsai, J.D. Association of exposure to hydrocarbon air pollution with the incidence of atopic dermatitis in children. Ital. J. Pediatr. 2021, 47, 202. [Google Scholar] [CrossRef]
- Rancière, F.; Bougas, N.; Viola, M.; Momas, I. Early Exposure to Traffic-Related Air Pollution, Respiratory Symptoms at 4 Years of Age, and Potential Effect Modification by Parental Allergy, Stressful Family Events, and Sex: A Prospective Follow-up Study of the PARIS Birth Cohort. Environ. Health Perspect. 2017, 125, 737–745. [Google Scholar] [CrossRef] [PubMed]
- Carroquino, M.J.; Posada, M.; Landrigan, P.J. Environmental Toxicology: Children at Risk. In Environmental Toxicology; Springer: New York, NY, USA, 2013; pp. 239–291. [Google Scholar] [CrossRef]
- Prunicki, M.; Stell, L.; Dinakarpandian, D.; de Planell-Saguer, M.; Lucas, R.W.; Katharine Hammond, S.; Balmes, J.R.; Zhou, X.; Paglino, T.; Sabatti, C.; et al. Exposure to NO2, CO, and PM2.5 is linked to regional DNA methylation differences in asthma. Clin. Epigenetics 2018, 10, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Min, K.D.; Yi, S.J.; Kim, H.C.; Leem, J.H.; Kwon, H.J.; Hong, S.; Kim, K.S.; Kim, S.Y. Association between exposure to traffic-related air pollution and pediatric allergic diseases based on modeled air pollution concentrations and traffic measures in Seoul, Korea: A comparative analysis. Environ. Health 2020, 19, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bowatte, G.; Lodge, C.J.; Knibbs, L.D.; Lowe, A.J.; Erbas, B.; Dennekamp, M.; Marks, G.B.; Giles, G.; Morrison, S.; Thompson, B.; et al. Traffic-related air pollution exposure is associated with allergic sensitization, asthma, and poor lung function in middle age. J. Allergy Clin. Immunol. 2017, 139, 122–129.e1. [Google Scholar] [CrossRef] [Green Version]
- Rouadi, P.W.; Idriss, S.A.; Naclerio, R.M.; Peden, D.B.; Ansotegui, I.J.; Canonica, G.W.; Gonzalez-Diaz, S.N.; Rosario Filho, N.A.; Ivancevich, J.C.; Hellings, P.W.; et al. Immunopathological features of air pollution and its impact on inflammatory airway diseases (IAD). World Allergy Organ. J. 2020, 13, 100467. [Google Scholar] [CrossRef] [PubMed]
- Mudway, I.S.; Kelly, F.J.; Holgate, S.T. Oxidative stress in air pollution research. Free Radic. Biol. Med. 2020, 151, 2–6. [Google Scholar] [CrossRef] [PubMed]
- Geiser, M.; Kreyling, W.G. Deposition and biokinetics of inhaled nanoparticles. Part. Fibre Toxicol. 2010, 7, 2. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Yokel, R.A.; Hennig, B.; Toborek, M. Manufactured aluminum oxide nanoparticles decrease expression of tight junction proteins in brain vasculature. J. Neuroimmune Pharm. 2008, 4, 286–295. [Google Scholar] [CrossRef] [Green Version]
- Oberdörster, G.; Sharp, Z.; Atudorei, V.; Elder, A.; Gelein, R.; Kreyling, W.; Cox, C. Translocation of inhaled ultrafine particles to the brain. Inhal. Toxicol. 2004, 16, 437–445. [Google Scholar] [CrossRef] [PubMed]
- Sunderman, F.W., Jr. Nasal toxicity, carcinogenicity, and olfactory uptake of metals. Ann. Clin. Lab. Sci. 2001, 31, 3–24. [Google Scholar] [PubMed]
- Elder, A.; Gelein, R.; Silva, V.; Feikert, T.; Opanashuk, L.; Carter, J.; Potter, R.; Maynard, A.; Ito, Y.; Finkelstein, J.; et al. Translocation of inhaled ultrafine manganese oxide particles to the central nervous system. Environ. Health Perspect. 2006, 114, 1172–1178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucchini, R.G.; Dorman, D.C.; Elder, A.; Veronesi, B. Neurological impacts from inhalation of pollutants and the nose-brain connection. Neurotoxicology 2012, 33, 838–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bromberg, P.A. Mechanisms of the acute effects of inhaled ozone in humans. Biochim. Biophys. Acta 2016, 1860, 2771–2781. [Google Scholar] [CrossRef] [PubMed]
- Thomson, E.M. Air Pollution, Stress, and Allostatic Load: Linking Systemic and Central Nervous System Impacts. J. Alzheimers Dis. 2019, 69, 597–614. [Google Scholar] [CrossRef] [Green Version]
- Costa, L.G.; Chang, Y.C.; Cole, T.B. Developmental Neurotoxicity of Traffic-Related Air Pollution: Focus on Autism. Curr. Environ. Health Rep. 2017, 4, 156–165. [Google Scholar] [CrossRef] [PubMed]
- Buoli, M.; Grassi, S.; Caldiroli, A.; Carnevali, G.S.; Mucci, F.; Iodice, S.; Cantone, L.; Pergoli, L.; Bollati, V. Is there a link between air pollution and mental disorders? Environ. Int. 2018, 118, 154–168. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.C.; Cole, T.B.; Costa, L.G. Prenatal and early-life diesel exhaust exposure causes autism-like behavioral changes in mice. Part. Fibre Toxicol. 2018, 15, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gładka, A.; Rymaszewska, J.; Zatoński, T. Impact of air pollution on depression and suicide. Int. J. Occup. Med. Environ. Health 2018, 31, 711–721. [Google Scholar] [CrossRef]
- Roberts, S.; Arseneault, L.; Barratt, B.; Beevers, S.; Danese, A.; Odgers, C.L.; Moffitt, T.E.; Reuben, A.; Kelly, F.J.; Fisher, H.L. Exploration of NO2 and PM2.5 air pollution and mental health problems using high-resolution data in London-based children from a UK longitudinal cohort study. Psychiatry Res. 2019, 272, 8–17. [Google Scholar] [CrossRef]
- Danysh, H.E.; Mitchell, L.E.; Zhang, K.; Scheurer, M.E.; Lupo, P.J. Traffic-related air pollution and the incidence of childhood central nervous system tumors: Texas, 2001–2009. Pediatr. Blood Cancer 2015, 62, 1572–1578. [Google Scholar] [CrossRef]
- Boda, E.; Rigamonti, A.E.; Bollati, V. Understanding the effects of air pollution on neurogenesis and gliogenesis in the growing and adult brain. Curr. Opin. Pharmacol. 2020, 50, 61–66. [Google Scholar] [CrossRef]
- Giovannini, N.; Schwartz, L.; Cipriani, S.; Parazzini, F.; Baini, I.; Signorelli, V.; Cetin, I. Particulate matter (PM10) exposure, birth and fetal-placental weight and umbilical arterial pH: Results from a prospective study. J. Matern. Fetal. Neonatal Med. 2018, 31, 651–655. [Google Scholar] [CrossRef]
- Calderón-Garcidueñas, L.; Mora-Tiscareño, A.; Ontiveros, E.; Gómez-Garza, G.; Barragán-Mejía, G.; Broadway, J.; Chapman, S.; Valencia-Salazar, G.; Jewells, V.; Maronpot, R.R.; et al. Air pollution, cognitive deficits and brain abnormalities: A pilot study with children and dogs. Brain Cogn. 2008, 68, 117–127. [Google Scholar] [CrossRef]
- Calderón-Garcidueñas, L.; Engle, R.; Mora-Tiscareño, A.; Styner, M.; Gómez-Garza, G.; Zhu, H.; Jewells, V.; Torres-Jardón, R.; Romero, L.; Monroy-Acosta, M.E.; et al. Exposure to severe urban air pollution influences cognitive outcomes, brain volume and systemic inflammation in clinically healthy children. Brain Cogn. 2011, 77, 345–355. [Google Scholar] [CrossRef]
- Calderón-Garcidueñas, L.; Kulesza, R.J.; Doty, R.L.; D’Angiulli, A.; Torres-Jardón, R. Megacities air pollution problems: Mexico City Metropolitan Area critical issues on the central nervous system pediatric impact. Environ. Res. 2015, 137, 157–169. [Google Scholar] [CrossRef]
- Newman, N.C.; Ryan, P.; Lemasters, G.; Levin, L.; Bernstein, D.; Khurana Hershey, G.K.; Lockey, J.E.; Villareal, M.; Reponen, T.; Grinshpun, S.; et al. Traffic-related air pollution exposure in the first year of life and behavioral scores at 7 years of age. Environ. Health Perspect. 2013, 121, 731–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perera, F.P.; Tang, D.; Wang, S.; Vishnevetsky, J.; Zhang, B.; Diaz, D.; Camann, D.; Rauh, V. Prenatal polycyclic aromatic hydrocarbon (PAH) exposure and child behavior at age 6–7 years. Environ. Health Perspect. 2012, 120, 921–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Suzuki, T.; Oshio, S.; Iwata, M.; Saburi, H.; Odagiri, T.; Udagawa, T.; Sugawara, I.; Umezawa, M.; Takeda, K. In utero exposure to a low concentration of diesel exhaust affects spontaneous locomotor activity and monoaminergic system in male mice. Part. Fibre Toxicol. 2010, 7, 7. [Google Scholar] [CrossRef] [Green Version]
- Yokota, S.; Moriya, N.; Iwata, M.; Umezawa, M.; Oshio, S.; Takeda, K. Exposure to diesel exhaust during fetal period affects behavior and neurotransmitters in male offspring mice. J. Toxicol. Sci. 2013, 38, 13–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Win-Shwe, T.T.; Yamamoto, S.; Fujitani, Y.; Hirano, S.; Fujimaki, H. Nanoparticle-rich diesel exhaust affects hippocampal-dependent spatial learning and NMDA receptor subunit expression in female mice. Nanotoxicology 2012, 6, 543–553. [Google Scholar] [CrossRef] [PubMed]
- Davis, D.A.; Bortolato, M.; Godar, S.C.; Sander, T.K.; Iwata, N.; Pakbin, P.; Shih, J.C.; Berhane, K.; McConnell, R.; Sioutas, C.; et al. Prenatal exposure to urban ai.ir nanoparticles in mice causes altered neuronal differentiation and depression-like responses. PLoS ONE 2013, 8, e64128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thirtamara Rajamani, K.; Doherty-Lyons, S.; Bolden, C.; Willis, D.; Hoffman, C.; Zelikoff, J.; Chen, L.C.; Gu, H. Prenatal and early-life exposure to high-level diesel exhaust particles leads to increased locomotor activity and repetitive behaviors in mice. Autism Res. 2013, 6, 248–257. [Google Scholar] [CrossRef] [PubMed]
- Babadjouni, R.M.; Hodis, D.M.; Radwanski, R.; Durazo, R.; Patel, A.; Liu, Q.; Mack, W.J. Clinical effects of air pollution on the central nervous system; a review. J. Clin. Neurosci. 2017, 43, 16–24. [Google Scholar] [CrossRef]
- Calderón-Garcidueñas, L.; Solt, A.C.; Henríquez-Roldán, C.; Torres-Jardón, R.; Nuse, B.; Herritt, L.; Villarreal-Calderón, R.; Osnaya, N.; Stone, I.; García, R.; et al. Long-term air pollution exposure is associated with neuroinflammation, an altered innate immune response, disruption of the blood-brain barrier, ultrafine particulate deposition, and accumulation of amyloid beta-42 and alpha-synuclein in children and young adults. Toxicol. Pathol. 2008, 36, 289–310. [Google Scholar] [CrossRef] [PubMed]
- Calderón-Garcidueñas, L.; Kavanaugh, M.; Block, M.; D’Angiulli, A.; Delgado-Chávez, R.; Torres-Jardón, R.; González-Maciel, A.; Reynoso-Robles, R.; Osnaya, N.; Villarreal-Calderon, R.; et al. Neuroinflammation, hyperphosphorylated tau, diffuse amyloid plaques, and down-regulation of the cellular prion protein in air pollution exposed children and young adults. J. Alzheimers Dis. 2012, 28, 93–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucchini, R.G.; Guazzetti, S.; Zoni, S.; Benedetti, C.; Fedrighi, C.; Peli, M.; Donna, F.; Bontempi, E.; Borgese, L.; Micheletti, S.; et al. Neurofunctional dopaminergic impairment in elderly after lifetime exposure to manganese. Neurotoxicology 2014, 45, 309–317. [Google Scholar] [CrossRef] [Green Version]
- Peters, R.; Ee, N.; Peters, J.; Booth, A.; Mudway, I.; Anstey, K.J. Air Pollution and Dementia: A Systematic Review. J. Alzheimers Dis. 2019, 70, S145–S163. [Google Scholar] [CrossRef] [Green Version]
- Tyler, C.R.; Noor, S.; Young, T.L.; Rivero, V.; Sanchez, B.; Lucas, S.; Caldwell, K.K.; Milligan, E.D.; Campen, M.J. Aging Exacerbates Neuroinflammatory Outcomes Induced by Acute Ozone Exposure. Toxicol. Sci. 2018, 163, 123–139. [Google Scholar] [CrossRef] [Green Version]
- Hahad, O.; Lelieveld, J.; Birklein, F.; Lieb, K.; Daiber, A.; Münzel, T. Ambient Air Pollution Increases the Risk of Cerebrovascular and Neuropsychiatric Disorders through Induction of Inflammation and Oxidative Stress. Int. J. Mol. Sci. 2020, 21, 4306. [Google Scholar] [CrossRef] [PubMed]
- Calderón-Garcidueñas, L.; Villarreal-Calderon, R.; Valencia-Salazar, G.; Henríquez-Roldán, C.; Gutiérrez-Castrellón, P.; Torres-Jardón, R.; Osnaya-Brizuela, N.; Romero, L.; Torres-Jardón, R.; Solt, A.; et al. Systemic inflammation, endothelial dysfunction, and activation in clinically healthy children exposed to air pollutants. Inhal. Toxicol. 2008, 20, 499–506. [Google Scholar] [CrossRef] [PubMed]
- Coburn, J.L.; Cole, T.B.; Dao, K.T.; Costa, L.G. Acute exposure to diesel exhaust impairs adult neurogenesis in mice: Prominence in males and protective effect of pioglitazone. Arch. Toxicol. 2018, 92, 1815–1829. [Google Scholar] [CrossRef]
- Chang, Y.C.; Daza, R.; Hevner, R.; Costa, L.G.; Cole, T.B. Prenatal and early life diesel exhaust exposure disrupts cortical lamina organization: Evidence for a reelin-related pathogenic pathway induced by interleukin-6. Brain Behav. Immun. 2019, 78, 105–115. [Google Scholar] [CrossRef] [PubMed]
- Gerlofs-Nijland, M.E.; van Berlo, D.; Cassee, F.R.; Schins, R.P.; Wang, K.; Campbell, A. Effect of prolonged exposure to diesel engine exhaust on proinflammatory markers in different regions of the rat brain. Part. Fibre Toxicol. 2010, 7, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van Berlo, D.; Albrecht, C.; Knaapen, A.M.; Cassee, F.R.; Gerlofs-Nijland, M.E.; Kooter, I.M.; Palomero-Gallagher, N.; Bidmon, H.J.; van Schooten, F.J.; Krutmann, J.; et al. Comparative evaluation of the effects of short-term inhalation exposure to diesel engine exhaust on rat lung and brain. Arch. Toxicol. 2010, 84, 553–562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cole, T.B.; Coburn, J.; Dao, K.; Roqué, P.; Chang, Y.C.; Kalia, V.; Guilarte, T.R.; Dziedzic, J.; Costa, L.G. Sex and genetic differences in the effects of acute diesel exhaust exposure on inflammation and oxidative stress in mouse brain. Toxicology 2016, 374, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ehsanifar, M.; Montazeri, Z.; Taheri, M.A.; Rafati, M.; Behjati, M.; Karimian, M. Hippocampal inflammation and oxidative stress following exposure to diesel exhaust nanoparticles in male and female mice. Neurochem. Int. 2021, 145, 104989. [Google Scholar] [CrossRef] [PubMed]
- Block, M.L.; Wu, X.; Pei, Z.; Li, G.; Wang, T.; Qin, L.; Wilson, B.; Yang, J.; Hong, J.S.; Veronesi, B. Nanometer size diesel exhaust particles are selectively toxic to dopaminergic neurons: The role of microglia, phagocytosis, and NADPH oxidase. FASEB J. 2004, 18, 1618–1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sama, P.; Long, T.C.; Hester, S.; Tajuba, J.; Parker, J.; Chen, L.C.; Veronesi, B. The cellular and genomic response of an immortalized microglia cell line (BV2) to concentrated ambient particulate matter. Inhal. Toxicol. 2007, 19, 1079–1087. [Google Scholar] [CrossRef] [PubMed]
- Woodward, N.C.; Levine, M.C.; Haghani, A.; Shirmohammadi, F.; Saffari, A.; Sioutas, C.; Morgan, T.E.; Finch, C.E. Toll-like receptor 4 in glial inflammatory responses to air pollution in vitro and in vivo. J. Neuroinflammation 2017, 14, 84. [Google Scholar] [CrossRef] [Green Version]
- Calderón-Garcidueñas, L.; Reed, W.; Maronpot, R.R.; Henríquez-Roldán, C.; Delgado-Chavez, R.; Calderón-Garcidueñas, A.; Dragustinovis, I.; Franco-Lira, M.; Aragón-Flores, M.; Solt, A.C.; et al. Brain inflammation and Alzheimer’s-like pathology in individuals exposed to severe air pollution. Toxicol. Pathol. 2004, 32, 650–658. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Budia, M.; Konttinen, H.; Saveleva, L.; Korhonen, P.; Jalava, P.I.; Kanninen, K.M.; Malm, T. Glial smog: Interplay between air pollution and astrocyte-microglia interactions. Neurochem. Int. 2020, 136, 104715. [Google Scholar] [CrossRef]
- Calderón-Garcidueñas, L.; Azzarelli, B.; Acuna, H.; Garcia, R.; Gambling, T.M.; Osnaya, N.; Monroy, S.; DEL Tizapantzi, M.R.; Carson, J.L.; Villarreal-Calderon, A.; et al. Air pollution and brain damage. Toxicol. Pathol. 2002, 30, 373–389. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.C.; Wang, X.; Wellenius, G.A.; Serre, M.L.; Driscoll, I.; Casanova, R.; McArdle, J.J.; Manson, J.E.; Chui, H.C.; Espeland, M.A. Ambient air pollution and neurotoxicity on brain structure: Evidence from women’s health initiative memory study. Ann. Neurol. 2015, 78, 466–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterson, B.S.; Rauh, V.A.; Bansal, R.; Hao, X.; Toth, Z.; Nati, G.; Walsh, K.; Miller, R.L.; Arias, F.; Semanek, D.; et al. Effects of prenatal exposure to air pollutants (polycyclic aromatic hydrocarbons) on the development of brain white matter, cognition, and behavior in later childhood. JAMA Psychiatry 2015, 72, 625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, B.; Guo, L.; Ku, T.; Chen, M.; Li, G.; Sang, N. PM2.5 exposure stimulates COX-2-mediated excitatory synaptic transmission via ROS-NF-κB pathway. Chemosphere 2018, 190, 124–134. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Kim, W.H.; Kim, Y.Y.; Park, H.Y. Air Pollution and Central Nervous System Disease: A Review of the Impact of Fine Particulate Matter on Neurological Disorders. Front. Public Health 2020, 8, 575330. [Google Scholar] [CrossRef] [PubMed]
- Puri, P.; Nandar, S.K.; Kathuria, S.; Ramesh, V. Effects of air pollution on the skin: A review. Indian J. Derm. Venereol. Leprol. 2017, 83, 415–423. [Google Scholar] [CrossRef]
- Arias-Pérez, R.D.; Taborda, N.A.; Gómez, D.M.; Narvaez, J.F.; Porras, J.; Hernandez, J.C. Inflammatory effects of particulate matter air pollution. Environ. Sci. Pollut. Res. Int. 2020, 27, 42390–42404. [Google Scholar] [CrossRef]
- Hung, S.C.; Cheng, H.Y.; Yang, C.C.; Lin, C.I.; Ho, C.K.; Lee, W.H.; Cheng, F.J.; Li, C.J.; Chuang, H.Y. The Association of White Blood Cells and Air Pollutants-A Population-Based Study. Int. J. Environ. Res. Public Health 2021, 18, 2370. [Google Scholar] [CrossRef]
- Valavanidis, A.; Vlachogianni, T.; Fiotakis, K.; Loridas, S. Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms. Int. J. Environ. Res. Public Health 2013, 10, 3886–3907. [Google Scholar] [CrossRef]
- Liu, A.H. Endotoxin exposure in allergy and asthma: Reconciling a paradox. J. Allergy Clin. Immunol. 2002, 109, 379–392. [Google Scholar] [CrossRef]
- Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
- Vidal Yucha, S.E.; Tamamoto, K.A.; Kaplan, D.L. The importance of the neuro-immuno-cutaneous system on human skin equivalent design. Cell Prolif. 2019, 52, e12677. [Google Scholar] [CrossRef] [Green Version]
- O’Sullivan, R.L.; Lipper, G.; Lerner, E.A. The neuro-immuno-cutaneous-endocrine network: Relationship of mind and skin. Arch. Derm. 1998, 134, 1431–1435. [Google Scholar] [CrossRef]
- Brazzini, B.; Ghersetich, I.; Hercogova, J.; Lotti, T. The neuro-immuno-cutaneous-endocrine network: Relationship between mind and skin. Dermatol. Ther. 2003, 16, 123–131. [Google Scholar] [CrossRef] [PubMed]
- Dantzer, R. Neuroimmune Interactions: From the Brain to the Immune System and Vice Versa. Physiol. Rev. 2018, 98, 477–504. [Google Scholar] [CrossRef] [PubMed]
- Misery L Atopic dermatitis and the nervous system. Clin. Rev. Allergy Immunol. 2011, 41, 259–266. [CrossRef]
- Tisoncik, J.R.; Korth, M.J.; Simmons, C.P.; Farrar, J.; Martin, T.R.; Katze, M.G. Into the eye of the cytokine storm. Microbiol. Mol. Biol. Rev. 2012, 76, 16–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Araviiskaia, E.; Berardesca, E.; Bieber, T.; Gontijo, G.; Sanchez Viera, M.; Marrot, L.; Chuberre, B.; Dreno, B. The impact of airborne pollution on skin. J. Eur. Acad. Derm. Venereol. 2019, 33, 1496–1505. [Google Scholar] [CrossRef]
- Kelley, K.W.; Réaux-Le Goazigo, A. Dialing in the dialogue between inflammation and the brain. Brain Behav. Immun. 2020, 88, 252–255. [Google Scholar] [CrossRef] [PubMed]
- Chavan, S.S.; Pavlov, V.A.; Tracey, K.J. Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity 2017, 46, 927–942. [Google Scholar] [CrossRef] [PubMed]
- Pavlov, V.A.; Chavan, S.S.; Tracey, K.J. Molecular and Functional Neuroscience in Immunity. Annu. Rev. Immunol. 2018, 36, 783–812. [Google Scholar] [CrossRef] [PubMed]
- Boche, D.; Perry, V.H.; Nicoll, J.A. Review: Activation patterns of microglia and their identification in the human brain. Neuropathol. Appl. Neurobiol. 2013, 39, 3–18. [Google Scholar] [CrossRef]
- D’Mello, C.; Swain, M.G. Immune-to-Brain Communication Pathways in Inflammation-Associated Sickness and Depression. Inflamm. Assoc. Depress. Evid. Mech. Implic. 2017, 31, 73–94. [Google Scholar] [CrossRef]
- von Mutius, E.; Vercelli, D. Farm living: Effects on childhood asthma and allergy. Nat. Rev. Immunol. 2010, 10, 861–868. [Google Scholar] [CrossRef] [PubMed]
- Panelli, S.; Epis, S.; Cococcioni, L.; Perini, M.; Paroni, M.; Bandi, C.; Drago, L.; Zuccotti, G.V. Inflammatory bowel diseases, the hygiene hypothesis and the other side of the microbiota: Parasites and fungi. Pharmacol Res. 2020, 159, 104962. [Google Scholar] [CrossRef] [PubMed]
- Da Costa Maranduba, C.M.; Ribeiro De Castro, S.B.; Torres de Souza, G.; Rossato, C.; Carlos da Guia, F.; Santana Valente, M.A.; Paes Rettore, J.V.; Pereira Maranduba, C.; Maurmann de Souza, C.; Resende do Carmo, A.M.; et al. Intestinal microbiota as modulators of the immune system and neuroimmune system: Impact on the host health and homeostasis. J. Immunol. Res. 2015, 2015, 931574. [Google Scholar] [CrossRef] [Green Version]
- Webley, W.C.; Aldridge, K.L. Infectious asthma triggers: Time to revise the hygiene hypothesis? Trends Microbiol. 2015, 23, 389–391. [Google Scholar] [CrossRef]
- Huang, Y.J.; Charlson, E.S.; Collman, R.G.; Colombini-Hatch, S.; Martinez, F.D.; Senior, R.M. The role of the lung microbiome in health and disease. A National Heart, Lung, and Blood Institute workshop report. Am. J. Respir. Crit. Care Med. 2013, 187, 1382–1387. [Google Scholar] [CrossRef] [Green Version]
- Kish, L.; Hotte, N.; Kaplan, G.G.; Vincent, R.; Tso, R.; Gänzle, M.; Rioux, K.P.; Thiesen, A.; Barkema, H.W.; Wine, E.; et al. Environmental particulate matter induces murine intestinal inflammatory responses and alters the gut microbiome. PLoS ONE 2013, 8, e62220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Allais, L.; Kerckhof, F.M.; Verschuere, S.; Bracke, K.R.; De Smet, R.; Laukens, D.; Van den Abbeele, P.; De Vos, M.; Boon, N.; Brusselle, G.G.; et al. Chronic cigarette smoke exposure induces microbial and inflammatory shifts and mucin changes in the murine gut. Environ. Microbiol. 2016, 18, 1352–1363. [Google Scholar] [CrossRef]
- Mutlu, E.A.; Comba, I.Y.; Cho, T.; Engen, P.A.; Yazıcı, C.; Soberanes, S.; Hamanaka, R.B.; Niğdelioğlu, R.; Meliton, A.Y.; Ghio, A.J.; et al. Inhalational exposure to particulate matter air pollution alters the composition of the gut microbiome. Environ. Pollut. 2018, 240, 817–830. [Google Scholar] [CrossRef] [PubMed]
- Yang, K.; Xu, M.; Cao, J.; Zhu, Q.; Rahman, M.; Holmén, B.A.; Fukagawa, N.K.; Zhu, J. Ultrafine particles altered gut microbial population and metabolic profiles in a sex-specific manner in an obese mouse model. Sci. Rep. 2021, 11, 6906. [Google Scholar] [CrossRef]
- Ridaura, V.; Belkaid, Y. Gut microbiota: The link to your second brain. Cell 2015, 161, 193–194. [Google Scholar] [CrossRef] [Green Version]
- Chu, C.; Murdock, M.H.; Jing, D.; Won, T.H.; Chung, H.; Kressel, A.M.; Tsaava, T.; Addorisio, M.E.; Putzel, G.G.; Zhou, L.; et al. The microbiota regulate neuronal function and fear extinction learning. Nature 2019, 574, 543–548. [Google Scholar] [CrossRef] [PubMed]
- El Aidy, S.; Dinan, T.G.; Cryan, J.F. Gut Microbiota: The Conductor in the Orchestra of Immune-Neuroendocrine Communication. Clin. Ther. 2015, 37, 954–967. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; Dinan, T.G. Gut microbiota: Microbiota and neuroimmune signalling-Metchnikoff to microglia. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 494–496. [Google Scholar] [CrossRef] [PubMed]
- Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [Green Version]
- Burokas, A.; Arboleya, S.; Moloney, R.D.; Peterson, V.L.; Murphy, K.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Targeting the Microbiota-Gut-Brain Axis: Prebiotics Have Anxiolytic and Antidepressant-like Effects and Reverse the Impact of Chronic Stress in Mice. Biol. Psychiatry 2017, 82, 472–487. [Google Scholar] [CrossRef] [PubMed]
- WHO. Health Consequences of Air Pollution on Populations. Available online: https://www.who.int/news/item/15-11-2019-what-are-health-consequences-of-air-pollution-on-populations (accessed on 15 November 2019).
- Pathway to a Healthy Planet for All. EU Action Plan: “Towards Zero Pollution for Air, Water and Soil”. Available online: https://eur-lex.europa.eu/resource.html?uri=cellar:a1c34a56-b314-11eb-8aca-01aa75ed71a1.0001.02/DOC_1&format=PDF (accessed on 9 November 2022).
- Sofia, D.; Gioiella, F.; Lotrecchiano, N.; Giuliano, A. Mitigation strategies for reducing air pollution. Environ. Sci. Pollut. Res. 2020, 27, 19226–19235. [Google Scholar] [CrossRef] [PubMed]
- Lindsay, G.; Macmillan, A.; Woodward, A. Moving urban trips from cars to bicycles: Impact on health and emissions. Aust. N. Z. J. Public Health 2011, 35, 54–60. [Google Scholar] [CrossRef]
- de Nazelle, A.; Bode, O.; Orjuela, J.P. Comparison of air pollution exposures in active vs. passive travel modes in European cities: A quantitative review. Envrion. Int. 2017, 99, 151–160. [Google Scholar] [CrossRef]
- Laumbach, R.; Meng, Q.; Kipen, H. What can individuals do to reduce personal health risks from air pollution? J. Thorac. Dis. 2015, 7, 96–107. [Google Scholar] [CrossRef] [PubMed]
- Carlsten, C.; Salvi, S.; Wong, G.W.K.; Chung, K.F. Personal strategies to minimise effects of air pollution on respiratory health: Advice for providers, patients and the public. Eur. Respir. J. 2020, 55, 1902056. [Google Scholar] [CrossRef] [PubMed]
- Giles, L.V.; Koehle, M.S. The health effects of exercising in air pollution. Sports Med. 2014, 44, 223–249. [Google Scholar] [CrossRef] [PubMed]
- Wright, R.J.; Brunst, K.J. Programming of respiratory health in childhood: Influence of outdoor air pollution. Curr. Opin. Pediatr. 2013, 25, 232–239. [Google Scholar] [CrossRef]
- Janghorbani, M.; Momeni, F.; Mansourian, M. Systematic review and metaanalysis of air pollution exposure and risk of diabetes. Eur. J. Epidemiol. 2014, 29, 231–242. [Google Scholar] [CrossRef]
- Backes, C.H.; Nelin, T.; Gorr, M.W.; Wold, L.E. Early life exposure to air pollution: How bad is it? Toxicol. Lett. 2013, 216, 47–53. [Google Scholar] [CrossRef] [Green Version]
- Laumbach, R.J. Outdoor air pollutants and patient health. Am. Fam. Physician 2010, 81, 175–180. [Google Scholar]
- Sun, Y.; Li, X.; Benmarhnia, T.; Chen, J.C.; Avila, C.; Sacks, D.A.; Chiu, V.; Slezak, J.; Molitor, J.; Getahun, D.; et al. Exposure to air pollutant mixture and gestational diabetes mellitus in Southern California: Results from electronic health record data of a large pregnancy cohort. Environ. Int. 2022, 158, 106888. [Google Scholar] [CrossRef]
- Li, R.; Hou, J.; Tu, R.; Liu, X.; Zuo, T.; Dong, X.; Pan, M.; Yin, S.; Hu, K.; Mao, Z.; et al. Associations of mixture of air pollutants with estimated 10-year atherosclerotic cardiovascular disease risk modified by socio-economic status: The Henan Rural Cohort Study. Sci. Total Environ. 2021, 793, 148542. [Google Scholar] [CrossRef]
- Nováková, Z.; Novák, J.; Kitanovski, Z.; Kukučka, P.; Smutná, M.; Wietzoreck, M.; Lammel, G.; Hilscherová, K. Toxic potentials of particulate and gaseous air pollutant mixtures and the role of PAHs and their derivatives. Environ. Int. 2020, 139, 105634. [Google Scholar] [CrossRef] [PubMed]
- Laine, J.E.; Bodinier, B.; Robinson, O.; Plusquin, M.; Scalbert, A.; Keski-Rahkonen, P.; Robinot, N.; Vermeulen, R.; Pizzi, C.; Asta, F.; et al. Prenatal Exposure to Multiple Air Pollutants, Mediating Molecular Mechanisms, and Shifts in Birthweight. Environ. Sci. Technol. 2020, 54, 14502–14513. [Google Scholar] [CrossRef] [PubMed]
- Occelli, F.; Lanier, C.; Cuny, D.; Deram, A.; Dumont, J.; Amouyel, P.; Montaye, M.; Dauchet, L.; Dallongeville, J.; Genin, M. Exposure to multiple air pollutants and the incidence of coronary heart disease: A fine-scale geographic analysis. Sci. Total Environ. 2020, 714, 136608. [Google Scholar] [CrossRef]
- Keil, A.P.; Buckley, J.P.; O’Brien, K.M.; Ferguson, K.K.; Zhao, S.; White, A.J. A Quantile-Based g-Computation Approach to Addressing the Effects of Exposure Mixtures. Environ. Health Perspect. 2020, 128, 47004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stafoggia, M.; Breitner, S.; Hampel, R.; Basagaña, X. Statistical Approaches to Address Multi-Pollutant Mixtures and Multiple Exposures: The State of the Science. Curr. Environ. Health Rep. 2017, 4, 481–490. [Google Scholar] [CrossRef] [PubMed]
Pollutant | Averaging Time | AQG Level |
---|---|---|
PM2.5 μg/m3 | Annual | 5 |
24 h a | 15 | |
PM10 μg/m3 | Annual | 15 |
24 h a | 45 | |
O3 μg/m3 | Peak season | 60 |
8 h b | 100 | |
NO2 μg/m3 | Annual | 10 |
24 h a | 25 | |
SO2 μg/m3 | 24 h a | 40 |
CO mg/m3 | 24 h a | 4 |
Air Pollutant | Effect | Type of Study | References |
---|---|---|---|
Active/passive cigarette smoke | Increased risk of AD. | Systematic review and meta-analysis | [75] |
CO | Modified methylation of FoxP3 promoter and IL-10 leading to asthma. | In vivo | [101] |
DEPs | Enhanced IgE mediated air-allergen sensitization. | Murine model | [45] |
Enhanced Th2 cytokines response. | Asthma murine model | [46] | |
Eosinophilic inflammation. | Guinea pigs sensitized to pollen | [47] | |
Increased production of Il-6 and IL-10 in patients with mild asthma. | Asthmatic patients | [48] | |
Decreased CD25 expression, IL-2 and IFN-γ in CD4+ and CD8+ T cells. | In vitro | [49] | |
Decreased release of IL-1β, IL-2, IL-4, IL-12p70, IFN-γ, and TNF-α by NK cells. | In vitro | [50] | |
Increase in ERK, p38, and NF-κB. | In vitro | [51] | |
Increase in MMP-1 and ERK1–2 phosphorylation. | In vitro | [52] | |
Activation of Nrf2. | In vitro | [53] | |
Increase in nuclear translocation NF-κB, AP-1, phosphorylated Jun kinase, and phosphorylated p38. | Nonatopic non-smokers (in vivo) | [54] | |
MAP kinase-mediated activation of NF-κB and AP-1. | In vitro | [55] | |
Increased oxidative damage of DNA. | In vitro | [56] | |
Changes in DNA methylation that increase the development and progression of allergic respiratory disease. | Randomized crossover-controlled exposure study | [63] | |
NO2 | Induction of blood DNA methylation and lung function changes. | Cohort study | [62] |
Modified methylation of FoxP3 promoter and IL-10 leading to asthma. | In vivo | [101] | |
O3 | Increased ROS formation and IL-8 gene expression in keratinocytes. | In vitro | [77] |
Imbalance of the Th1/Th2 differentiation in the offspring, increasing the severity of asthma. | In vivo | [92] | |
PM | Increase in ROS production, pro-inflammatory markers (IL-1β, IL-6, IL-8 e TNF-α) and phosphorylation of p38 MAPK. | In vitro | [59] |
Increase in ROS production, TNF-α and involvement of MAPK and NF-κB pathways. | In vitro | [60] | |
Increased GM-CSF levels, MIP-1β, MCP-1, IL-6, and ICAM-1. | In vitro | [61] | |
Decrease in DNA methyltransferase and increase in DNA demethylase leading to skin senescence. | In vitro | [68] | |
Increased levels of TLR2 and TLR4. | In vivo | [86] | |
Modified methylation of FoxP3 promoter and IL-10 leading to asthma. | In vivo | [101] | |
UVA | Induced ROS production in keratinocytes and fibroblasts, leading to the transcription of nuclear transcription factors AP-1 and NF-κB. | In vitro | [65] |
Air Pollutant | Effect | Type of Study | References |
---|---|---|---|
Air pollution | Up-regulation of COX-2, IL-1β, CD14 in olfactory bulb, frontal cortex, substantia nigra and vagus nerve, and increase in infiltrating monocytes or resident microglia. Disruption of BBB, higher oxidative stress, and inflammatory mediators. | In vivo, children and young adults | [133,139] |
DEPs | Alteration in dopamine turnover and monoamine metabolisms, leading to decrease in spontaneous locomotor activity. | In vivo, mice (in utero) | [127,128] |
Higher expression of NMDA receptor subunit GluN2A, CCL3, and BDNF indicating an impaired special learning and memory function. | In vivo, mice | [129] | |
Increase locomotor activity and repetitive behaviors in offspring, referable to autism. | In vivo, mice (during pregnancy and nursing) | [131] | |
Impaired cell proliferation only in males, reduction in adult neurogenesis, microglial activation, neuroinflammation and oxidative stress, indication of a sex-dependent impairment. | In vivo, mice | [140] | |
Increased IL-6 release in the placenta and neonatal brain with the consequent activation of the JAK2/STAT3 pathway in neonatal brain. | In vivo, mice | [141] | |
Increase in TNF-α and IL-1α in the striatum region. | In vivo, rats | [142] | |
Increased CYP1A1, iNOS, and oxygenase-1 and COX-2 in different brain regions. | In vivo, rats | [143] | |
Increase in IL-1α, IL-1β, TNF-α, IL-6 with some gender difference in olfactory bulb and hippocampus. Increase in oxygenase-1, nNOS and NMDA subunit GluN2A in male hippocampus. | In vivo, mice | [145] | |
Dose-dependent decrease in dopaminergic neurons. Increase superoxide and ROS production. | In vitro | [146] | |
Elemental carbon attributed to traffic | Association with hyperactivity T-score. | In vivo, children | [125] |
O3 | Structural damage at prefrontal cortex, elevated neuroinflammatory markers associated with cognitive deficits. | In vivo, children | [122,123,124] |
PAH | Increase in anxious/depressive symptoms. | In vivo, children | [125] |
Dose–response reduction in white matter. | Cross-sectional imaging study in school-age children | [153] | |
PM | Reduce weight at birth. | Prospective study in women during the first trimester of pregnancy | [121] |
Structural damage at prefrontal cortex, increase neuroinflammatory markers associated with cognitive deficits. | In vivo, children | [122,123,124] | |
Altered neuronal differentiation in the offspring and depression-like symptoms in adult males. | In vivo, mice | [130] | |
PM reached in Mn is associated with motor coordination and cognitive abilities deficits and increased levels of prolactin in serum. | In vivo, elderly human | [135] | |
Reduces intracellular levels of ATP and increase in TNF-α and IL-6. | In vitro | [147] | |
Activation of TLR.4 and NF-κB (in vitro). Increase in TLR.4, MyD88, TNF-α and TNFR2, and decrease in NF-κB in the hippocampus (in vivo). | In vitro/in vivo | [148] | |
Decrease in white matter. | Prospective study in women | [149] | |
Increase in COX-2 expression, ROS production and NF-κB phosphorylation. | In vitro | [154] | |
Ultrafine Mn oxide particles | Increased TNF-α, macrophage inflammatory protein-2, and neuronal cell adhesion molecules in olfactory bulb. | In vivo, rats | [110] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Serafini, M.M.; Maddalon, A.; Iulini, M.; Galbiati, V. Air Pollution: Possible Interaction between the Immune and Nervous System? Int. J. Environ. Res. Public Health 2022, 19, 16037. https://doi.org/10.3390/ijerph192316037
Serafini MM, Maddalon A, Iulini M, Galbiati V. Air Pollution: Possible Interaction between the Immune and Nervous System? International Journal of Environmental Research and Public Health. 2022; 19(23):16037. https://doi.org/10.3390/ijerph192316037
Chicago/Turabian StyleSerafini, Melania Maria, Ambra Maddalon, Martina Iulini, and Valentina Galbiati. 2022. "Air Pollution: Possible Interaction between the Immune and Nervous System?" International Journal of Environmental Research and Public Health 19, no. 23: 16037. https://doi.org/10.3390/ijerph192316037
APA StyleSerafini, M. M., Maddalon, A., Iulini, M., & Galbiati, V. (2022). Air Pollution: Possible Interaction between the Immune and Nervous System? International Journal of Environmental Research and Public Health, 19(23), 16037. https://doi.org/10.3390/ijerph192316037