1
|
Esposito S, Fainardi V, Titolo A, Lazzara A, Menzella M, Campana B, Argentiero A, Principi N. How air pollution fuels respiratory infections in children: current insights. Front Public Health 2025; 13:1567206. [PMID: 40365435 PMCID: PMC12070440 DOI: 10.3389/fpubh.2025.1567206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Background Air pollution is a significant global health concern, particularly for younger children who are especially susceptible to its adverse effects. Pollutants such as particulate matter (PM), nitrogen oxides (NO and NO₂), sulfur dioxide (SO2), ozone (O3), and carbon monoxide (CO) are associated with increased risks of upper respiratory tract infections (URTI) and lower respiratory tract infections (LRTI). While this association is well-documented, there are critical gaps in understanding the magnitude of these risks, the roles of specific pollutants, and the influence of age, sex, and exposure duration. Methods To confirm the relationship between air pollution and respiratory tract infections in children and to identify areas for further research on reducing pollution-related respiratory damage, a literature review was conducted using the MEDLINE/PubMed database for studies published from January 2000 to December 2024. Eligible studies included randomized controlled trials, cohort studies, and meta-analyses focusing on the relationship between air pollution and respiratory infections in children. Studies were grouped by pollutant type, exposure timing, and infection type. Results The literature analysis confirmed that pollution significantly increases the risk of URTI and LRTI in children, with infants and young children being the most vulnerable. Potential mechanisms for the development of respiratory tract pollution-related diseases include the promotion of oxidative stress, induction of inflammatory responses, deregulation of the immune system, and genetic alterations. Prenatal exposure significantly alters respiratory tract development, increasing the risk of LRTI and acute otitis media (AOM) early in life. Both short-term and long-term postnatal exposures can cause severe and recurrent LRTIs, reducing quality of life and leading to frequent hospitalizations and early death. However, the available data do not allow for precise definition of the magnitude of the risk, the individual and combined roles of specific pollutants, and the influence of factors such as age, sex, duration, and site of exposure on the development and severity of respiratory infections. Inconsistent findings on pollutant combinations and specific diseases like otitis media highlight the need for further research. Conclusion Air pollution is a major risk factor for respiratory infections in children, both prenatal and postnatal exposure can have significant negative impact. However, present knowledge is inadequate to develop effective preventive and therapeutic measures. Further studies are needed to minimize these cultural limits. In particular, it is necessary to delve deeper into how the various pollutants circulate, how they interact with each other, and how they are influenced by climate change and other environmental drivers. Results of these key researches can be translate into clinical and public health practice capable to help protect and improve children's environmental health.
Collapse
Affiliation(s)
- Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Valentina Fainardi
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Annachiara Titolo
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Angela Lazzara
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Marialaura Menzella
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Beatrice Campana
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | - Alberto Argentiero
- Department of Medicine and Surgery, Pediatric Clinic, University Hospital, University of Parma, Parma, Italy
| | | |
Collapse
|
2
|
Butler HM, Keller E, McCrorey M, Keceli G, Combs CK, Kayed R, Namakkal-S R, Paolocci N, Jacobs Wolf B, Wold LE, Del Monte F. Particulate matter and co-occurring genetic risk induce oxidative stress and cardiac and brain Alzheimer's pathology. Commun Biol 2025; 8:603. [PMID: 40221628 PMCID: PMC11993720 DOI: 10.1038/s42003-025-07701-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/07/2025] [Indexed: 04/14/2025] Open
Abstract
Amyloid-beta (Aβ) aggregates, an Alzheimer's disease (AD) pathological hallmark, extend beyond the brain to the heart of heart failure (HF) and AD patients. Being diseases of the elderly, increased prevalence is expected as the population ages. However, changes in the incidence and prevalence of dementia over the past decades, and the independent association of exposure to air particulate matter (PM) with poor cognitive function, adverse cardiovascular effects, and oxidative stress hint to the contribution of other factors beyond senescence. Therefore we evaluate whether, and by which mechanism(s), PM exposure affects heart and brain proteinopathy with/without genetic predisposition.AD-prone and control mice are exposed for three months to filtered air (FA) or concentrated ambient PM < 2.5μm in diameter (PM2.5), and evaluated for Aβ pathology, cognitive and cardiac function, and markers of oxidative stress. Aβ pathology become noticeable in AD hearts and worsens with PM2.5 in AD brains. Functionally, PM2.5 lead to anxiety and memory deficits and worsens diastolic function. Redox homeostasis is negatively impacted by genotype and PM2.5. This study identifies environmental pollution as a potential key contributor to early progression of heart and brain proteinopathy, delineating a crucial timepoint for early interventions to limit multiorgan damage in vulnerable patients.
Collapse
Affiliation(s)
- Helen M Butler
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Everette Keller
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Marice McCrorey
- College of Graduate Studies, Medical University of South Carolina, Charleston, SC, USA
| | - Gizem Keceli
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Colin K Combs
- School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Rakez Kayed
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rajasekaran Namakkal-S
- Department of Pathology/Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nazareno Paolocci
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Bethany Jacobs Wolf
- College of Medicine, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Loren E Wold
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Medicine and Surgery, University of Bologna Alma Mater, Bologna, Italy.
| |
Collapse
|
3
|
Burbank AJ. Climate Change and the Future of Allergies and Asthma. Curr Allergy Asthma Rep 2025; 25:20. [PMID: 40146339 DOI: 10.1007/s11882-025-01201-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE OF THIS REVIEW Climate change affects global temperature, meteorological variables, plant aerobiology, air pollution exposure and a host of other factors that individually have been implicated in the inception and/or exacerbation of allergic disease like asthma and allergic rhinitis. It is unknown how climate change will impact allergic disease prevalence and morbidity in the future. RECENT FINDINGS Pollen seasons are lengthening with variable effects on pollen peak concentrations and allergenicity. Air pollution exposure is linked with enhance susceptibility to allergic inflammation induced by pollen and with enhanced susceptibility to infection with a morbidity/mortality from respiratory viruses, including SARS-CoV-2. The available literature largely supports the association between climate change and three of the most salient factors for allergic respiratory disease prevalence and morbidity: changes in allergen exposure, pollution exposure, and viral respiratory infection. More research is needed to understand the complex interactions between these factors and individual-level variables that influence disease susceptibility.
Collapse
Affiliation(s)
- Allison J Burbank
- Division of Pediatric Allergy and Immunology, University of North Carolina, Mary Ellen Jones Bldg, 5008B 116 Manning Dr, CB#7231, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Burbank AJ, Penrice AJ, Rorie AC, Oh JW. Climate Change and Allergens: Current and Future Impacts. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025:S2213-2198(25)00212-0. [PMID: 40074172 DOI: 10.1016/j.jaip.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
Climate change will continue to impact allergic diseases in direct and indirect ways. Rising global temperatures are contributing to increased duration of pollen seasons, altered aeroallergen production and potency of allergens, and changes in the geographic distribution of allergenic plants that drive increased human exposure to aeroallergens and increased allergic disease morbidity. Climate change is inextricably linked with air pollution, the latter of which was shown to act as an adjuvant for allergic inflammatory processes promoting allergic sensitization. Pollutant exposure is also linked with higher prevalence of childhood asthma and exacerbation of existing asthma and allergic disease. Increased exposure, or co-exposure, to aeroallergens and air pollution as a result of climate change will result in higher rates of sensitization, and incident allergic disease remains uncertain. Vulnerable populations, including children, the elderly, and marginalized groups, are likely to be disproportionately affected. This review summarizes the current knowledge of the effects of climate change on aeroallergens, and by extension, allergic disease. Addressing these health challenges requires a comprehensive understanding of the interaction between climate change, allergens, pollution and public health, alongside proactive measures to mitigate these effects.
Collapse
Affiliation(s)
- Allison J Burbank
- Department of Pediatrics, Division of Allergy and Immunology, University of North Carolina, Chapel Hill, NC.
| | - Alexander J Penrice
- Department of Medicine, Division of Allergy and Immunology, University of Nebraska Medical Center, Omaha, Neb
| | - Andrew C Rorie
- Department of Medicine, Division of Allergy and Immunology, University of Nebraska Medical Center, Omaha, Neb
| | - Jae-Won Oh
- Department of Pediatrics, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
5
|
Balmes JR, Hicks A, Johnson MM, Nadeau KC. The Effect of Wildfires on Asthma and Allergies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025; 13:280-287. [PMID: 39672379 PMCID: PMC11807743 DOI: 10.1016/j.jaip.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/22/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Climate change is a major driver of the frequency and severity of wildfires caused by extended periods of drought and hotter, drier weather superimposed on the legacy of fire suppression in the Mountain West of the United States. In recent years, increased wildfire smoke has negated the improvements in air quality made by clean energy transitions. Wildfire smoke is a complex mixture of gases and solids, a chief constituent of which is fine particulate matter (PM2.5). Exposure to PM2.5 is associated with adverse respiratory outcomes, including exacerbations of asthma and chronic obstructive pulmonary disease. In the face of increasing wildfire smoke exposures, it is critical that adaptation and mitigation strategies be put in place to minimize health effects. Individual strategies include modifying behavior and creating clean air spaces in homes to avoid wildfire smoke exposure. Community strategies include regulations promoting fire-resistant buildings and landscaping; establishing wildfire monitoring and alert systems; providing safe clean spaces where individuals can minimize wildfire smoke exposure and find evacuation routes; and creating health care response teams. Mitigation to prevent wildfires includes forest management and establishing monitoring systems and protocols to control forest fires in the wildland urban interface before they increase in size and intensity. Research into understanding the mechanism by which wildfire smoke mediates adverse health effects can inform guidelines to mitigate its health effects further.
Collapse
Affiliation(s)
- John R Balmes
- Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, Calif; Department of Medicine, University of California, San Francisco, Calif
| | - Anne Hicks
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Mary M Johnson
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass.
| |
Collapse
|
6
|
Kim HM, Kim JH, Park BJ, Park HJ. Chitosan Nanoparticle-Encapsulated Cordyceps militaris Grown on Germinated Rhynchosia nulubilis Reduces Type II Alveolar Epithelial Cell Apoptosis in PM 2.5-Induced Lung Injury. Int J Mol Sci 2025; 26:1105. [PMID: 39940873 PMCID: PMC11817496 DOI: 10.3390/ijms26031105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Chitosan nanoparticles (CNPs) were synthesized in this study to enhance the limited bioactivity and stability of Cordyceps militaris grown on germinated Rhynchosia nulubilis (GRC) and effectively deliver it to target tissues. Under optimized conditions, stable encapsulation of GRC was achieved by setting the chitosan (CHI)-to-tripolyphosphate (TPP) ratio to 4:1 and adjusting the pH of TPP to 2, resulting in a zeta potential of +22.77 mV, which indicated excellent stability. As the concentration of GRC increased, the encapsulation efficiency decreased, whereas the loading efficiency increased. Fourier-transform infrared (FT-IR) spectroscopy revealed shifts in the amide I and II bands of CHI from 1659 and 1578 to 1639 cm⁻1, indicating hydrogen bonding and successful encapsulation of GRC encapsulated with CNPs (GCN). X-ray diffraction (XRD) examination revealed the transition of the nanoparticles from a crystalline to an amorphous state, further confirming successful encapsulation. In vivo experiments demonstrated that GCN treatment significantly reduced lung injury scores in fine particulate matter (PM2.5)-exposed mice (p < 0.05) and alleviated lung epithelial barrier damage by restoring the decreased expression of occludin protein (p < 0.05). In addition, GCN decreased the PM2.5-induced upregulation of MMP-9 and COL1A1 mRNA expression levels, preventing extracellular matrix (ECM) degradation and collagen accumulation (p < 0.05). GCN exhibited antioxidant effects by reducing the mRNA expression of nitric oxide synthase (iNOS) and enhancing both the protein and mRNA expression of superoxide dismutase (SOD-1) caused by PM2.5, thereby alleviating oxidative stress (p < 0.05). In A549 cells, GCN significantly reduced PM2.5-induced reactive oxygen species (ROS) production compared with GRC (p < 0.05), with enhanced intracellular uptake confirmed using fluorescence microscopy (p < 0.05). In conclusion, GCN effectively alleviated PM2.5-induced lung damage by attenuating oxidative stress, suppressing apoptosis, and preserving the lung epithelial barrier integrity. These results emphasize its potential as a therapeutic candidate for preventing and treating the lung diseases associated with PM2.5 exposure.
Collapse
Affiliation(s)
| | | | | | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea; (H.-M.K.); (J.-H.K.); (B.-J.P.)
| |
Collapse
|
7
|
Zeng F, Pang G, Hu L, Sun Y, Peng W, Chen Y, Xu D, Xia Q, Zhao L, Li Y, He M. Subway Fine Particles (PM 2.5 )-Induced Pro-Inflammatory Response Triggers Airway Epithelial Barrier Damage Through the TLRs/NF-κB-Dependent Pathway In Vitro. ENVIRONMENTAL TOXICOLOGY 2024; 39:5296-5308. [PMID: 39189708 DOI: 10.1002/tox.24403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 08/28/2024]
Abstract
Subways are widely used in major cities around the world, and subway fine particulate matter (PM2.5) is the main source of daily PM2.5 exposure for urban residents. Exposure to subway PM2.5 leads to acute inflammatory damage in humans, which has been confirmed in mouse in vivo studies. However, the concrete mechanism by which subway PM2.5 causes airway damage remains obscure. In this study, we found that subway PM2.5 triggered release of pro-inflammatory cytokines such as interleukin 17E, tumor necrosis factor α, transforming growth factor β, and thymic stromal lymphopoietin from human bronchial epithelial cells (BEAS-2B) in a dose-effect relationship. Subsequently, supernatant recovered from the subway PM2.5 group significantly increased expression of the aforementioned cytokines in BEAS-2B cells compared with the subway PM2.5 group. Additionally, tight junctions (TJs) of BEAS-2B cells including zonula occludens-1, E-cadherin, and occludin were decreased by subway PM2.5 in a dose-dependent manner. Moreover, supernatant recovered from the subway PM2.5 group markedly decreased the expression of these TJs compared with the control group. Furthermore, inhibitors of toll-like receptors (TLRs) and nuclear factor-kappa B (NF-κB), as well as chelate resins (e.g., chelex) and deferoxamine, remarkably ameliorated the observed changes of cytokines and TJs caused by subway PM2.5 in BEAS-2B cells. Therefore, these results suggest that subway PM2.5 induced a decline of TJs after an initial ascent of cytokine expression, and subway PM2.5 altered expression of both cytokines and TJs by activating TLRs/NF-κB-dependent pathway in BEAS-2B cells. The metal components of subway PM2.5 may contribute to the airway epithelial injury.
Collapse
Affiliation(s)
- Fanmei Zeng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Guanhua Pang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Liwen Hu
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Yuan Sun
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Wen Peng
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yuwei Chen
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Dan Xu
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Qing Xia
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Luwei Zhao
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Yifei Li
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Miao He
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
- Liaoning Key Laboratory of Environmental Health Damage Research and Assessment, Shenyang, China
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, Shenyang, China
| |
Collapse
|
8
|
Ardicli S, Ardicli O, Yazici D, Pat Y, Babayev H, Xiong P, Zeyneloglu C, Garcia-Sanchez A, Shi LL, Viscardi OG, Skolnick S, Ogulur I, Dhir R, Jutel M, Agache I, Janda J, Pali-Schöll I, Nadeau KC, Akdis M, Akdis CA. Epithelial barrier dysfunction and associated diseases in companion animals: Differences and similarities between humans and animals and research needs. Allergy 2024; 79:3238-3268. [PMID: 39417247 DOI: 10.1111/all.16343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Since the 1960s, more than 350,000 new chemicals have been introduced into the lives of humans and domestic animals. Many of them have become part of modern life and some are affecting nature as pollutants. Yet, our comprehension of their potential health risks for both humans and animals remains partial. The "epithelial barrier theory" suggests that genetic predisposition and exposure to diverse factors damaging the epithelial barriers contribute to the emergence of allergic and autoimmune conditions. Impaired epithelial barriers, microbial dysbiosis, and tissue inflammation have been observed in a high number of mucosal inflammatory, autoimmune and neuropsychiatric diseases, many of which showed increased prevalence in the last decades. Pets, especially cats and dogs, share living spaces with humans and are exposed to household cleaners, personal care products, air pollutants, and microplastics. The utilisation of cosmetic products and food additives for pets is on the rise, unfortunately, accompanied by less rigorous safety regulations than those governing human products. In this review, we explore the implications of disruptions in epithelial barriers on the well-being of companion animals, drawing comparisons with humans, and endeavour to elucidate the spectrum of diseases that afflict them. In addition, future research areas with the interconnectedness of human, animal, and environmental well-being are highlighted in line with the "One Health" concept.
Collapse
Affiliation(s)
- Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Türkiye
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Türkiye
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Peng Xiong
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical & Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | | | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SEED Inc. Co., Los Angeles, California, USA
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Raja Dhir
- SEED Inc. Co., Los Angeles, California, USA
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wrocław, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Jozef Janda
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine and Medical University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
9
|
Wang C, Niu Z, Zhang Y, Liu N, Ji X, Tian J, Guan L, Shi D, Zheng H, Gao Y, Zhao L, Zhang W, Zhang Z. Exosomal miR-129-2-3p promotes airway epithelial barrier disruption in PM 2.5-aggravated asthma. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 370:123053. [PMID: 39467462 DOI: 10.1016/j.jenvman.2024.123053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/20/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
Particulate matter 2.5 (PM2.5) exposure is intricately linked to asthma exacerbations. Damage to the airway epithelial barrier function serves as an initiating factor for asthma attacks and worsening symptoms. In recent years, numerous exosomal microRNAs (miRNAs) have emerged as potential biomarkers for diagnosing asthma. However, the mechanisms by which PM2.5-induced exosomes exacerbate asthma remain unclear. This study aims to investigate the role of exosomal miR-129-2-3p in regulating airway epithelial cell barrier function, its potential targets, and signaling pathways involved in PM2.5-induced aggravation of asthma. In this study, miR-129-2-3p is highly expressed in plasma exosomes from patients with asthma, mouse lung tissue and plasma exosomes, and exosomes produced by PM2.5-stimulated 16HBE cells. Moreover, the exposure level of PM2.5 is positively correlated with exosomal miR-129-2-3p in plasma in patients with asthma. As the concentration of PM2.5 increases, the synthesis of connexin (ZO-1, occludin, and E-cadherin) is gradually weakened, while the content of inflammatory factors (IL-6, IL-8, and TNF-α) is notably upregulated in PM2.5 exacerbated asthmatic mice. PM2.5-induced exosomes can decrease the level of connexin, enhance cell permeability and promote the secretion of inflammatory factors in 16HBE cells. TIAM1, a specific target gene for miR-129-2-3p, regulates the synthesis of connexin. Exosomal miR-129-2-3p exacerbates airway epithelial barrier dysfunction by targeted inhibition of the TIAM1/RAC1/PAK1 signaling pathway in PM2.5 aggravated asthma. In contrast, blocking miR-129-2-3p may be an alternative approach to therapeutic intervention in asthma.
Collapse
Affiliation(s)
- Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Zeyu Niu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Yan Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Xiaotong Ji
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Linlin Guan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Dongxing Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Huiqiu Zheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Yuhui Gao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Lifang Zhao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| | - Wenping Zhang
- MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China; Department of Toxicology, School of Public Health, Shanxi Medical University, China.
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, China; Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, China.
| |
Collapse
|
10
|
Somayajulu M, Wright R, Muhammed F, McClellan SA, Ibrahim A, Hazlett LD. PM 10 dysregulates epithelial barrier function in human corneal epithelial cells that is restored by antioxidant SKQ1. Toxicol Appl Pharmacol 2024; 492:117122. [PMID: 39393465 PMCID: PMC11563859 DOI: 10.1016/j.taap.2024.117122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
Exposure to airborne particulate <10 μm (PM10) adversely affects the ocular surface. This study tested PM10 on epithelial barrier integrity in immortalized human corneal epithelial cells (HCE-2) and mouse cornea, and whether antioxidant SKQ1 is restorative. HCE-2 were exposed to 100 μg/ml PM10 ± SKQ1 for 24 h. An Electric Cell-Substrate Impedance Sensing (ECIS) system monitored the impact of PM10. RT-PCR, western blotting and immunofluorescence measured levels of barrier and associated proteins, stanniocalcin 2 (STC2), and a kit measured total calcium. In vivo, female C57BL/6 mice were exposed to either control air or PM10 (±SKQ1) in a whole-body exposure chamber, and barrier associated proteins tested. Tight junction and mucins proteins in the cornea were tested. In HCE-2, PM0 vs control significantly reduced mRNA and protein levels of tight junction and adherence proteins, and mucins. ECIS data demonstrated that PM10 vs control cells exhibited a significant decrease in epithelial barrier strength at 4000 Hz indicated by reduced impedance and resistance. PM10 also upregulated STC2 protein and total calcium levels. In vivo, PM10 vs control reduced zonula occludens 1 and mucins. SKQ1 pre-treatment reversed PM10 effects both in vitro and in vivo. In conclusion, PM10 exposure reduced tight junction and mucin proteins, and compromised the seal between cells in the corneal epithelium leading to decreased epithelial barrier strength. This effect was reversed by SKQ1. Since the corneal epithelium forms the first line of defense against air pollutants, including PM10, preserving its integrity using antioxidants such as SKQ1 is crucial in reducing the occurrence of ocular surface disorders.
Collapse
Affiliation(s)
- Mallika Somayajulu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Robert Wright
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Farooq Muhammed
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Sharon A McClellan
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Ahmed Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA; Department of Pharmacology, Wayne State University, School of Medicine, Detroit, MI 48201, USA
| | - Linda D Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
11
|
Mazumder MHH, Hussain S. Air-Pollution-Mediated Microbial Dysbiosis in Health and Disease: Lung-Gut Axis and Beyond. J Xenobiot 2024; 14:1595-1612. [PMID: 39449427 PMCID: PMC11503347 DOI: 10.3390/jox14040086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Growing evidence suggests physiological and pathological functions of lung and gut microbiomes in various pathologies. Epidemiological and experimental data associate air pollution exposure with host microbial dysbiosis in the lungs and gut. Air pollution through increased reactive oxygen species generation, the disruption of epithelial barrier integrity, and systemic inflammation modulates microbial imbalance. Microbiome balance is crucial in regulating inflammation and metabolic pathways to maintain health. Microbiome dysbiosis is proposed as a potential mechanism for the air-pollution-induced modulation of pulmonary and systemic disorders. Microbiome-based therapeutic approaches are increasingly gaining attention and could have added value in promoting lung health. This review summarizes and discusses air-pollution-mediated microbiome alterations in the lungs and gut in humans and mice and elaborates on their role in health and disease. We discuss and summarize the current literature, highlight important mechanisms that lead to microbial dysbiosis, and elaborate on pathways that potentially link lung and lung microbiomes in the context of environmental exposures. Finally, we discuss the lung-liver-gut axis and its potential pathophysiological implications in air-pollution-mediated pathologies through microbial dysbiosis.
Collapse
Affiliation(s)
- Md Habibul Hasan Mazumder
- Department of Physiology, Pharmacology & Toxicology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV 26506, USA
- Department of Pharmaceutical and Pharmacological Sciences, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA
| | - Salik Hussain
- Department of Physiology, Pharmacology & Toxicology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
12
|
Park BJ, Dhong KR, Park HJ. Cordyceps militaris Grown on Germinated Rhynchosia nulubilis (GRC) Encapsulated in Chitosan Nanoparticle (GCN) Suppresses Particulate Matter (PM)-Induced Lung Inflammation in Mice. Int J Mol Sci 2024; 25:10642. [PMID: 39408971 PMCID: PMC11477187 DOI: 10.3390/ijms251910642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Cordyceps militaris grown on germinated Rhynchosia nulubilis (GRC) exerts various biological effects, including anti-allergic, anti-inflammatory, and immune-regulatory effects. In this study, we investigated the anti-inflammatory effects of GRC encapsulated in chitosan nanoparticles (CN) against particulate matter (PM)-induced lung inflammation. Optimal CN (CN6) (CHI: TPP w/w ratio of 4:1; TPP pH 2) exhibited a zeta potential of +22.77 mV, suitable for GRC encapsulation. At different GRC concentrations, higher levels (60 and 120 mg/mL) led to increased negative zeta potential, enhancing stability. The optimal GRC concentration for maximum entrapment (31.4 ± 1.35%) and loading efficiency (7.6 ± 0.33%) of GRC encapsulated in CN (GCN) was 8 mg/mL with a diameter of 146.1 ± 54 nm and zeta potential of +30.68. In vivo studies revealed that administering 300 mg/kg of GCN significantly decreased the infiltration of macrophages and T cells in the lung tissues of PM-treated mice, as shown by immunohistochemical analysis of CD4 and F4/80 markers. Additionally, GCN ameliorated PM-induced lung tissue damage, inflammatory cell infiltration, and alveolar septal hypertrophy. GCN also decreased total cells and neutrophils, showing notable anti-inflammatory effects in the bronchoalveolar lavage fluid (BALF) from PM-exposed mice, compared to GRC. Next the anti-inflammatory properties of GCN were further explored in PM- and LPS-exposed RAW264.7 cells; it significantly reduced PM- and LPS-induced cell death, NO production, and levels of inflammatory cytokine mRNAs (IL-1β, IL-6, and COX-2). GCN also suppressed NF-κB/MAPK signaling pathways by reducing levels of p-NF-κB, p-ERK, and p-c-Jun proteins, indicating its potential in managing PM-related inflammatory lung disease. Furthermore, GCN significantly reduced PM- and LPS-induced ROS production. The enhanced bioavailability of GRC components was demonstrated by an increase in fluorescence intensity in the intestinal absorption study using FITC-GCN. Our data indicated that GCN exhibited enhanced bioavailability and potent anti-inflammatory and antioxidant effects in cells and in vivo, making it a promising candidate for mitigating PM-induced lung inflammation and oxidative stress.
Collapse
Affiliation(s)
- Byung-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| | - Kyu-Ree Dhong
- Magicbullettherapeutics Inc., 150 Yeongdeungpo-ro, Yeongdeungpo-gu, Seoul 07292, Republic of Korea;
| | - Hye-Jin Park
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Republic of Korea;
| |
Collapse
|
13
|
Park YC, Choi SY, Cha Y, Yoon HW, Son YM. Microbiome-Mucosal Immunity Nexus: Driving Forces in Respiratory Disease Progression. J Microbiol 2024; 62:709-725. [PMID: 39240507 DOI: 10.1007/s12275-024-00167-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/07/2024]
Abstract
The importance of the complex interplay between the microbiome and mucosal immunity, particularly within the respiratory tract, has gained significant attention due to its potential implications for the severity and progression of lung diseases. Therefore, this review summarizes the specific interactions through which the respiratory tract-specific microbiome influences mucosal immunity and ultimately impacts respiratory health. Furthermore, we discuss how the microbiome affects mucosal immunity, considering tissue-specific variations, and its capacity in respiratory diseases containing asthma, chronic obstructive pulmonary disease, and lung cancer. Additionally, we investigate the external factors which affect the relationship between respiratory microbiome and mucosal immune responses. By exploring these intricate interactions, this review provides valuable insights into the potential for microbiome-based interventions to modulate mucosal immunity and alleviate the severity of respiratory diseases.
Collapse
Affiliation(s)
- Young Chae Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Soo Yeon Choi
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Yunah Cha
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Hyeong Won Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Young Min Son
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
14
|
Zhou Q, Ma J, Biswal S, Rowan NR, London NR, Riley CA, Lee SE, Pinto JM, Ahmed OG, Su M, Liang Z, Du R, Ramanathan M, Zhang Z. Air pollution, genetic factors, and chronic rhinosinusitis: A prospective study in the UK Biobank. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 940:173526. [PMID: 38825199 DOI: 10.1016/j.scitotenv.2024.173526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a prevalent upper respiratory condition that manifests in two primary subtypes: CRS with nasal polyps (CRSwNP) and CRS without nasal polyps (CRSsNP). While previous studies indicate a correlation between air pollution and CRS, the role of genetic predisposition in this relationship remains largely unexplored. We hypothesized that higher air pollution exposure would lead to the development of CRS, and that genetic susceptibility might modify this association. METHODS This cohort study involving 367,298 adult participants from the UK Biobank, followed from March 2006 to October 2021. Air pollution metrics were estimated at residential locations using land-use regression models. Cox proportional hazard models were employed to explore the associations between air pollution exposure and CRS, CRSwNP, and CRSsNP. A polygenic risk score (PRS) was constructed to evaluate the joint effect of air pollution and genetic predisposition on the development of CRS. RESULTS We found that the risk of CRS increased under long-term exposure to PM2.5 [the hazard ratios (HRs) with 95 % CIs: 1.59 (1.26-2.01)], PM10 [1.64 (1.26-2.12)], NO2 [1.11 (1.04-1.17)], and NOx [1.18 (1.12-1.25)], respectively. These effects were more pronounced among participants with CRSwNP, although the differences were not statistically significant. Additionally, we found that the risks for CRS and CRSwNP increased in a graded manner among participants with higher PRS or higher exposure to PM2.5, PM10, or NOx concentrations. However, no multiplicative or additive interactions were observed. CONCLUSIONS Long-term exposure to air pollution increases the risk of CRS, particularly CRSwNP underscoring the need to prioritize clean air initiatives and environmental regulations.
Collapse
Affiliation(s)
- Qinfeng Zhou
- Department of Global Health, The Peking University School of Public Health, Beijing, China
| | - Junxiong Ma
- Department of Global Health, The Peking University School of Public Health, Beijing, China
| | - Shyam Biswal
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas R Rowan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nyall R London
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Charles A Riley
- Department of Otolaryngology-Head and Neck Surgery, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Stella E Lee
- Division of Otolaryngology, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jayant M Pinto
- Section of Otolaryngology, Department of Surgery, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | - Omar G Ahmed
- Department of Otolaryngology, Houston Methodist Academic Institute, Houston, TX, USA
| | - Mintao Su
- Department of Global Health, The Peking University School of Public Health, Beijing, China
| | - Zhisheng Liang
- Department of Global Health, The Peking University School of Public Health, Beijing, China
| | - Runming Du
- Department of Global Health, The Peking University School of Public Health, Beijing, China
| | - Murugappan Ramanathan
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Zhenyu Zhang
- Department of Global Health, The Peking University School of Public Health, Beijing, China; Institute for Global Health and Development, Peking University, Beijing, China; Institute of Carbon Neutrality, Peking University, Beijing, China.
| |
Collapse
|
15
|
Poniedziałek B, Rzymski P, Zarębska-Michaluk D, Flisiak R. Viral respiratory infections and air pollution: A review focused on research in Poland. CHEMOSPHERE 2024; 359:142256. [PMID: 38723686 DOI: 10.1016/j.chemosphere.2024.142256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/14/2024]
Abstract
The COVID-19 pandemic has reinforced an interest in the relationship between air pollution and respiratory viral infections, indicating that their burden can be increased under poor air quality. This paper reviews the pathways through which air pollutants can enhance susceptibility to such infections and aggravate their clinical course and outcome. It also summarizes the research exploring the links between various viral infections and exposure to solid and gaseous pollution in Poland, a region characterized by poor air quality, especially during a heating season. The majority of studies focused on concentrations of particulate matter (PM; 86.7%); the other pollutants, i.e., BaP, benzene, CO, NOx, O3, and SO2, were studied less often and sometimes only in the context of a particular infection type. Most research concerned COVID-19, showing that elevated levels of PM and NO2 correlated with higher morbidity and mortality, while increased PM2.5 and benzo[a]pyrene levels were related to worse clinical course and outcome in hospitalized, regardless of age and dominant SARS-CoV-2 variant. PM10 and PM2.5 levels were also associated with the incidence of influenza-like illness and, along with NO2 concentrations, with a higher rate of children's hospitalizations due to lower respiratory tract RSV infections. Higher levels of air pollutants also increased hospitalization due to bronchitis (PM, NOx, and O3) and emergency department admission due to viral croup (PM10, PM2.5, NOx, CO, and benzene). Although the conducted studies imply only correlations and have other limitations, as discussed in the present paper, it appears that improving air quality through reducing combustion processes in energy production in Poland should be perceived as a part of multilayered protection measures against respiratory viral infections, decreasing the healthcare costs of COVID-19, lower tract RSV infections, influenza, and other respiratory viral diseases prevalent between autumn and early spring, in addition to other health and climate benefits.
Collapse
Affiliation(s)
- Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.
| | | | - Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University of Białystok, Białystok, Poland.
| |
Collapse
|
16
|
Weidinger D, Jacobsen J, Alisch D, Uebner H, Heinen N, Greune L, Westhoven S, Jamal Jameel K, Kronsbein J, Pfaender S, Taube C, Reuter S, Peters M, Hatt H, Knobloch J. Olfactory receptors impact pathophysiological processes of lung diseases in bronchial epithelial cells. Eur J Cell Biol 2024; 103:151408. [PMID: 38583306 DOI: 10.1016/j.ejcb.2024.151408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Therapeutic options for steroid-resistant non-type 2 inflammation in obstructive lung diseases are limited. Bronchial epithelial cells are key in the pathogenesis by releasing the central proinflammatory cytokine interleukine-8 (IL-8). Olfactory receptors (ORs) are expressed in various cell types. This study examined the drug target potential of ORs by investigating their impact on associated pathophysiological processes in lung epithelial cells. METHODS Experiments were performed in the A549 cell line and in primary human bronchial epithelial cells. OR expression was investigated using RT-PCR, Western blot, and immunocytochemical staining. OR-mediated effects were analyzed by measuring 1) intracellular calcium concentration via calcium imaging, 2) cAMP concentration by luminescence-based assays, 3) wound healing by scratch assays, 4) proliferation by MTS-based assays, 5) cellular vitality by Annexin V/PI-based FACS staining, and 6) the secretion of IL-8 in culture supernatants by ELISA. RESULTS By screening 100 potential OR agonists, we identified two, Brahmanol and Cinnamaldehyde, that increased intracellular calcium concentrations. The mRNA and proteins of the corresponding receptors OR2AT4 and OR2J3 were detected. Stimulation of OR2J3 with Cinnamaldehyde reduced 1) IL-8 in the absence and presence of bacterial and viral pathogen-associated molecular patterns (PAMPs), 2) proliferation, and 3) wound healing but increased cAMP. In contrast, stimulation of OR2AT4 by Brahmanol increased wound healing but did not affect cAMP and proliferation. Both ORs did not influence cell vitality. CONCLUSION ORs might be promising drug target candidates for lung diseases with non-type 2 inflammation. Their stimulation might reduce inflammation or prevent tissue remodeling by promoting wound healing.
Collapse
Affiliation(s)
- Daniel Weidinger
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Julian Jacobsen
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Desiree Alisch
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Hendrik Uebner
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Tüschener Weg 40, Essen 45239, Germany
| | - Natalie Heinen
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Lea Greune
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Saskia Westhoven
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany; Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Kaschin Jamal Jameel
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum 44801, Germany; Research Unit Emerging Viruses, Leibniz Institute of Virology (LIV), Hamburg, Germany; University of Lübeck, Lübeck, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Tüschener Weg 40, Essen 45239, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Tüschener Weg 40, Essen 45239, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, Bochum 44801, Germany
| | - Hanns Hatt
- Cell Physiology ND4/35, Ruhr-University Bochum, Universitätsstraße 150, Bochum 44801, Germany
| | - Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-la-Camp-Platz 1, Bochum 44789, Germany.
| |
Collapse
|
17
|
Simões M, Zorn J, Hogerwerf L, Velders GJM, Portengen L, Gerlofs-Nijland M, Dijkema M, Strak M, Jacobs J, Wesseling J, de Vries WJ, Mijnen-Visser S, Smit LAM, Vermeulen R, Mughini-Gras L. Outdoor air pollution as a risk factor for testing positive for SARS-CoV-2: A nationwide test-negative case-control study in the Netherlands. Int J Hyg Environ Health 2024; 259:114382. [PMID: 38652943 DOI: 10.1016/j.ijheh.2024.114382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Air pollution is a known risk factor for several diseases, but the extent to which it influences COVID-19 compared to other respiratory diseases remains unclear. We performed a test-negative case-control study among people with COVID-19-compatible symptoms who were tested for SARS-CoV-2 infection, to assess whether their long- and short-term exposure to ambient air pollution (AAP) was associated with testing positive (vs. negative) for SARS-CoV-2. We used individual-level data for all adult residents in the Netherlands who were tested for SARS-CoV-2 between June and November 2020, when only symptomatic people were tested, and modeled ambient concentrations of PM10, PM2.5, NO2 and O3 at geocoded residential addresses. In long-term exposure analysis, we selected individuals who did not change residential address in 2017-2019 (1.7 million tests) and considered the average concentrations of PM10, PM2.5 and NO2 in that period, and different sources of PM (industry, livestock, other agricultural activities, road traffic, other Dutch sources, foreign sources). In short-term exposure analysis, individuals not changing residential address in the two weeks before testing day (2.7 million tests) were included in the analyses, thus considering 1- and 2-week average concentrations of PM10, PM2.5, NO2 and O3 before testing day as exposure. Mixed-effects logistic regression analysis with adjustment for several confounders, including municipality and testing week to account for spatiotemporal variation in viral circulation, was used. Overall, there was no statistically significant effect of long-term exposure to the studied pollutants on the odds of testing positive vs. negative for SARS-CoV-2. However, significant positive associations of long-term exposure to PM10 and PM2.5 from specifically foreign and livestock sources, and to PM10 from other agricultural sources, were observed. Short-term exposure to PM10 (adjusting for NO2) and PM2.5 were also positively associated with increased odds of testing positive for SARS-CoV-2. While these exposures seemed to increase COVID-19 risk relative to other respiratory diseases, the underlying biological mechanisms remain unclear. This study reinforces the need to continue to strive for better air quality to support public health.
Collapse
Affiliation(s)
- Mariana Simões
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Jelle Zorn
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control (CIb), Bilthoven, the Netherlands
| | - Lenny Hogerwerf
- National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control (CIb), Bilthoven, the Netherlands
| | - Guus J M Velders
- Institute for Marine and Atmospheric Research Utrecht, Utrecht University, Utrecht, the Netherlands; National Institute for Public Health and the Environment (RIVM), Center for Environmental Quality (MIL), Bilthoven, the Netherlands
| | - Lützen Portengen
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Miriam Gerlofs-Nijland
- National Institute for Public Health and the Environment (RIVM), Center for Sustainability, Environment and Health (DMG), Bilthoven, the Netherlands
| | - Marieke Dijkema
- Municipal Health Services, Provinces of Overijssel and Gelderland, the Netherlands
| | - Maciek Strak
- National Institute for Public Health and the Environment (RIVM), Center for Sustainability, Environment and Health (DMG), Bilthoven, the Netherlands
| | - José Jacobs
- National Institute for Public Health and the Environment (RIVM), Center for Sustainability, Environment and Health (DMG), Bilthoven, the Netherlands
| | - Joost Wesseling
- National Institute for Public Health and the Environment (RIVM), Center for Environmental Quality (MIL), Bilthoven, the Netherlands
| | - Wilco J de Vries
- National Institute for Public Health and the Environment (RIVM), Center for Environmental Quality (MIL), Bilthoven, the Netherlands
| | - Suzanne Mijnen-Visser
- National Institute for Public Health and the Environment (RIVM), Center for Environmental Quality (MIL), Bilthoven, the Netherlands
| | - Lidwien A M Smit
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Roel Vermeulen
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Lapo Mughini-Gras
- Institute for Risk Assessment Sciences (IRAS), Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; National Institute for Public Health and the Environment (RIVM), Centre for Infectious Disease Control (CIb), Bilthoven, the Netherlands.
| |
Collapse
|
18
|
Ruggles A, Benakis C. Exposure to Environmental Toxins: Potential Implications for Stroke Risk via the Gut- and Lung-Brain Axis. Cells 2024; 13:803. [PMID: 38786027 PMCID: PMC11119296 DOI: 10.3390/cells13100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Recent evidence indicates that exposure to environmental toxins, both short-term and long-term, can increase the risk of developing neurological disorders, including neurodegenerative diseases (i.e., Alzheimer's disease and other dementias) and acute brain injury (i.e., stroke). For stroke, the latest systematic analysis revealed that exposure to ambient particulate matter is the second most frequent stroke risk after high blood pressure. However, preclinical and clinical stroke investigations on the deleterious consequences of environmental pollutants are scarce. This review examines recent evidence of how environmental toxins, absorbed along the digestive tract or inhaled through the lungs, affect the host cellular response. We particularly address the consequences of environmental toxins on the immune response and the microbiome at the gut and lung barrier sites. Additionally, this review highlights findings showing the potential contribution of environmental toxins to an increased risk of stroke. A better understanding of the biological mechanisms underlying exposure to environmental toxins has the potential to mitigate stroke risk and other neurological disorders.
Collapse
Affiliation(s)
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, 81337 Munich, Germany;
| |
Collapse
|
19
|
Lu HF, Zhou YC, Yang LT, Zhou Q, Wang XJ, Qiu SQ, Cheng BH, Zeng XH. Involvement and repair of epithelial barrier dysfunction in allergic diseases. Front Immunol 2024; 15:1348272. [PMID: 38361946 PMCID: PMC10867171 DOI: 10.3389/fimmu.2024.1348272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
The epithelial barrier serves as a critical defense mechanism separating the human body from the external environment, fulfilling both physical and immune functions. This barrier plays a pivotal role in shielding the body from environmental risk factors such as allergens, pathogens, and pollutants. However, since the 19th century, the escalating threats posed by environmental pollution, global warming, heightened usage of industrial chemical products, and alterations in biodiversity have contributed to a noteworthy surge in allergic disease incidences. Notably, allergic diseases frequently exhibit dysfunction in the epithelial barrier. The proposed epithelial barrier hypothesis introduces a novel avenue for the prevention and treatment of allergic diseases. Despite increased attention to the role of barrier dysfunction in allergic disease development, numerous questions persist regarding the mechanisms underlying the disruption of normal barrier function. Consequently, this review aims to provide a comprehensive overview of the epithelial barrier's role in allergic diseases, encompassing influencing factors, assessment techniques, and repair methodologies. By doing so, it seeks to present innovative strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Hui-Fei Lu
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Yi-Chi Zhou
- Department of Gastroenterology, Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, China
| | - Li-Tao Yang
- Clinical Laboratory Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen, China
| | - Qian Zhou
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xi-Jia Wang
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Shu-Qi Qiu
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Bao-Hui Cheng
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| | - Xian-Hai Zeng
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
- Department of Otolaryngology, Longgang Otolaryngology Hospital & Shenzhen Key Laboratory of Otolaryngology, Institute of Otolaryngology Shenzhen, Shenzhen, China
| |
Collapse
|
20
|
Lopes CDH, Antonacio FF, Moraes PMG, Asprino PF, Galante PAF, Jardim DL, de Macedo MP, Sandoval RL, Katz A, de Castro G, Achatz MI. The Clinical and Molecular Profile of Lung Cancer Patients Harboring the TP53 R337H Germline Variant in a Brazilian Cancer Center: The Possible Mechanism of Carcinogenesis. Int J Mol Sci 2023; 24:15035. [PMID: 37894716 PMCID: PMC10606350 DOI: 10.3390/ijms242015035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
In southern and southeastern Brazil, the TP53 founder variant c.1010G>A (R337H) has been previously documented with a prevalence of 0.3% within the general population and linked to a heightened incidence of lung adenocarcinomas (LUADs). In the present investigation, we cover clinical and molecular characterizations of lung cancer patients from the Brazilian Li-Fraumeni Syndrome Study (BLISS) database. Among the 175 diagnosed malignant neoplasms, 28 (16%) were classified as LUADs, predominantly occurring in females (68%), aged above 50 years, and never-smokers (78.6%). Significantly, LUADs manifested as the initial clinical presentation of Li-Fraumeni Syndrome in 78.6% of cases. Molecular profiling was available for 20 patients, with 14 (70%) revealing EGFR family alterations. In total, 23 alterations in cancer driver genes were identified, comprising 7 actionable mutations and 4 linked to resistance against systemic treatments. In conclusion, the carriers of TP53 R337H demonstrate a predisposition to LUAD development. Furthermore, our results indicate that environmental pollution potentially impacts the carcinogenesis of lung tumors in the carriers of TP53 R337H.
Collapse
Affiliation(s)
- Carlos D. H. Lopes
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Fernanda F. Antonacio
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Priscila M. G. Moraes
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Paula F. Asprino
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Pedro A. F. Galante
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Denis L. Jardim
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
- Oncoclínicas, São Paulo 04543-906, Brazil
| | - Mariana P. de Macedo
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Renata L. Sandoval
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Artur Katz
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Gilberto de Castro
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| | - Maria Isabel Achatz
- Hospital Sirio Libanes, São Paulo 01308-050, Brazil; (F.F.A.); (P.M.G.M.); (P.F.A.); (P.A.F.G.); (D.L.J.); (M.P.d.M.); (R.L.S.); (A.K.); (G.d.C.J.)
| |
Collapse
|
21
|
Somayajulu M, McClellan SA, Muhammed F, Wright R, Hazlett LD. PM 10 and Pseudomonas aeruginosa: effects on corneal epithelium. Front Cell Infect Microbiol 2023; 13:1240903. [PMID: 37868351 PMCID: PMC10585254 DOI: 10.3389/fcimb.2023.1240903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose In vivo data indicate that mouse corneas exposed to PM10 showed early perforation and thinning after infection with Pseudomonas aeruginosa. To understand the mechanisms underlying this finding, we tested the effects of PM10 and the mitochondria targeted anti-oxidant SKQ1 in immortalized human corneal epithelial cells (HCET) that were challenged with Pseudomonas aeruginosa strain 19660. Methods Mouse corneas were infected with strain 19660 after a 2 week whole-body exposure to PM10 or control air and assessed by clinical scores, slit lamp photography and western blot. HCET were exposed to 100μg/ml PM10 for 24h before challenge with strain 19660 (MOI 20). A subset of cells were pre-treated with 50nM SKQ1 for 1h before PM10 exposure. Phase contrast microscopy was used to study cell morphology, cell viability was measured by an MTT assay, and ROS by DCFH-DA. Levels of pro-inflammatory markers and anti-oxidant enzymes were evaluated by RT-PCR, western blot and ELISA. Reduced glutathione (GSH) and malondialdehyde (MDA) levels were evaluated by assay kits. Results In vivo, whole body exposure to PM10 vs. control air exposed mouse corneas showed early perforation and/or corneal thinning at 3 days post infection, accompanied by increased TNF-α and decreased SOD2 protein levels. In vitro, PM10 induced a dose dependent reduction in cell viability of HCET and significantly increased mRNA levels of pro-inflammatory molecules compared to control. Exposure to PM10 before bacterial challenge further amplified the reduction in cell viability and GSH levels. Furthermore, PM10 exposure also exacerbated the increase in MDA and ROS levels and phase contrast microscopy revealed more rounded cells after strain 19660 challenge. PM10 exposure also further increased the mRNA and protein levels of pro-inflammatory molecules, while anti-inflammatory IL-10 was decreased. SKQ1 reversed the rounded cell morphology observed by phase contrast microscopy, increased levels of MDA, ROS and pro-inflammatory molecules, and restored IL-10. Conclusions PM10 induces decreased cell viability, oxidative stress and inflammation in HCET and has an additive effect upon bacterial challenge. SKQ1 protects against oxidative stress and inflammation induced by PM10 after bacterial challenge by reversing these effects. The findings provide insight into mechanisms underlying early perforation and thinning observed in infected corneas of PM10 exposed mice.
Collapse
Affiliation(s)
| | | | | | | | - Linda D. Hazlett
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, School of Medicine, Detroit, MI, United States
| |
Collapse
|
22
|
Holme JA, Vondráček J, Machala M, Lagadic-Gossmann D, Vogel CFA, Le Ferrec E, Sparfel L, Øvrevik J. Lung cancer associated with combustion particles and fine particulate matter (PM 2.5) - The roles of polycyclic aromatic hydrocarbons (PAHs) and the aryl hydrocarbon receptor (AhR). Biochem Pharmacol 2023; 216:115801. [PMID: 37696458 PMCID: PMC10543654 DOI: 10.1016/j.bcp.2023.115801] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Air pollution is the leading cause of lung cancer after tobacco smoking, contributing to 20% of all lung cancer deaths. Increased risk associated with living near trafficked roads, occupational exposure to diesel exhaust, indoor coal combustion and cigarette smoking, suggest that combustion components in ambient fine particulate matter (PM2.5), such as polycyclic aromatic hydrocarbons (PAHs), may be central drivers of lung cancer. Activation of the aryl hydrocarbon receptor (AhR) induces expression of xenobiotic-metabolizing enzymes (XMEs) and increase PAH metabolism, formation of reactive metabolites, oxidative stress, DNA damage and mutagenesis. Lung cancer tissues from smokers and workers exposed to high combustion PM levels contain mutagenic signatures derived from PAHs. However, recent findings suggest that ambient air PM2.5 exposure primarily induces lung cancer development through tumor promotion of cells harboring naturally acquired oncogenic mutations, thus lacking typical PAH-induced mutations. On this background, we discuss the role of AhR and PAHs in lung cancer development caused by air pollution focusing on the tumor promoting properties including metabolism, immune system, cell proliferation and survival, tumor microenvironment, cell-to-cell communication, tumor growth and metastasis. We suggest that the dichotomy in lung cancer patterns observed between smoking and outdoor air PM2.5 represent the two ends of a dose-response continuum of combustion PM exposure, where tumor promotion in the peripheral lung appears to be the driving factor at the relatively low-dose exposures from ambient air PM2.5, whereas genotoxicity in the central airways becomes increasingly more important at the higher combustion PM levels encountered through smoking and occupational exposure.
Collapse
Affiliation(s)
- Jørn A Holme
- Department of Air Quality and Noise, Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box PO Box 222 Skøyen, 0213 Oslo, Norway
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic
| | - Miroslav Machala
- Department of Pharmacology and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Dominique Lagadic-Gossmann
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Lydie Sparfel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France
| | - Johan Øvrevik
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, PO Box 1066 Blindern, 0316 Oslo, Norway; Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box 222 Skøyen, 0213 Oslo, Norway.
| |
Collapse
|
23
|
Kewcharoenwong C, Khongmee A, Nithichanon A, Palaga T, Prueksasit T, Mudway IS, Hawrylowicz CM, Lertmemongkolchai G. Vitamin D3 regulates PM-driven primary human neutrophil inflammatory responses. Sci Rep 2023; 13:15850. [PMID: 37740033 PMCID: PMC10516903 DOI: 10.1038/s41598-023-43252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023] Open
Abstract
Recent evidence has demonstrated that both acute and chronic exposure to particulate air pollution are risk factors for respiratory tract infections and increased mortality from sepsis. There is therefore an urgent need to establish the impact of ambient particulate matter (PM) on innate immune cells and to establish potential strategies to mitigate against adverse effects. PM has previously been reported to have potential adverse effects on neutrophil function. In the present study, we investigated the impact of standard urban PM (SRM1648a, NIST) and PM2.5 collected from Chiang Mai, Thailand, on human peripheral blood neutrophil functions, including LPS-induced migration, IL-8 production, and bacterial killing. Both NIST and the PM2.5, being collected in Chiang Mai, Thailand, increased IL-8 production, but reduced CXCR2 expression and migration of human primary neutrophils stimulated with Escherichia coli LPS. Moreover, PM-pretreated neutrophils from vitamin D-insufficient participants showed reduced E. coli-killing activity. Furthermore, in vitro vitamin D3 supplementation attenuated IL-8 production and improved bacterial killing by cells from vitamin D-insufficient participants. Our findings suggest that provision of vitamin D to individuals with insufficiency may attenuate adverse acute neutrophilic responses to ambient PM.
Collapse
Affiliation(s)
- Chidchamai Kewcharoenwong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Aranya Khongmee
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Arnone Nithichanon
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Tassanee Prueksasit
- Department of Environmental Science, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Ian S Mudway
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute of Health Research, Health Protection Research Unit in Environmental Exposures and Health, Imperial College London and King's College London, London, W12 OBZ, UK
| | - Catherine M Hawrylowicz
- King's Centre for Lung Health, School of Immunology and Microbial Sciences, King's College London, London, W2 1PG, UK
- National Institute of Health Research, Health Protection Research Unit in Environmental Exposures and Health, Imperial College London and King's College London, London, W12 OBZ, UK
| | - Ganjana Lertmemongkolchai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
24
|
Kim HR, Ingram JL, Que LG. Effects of Oxidative Stress on Airway Epithelium Permeability in Asthma and Potential Implications for Patients with Comorbid Obesity. J Asthma Allergy 2023; 16:481-499. [PMID: 37181453 PMCID: PMC10171222 DOI: 10.2147/jaa.s402340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/15/2023] [Indexed: 05/16/2023] Open
Abstract
20 million adults and 4.2 million children in the United States have asthma, a disease resulting in inflammation and airway obstruction in response to various factors, including allergens and pollutants and nonallergic triggers. Obesity, another highly prevalent disease in the US, is a major risk factor for asthma and a significant cause of oxidative stress throughout the body. People with asthma and comorbid obesity are susceptible to developing severe asthma that cannot be sufficiently controlled with current treatments. More research is needed to understand how asthma pathobiology is affected when the patient has comorbid obesity. Because the airway epithelium directly interacts with the outside environment and interacts closely with the immune system, understanding how the airway epithelium of patients with asthma and comorbid obesity is altered compared to that of lean asthma patients will be crucial for developing more effective treatments. In this review, we discuss how oxidative stress plays a role in two chronic inflammatory diseases, obesity and asthma, and propose a mechanism for how these conditions may compromise the airway epithelium.
Collapse
Affiliation(s)
- Haein R Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jennifer L Ingram
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, USA
| | - Loretta G Que
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
25
|
Zhang J, Cheng H, Di Narzo A, Zhu Y, Xie S, Shao X, Zhang Z, Chung SK, Hao K. Profiling Microbiota from Multiple Sites in the Respiratory Tract to Identify a Biomarker for PM 2.5 Nitrate Exposure-Induced Pulmonary Damages. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:7346-7357. [PMID: 37133311 DOI: 10.1021/acs.est.2c08807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The microbiota present in the respiratory tract (RT) responds to environmental stimuli and engages in a continuous interaction with the host immune system to maintain homeostasis. A total of 40 C57BL/6 mice were divided into four groups and exposed to varying concentrations of PM2.5 nitrate aerosol and clean air. After 10 weeks of exposure, assessments were conducted on the lung and airway microbiome, lung functions, and pulmonary inflammation. Additionally, we analyzed data from both mouse and human respiratory tract (RT) microbiomes to identify possible biomarkers for PM2.5 exposure-induced pulmonary damages. On average, 1.5 and 13.5% inter-individual microbiome variations in the lung and airway were explained by exposure, respectively. In the airway, among the 60 bacterial OTUs (operational taxonomic units) > 0.05% proportion, 40 OTUs were significantly affected by PM2.5 exposure (FDR ≤ 10%). Further, the airway microbiome was associated with peak expiratory flow (PEF) (p = 0.003), pulmonary neutrophil counts (p = 0.01), and alveolar 8-OHdG oxidative lesions (p = 0.0078). The Clostridiales order bacteria showed the strongest signals. For example, the o_Clostridiales;f_;g_ OTU was elevated by PM2.5 nitrate exposure (p = 4.98 × 10-5) and negatively correlated with PEF (r = -0.585 and p = 2.4 × 10-4). It was also associated with the higher pulmonary neutrophil count (p = 8.47 × 10-5) and oxidative lesion (p = 7.17 × 10-3). In human data, we confirmed the association of airway Clostridiales order bacteria with PM2.5 exposure and lung function. For the first time, this study characterizes the impact of PM2.5 exposure on the microbiome of multiple sites in the respiratory tract (RT) and its relevance to airflow obstructive diseases. By analyzing data from both humans and mice, we have identified bacteria belonging to the Clostridiales order as a promising biomarker for PM2.5 exposure-induced decline in pulmonary function and inflammation.
Collapse
Affiliation(s)
- Jushan Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, Shanghai 200072, China
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Yujie Zhu
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Shuanshuan Xie
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xiaowen Shao
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Zhongyang Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Sookja Kim Chung
- Medical Faculty, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| | - Ke Hao
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| |
Collapse
|
26
|
Chen Y, Wu Y, Qi Y, Liu S. Cell Death Pathways: The Variable Mechanisms Underlying Fine Particulate Matter-Induced Cytotoxicity. ACS NANOSCIENCE AU 2023; 3:130-139. [PMID: 37101591 PMCID: PMC10125306 DOI: 10.1021/acsnanoscienceau.2c00059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 04/28/2023]
Abstract
Recently, the advent of health risks due to the cytotoxicity of fine particulate matter (FPM) is concerning. Numerous studies have reported abundant data elucidating the FPM-induced cell death pathways. However, several challenges and knowledge gaps are still confronted nowadays. On one hand, the undefined components of FPM (such as heavy metals, polycyclic aromatic hydrocarbons, and pathogens) are all responsible for detrimental effects, thus rendering it difficult to delineate the specific roles of these copollutants. On the other hand, owing to the crosstalk and interplay among different cell death signaling pathways, precisely determining the threats and risks posed by FPM is difficult. Herein, we recapitulate the current knowledge gaps present in the recent studies regarding FPM-induced cell death, and propose future research directions for policy-making to prevent FPM-induced diseases and improve knowledge concerning the adverse outcome pathways and public health risks of FPM.
Collapse
Affiliation(s)
- Yucai Chen
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Yu Qi
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Sijin Liu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
- Science
and Technology Innovation Center, Shandong
First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| |
Collapse
|
27
|
Wang J, Yan Y, Si H, Li J, Zhao Y, Gao T, Pi J, Zhang R, Chen R, Chen W, Zheng Y, Jiang M. The effect of real-ambient PM2.5 exposure on the lung and gut microbiomes and the regulation of Nrf2. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114702. [PMID: 36950983 DOI: 10.1016/j.ecoenv.2023.114702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 06/18/2023]
Abstract
The influence of air pollution on human health has sparked widespread concerns across the world. Previously, we found that exposure to ambient fine particulate matter (PM2.5) in our "real-ambient exposure" system can result in reduced lung function. However, the mechanism of organ-specific toxicity is still not fully elucidated. The balance of the microbiome contributes to maintaining lung and gut health, but the changes in the microbiome under PM2.5 exposure are not fully understood. Recently, crosstalk between nuclear factor E2-related factor 2 (Nrf2) and the microbiome was reported. However, it is unclear whether Nrf2 affects the lung and gut microbiomes under PM2.5 exposure. In this study, wild-type (WT) and Nrf2-/- (KO) mice were exposed to filtered air (FA) and real ambient PM2.5 (PM) in the " real-ambient exposure" system to examine changes in the lung and gut microbiomes. Here, our data suggested microbiome dysbiosis in lung and gut of KO mice under PM2.5 exposure, and Nrf2 ameliorated the microbiome disorder. Our study demonstrated the detrimental impacts of PM2.5 on the lung and gut microbiome by inhaled exposure to air pollution and supported the protective role of Nrf2 in maintaining microbiome homeostasis under PM2.5 exposure.
Collapse
Affiliation(s)
- Jianxin Wang
- School of Public Health, Qingdao University, Qingdao, China
| | - Yongwei Yan
- Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture and Rural Affairs, Yellow Sea fisheries research institute, Chinese Academy of Fishery Sciences, Qingdao, Shandong, China
| | - Honglin Si
- School of Public Health, Qingdao University, Qingdao, China
| | - Jianyu Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Yanjie Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Tianlin Gao
- School of Public Health, Qingdao University, Qingdao, China
| | - Jingbo Pi
- School of Public Health, China Medical University, Shenyang, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yaseen University, Guangzhou, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Menghui Jiang
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
28
|
Young TL, Scieszka D, Begay JG, Lucas SN, Herbert G, Zychowski K, Hunter R, Salazar R, Ottens AK, Erdely A, Gu H, Campen MJ. Aging influence on pulmonary and systemic inflammation and neural metabolomics arising from pulmonary multi-walled carbon nanotube exposure in apolipoprotein E-deficient and C57BL/6 female mice. Inhal Toxicol 2023; 35:86-100. [PMID: 35037817 PMCID: PMC10037439 DOI: 10.1080/08958378.2022.2026538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/03/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Environmental exposures exacerbate age-related pathologies, such as cardiovascular and neurodegenerative diseases. Nanoparticulates, and specifically carbon nanomaterials, are a fast-growing contributor to the category of inhalable pollutants, whose risks to health are only now being unraveled. The current study assessed the exacerbating effect of age on multiwalled-carbon nanotube (MWCNT) exposure in young and old C57BL/6 and ApoE-/- mice. MATERIALS AND METHODS Female C57BL/6 and apolipoprotein E-deficient (ApoE-/-) mice, aged 8 weeks and 15 months, were exposed to 0 or 40 µg MWCNT via oropharyngeal aspiration. Pulmonary inflammation, inflammatory bioactivity of serum, and neurometabolic changes were assessed at 24 h post-exposure. RESULTS Pulmonary neutrophil infiltration was induced by MWCNT in bronchoalveolar lavage fluid in both C57BL/6 and ApoE-/-. Macrophage counts decreased with MWCNT exposure in ApoE-/- mice but were unaffected by exposure in C57BL/6 mice. Older mice appeared to have greater MWCNT-induced total protein in lavage fluid. BALF cytokines and chemokines were elevated with MWCNT exposure, but CCL2, CXCL1, and CXCL10 showed reduced responses to MWCNT in older mice. However, no significant serum inflammatory bioactivity was detected. Cerebellar metabolic changes in response to MWCNT were modest, but age and strain significantly influenced metabolite profiles assessed. ApoE-/- mice and older mice exhibited less robust metabolite changes in response to exposure, suggesting a reduced health reserve. CONCLUSIONS Age influences the pulmonary and neurological responses to short-term MWCNT exposure. However, with only the model of moderate aging (15 months) in this study, the responses appeared modest compared to inhaled toxicant impacts in more advanced aging models.
Collapse
Affiliation(s)
- Tamara L. Young
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - David Scieszka
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Jessica G. Begay
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Selita N. Lucas
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Guy Herbert
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | | | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Raul Salazar
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| | - Andrew K. Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, Richmond, VA 23298
| | - Aaron Erdely
- Pathology and Physiology Research Branch, National Institute for Occupational Safety and Health, Morgantown, WV 26505
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, US 85004
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
29
|
Chen SJ, Huang Y, Yu F, Feng X, Zheng YY, Li Q, Niu Q, Jiang YH, Zhao LQ, Wang M, Cheng PP, Song LJ, Liang LM, He XL, Xiong L, Xiang F, Wang X, Ma WL, Ye H. BMAL1/p53 mediating bronchial epithelial cell autophagy contributes to PM2.5-aggravated asthma. Cell Commun Signal 2023; 21:39. [PMID: 36803515 PMCID: PMC9940367 DOI: 10.1186/s12964-023-01057-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Fine particulate matter (PM2.5) is associated with increased incidence and severity of asthma. PM2.5 exposure disrupts airway epithelial cells, which elicits and sustains PM2.5-induced airway inflammation and remodeling. However, the mechanisms underlying development and exacerbation of PM2.5-induced asthma were still poorly understood. The aryl hydrocarbon receptor nuclear translocator-like protein 1 (BMAL1) is a major circadian clock transcriptional activator that is also extensively expressed in peripheral tissues and plays a crucial role in organ and tissue metabolism. RESULTS In this study, we found PM2.5 aggravated airway remodeling in mouse chronic asthma, and exacerbated asthma manifestation in mouse acute asthma. Next, low BMAL1 expression was found to be crucial for airway remodeling in PM2.5-challenged asthmatic mice. Subsequently, we confirmed that BMAL1 could bind and promote ubiquitination of p53, which can regulate p53 degradation and block its increase under normal conditions. However, PM2.5-induced BMAL1 inhibition resulted in up-regulation of p53 protein in bronchial epithelial cells, then increased-p53 promoted autophagy. Autophagy in bronchial epithelial cells mediated collagen-I synthesis as well as airway remodeling in asthma. CONCLUSIONS Taken together, our results suggest that BMAL1/p53-mediated bronchial epithelial cell autophagy contributes to PM2.5-aggravated asthma. This study highlights the functional importance of BMAL1-dependent p53 regulation during asthma, and provides a novel mechanistic insight into the therapeutic mechanisms of BMAL1. Video Abstract.
Collapse
Affiliation(s)
- Shuai-Jun Chen
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Yi Huang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Yuan-Yi Zheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Qian Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Ye-Han Jiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Li-Qin Zhao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China
| | - Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Xiaorong Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China. .,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China.
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hang Kong Road, Wuhan, 430030, China. .,Key Laboratory of Respiratory Diseases, National Health Commission of China, Wuhan, China.
| |
Collapse
|
30
|
Burbank AJ. Risk Factors for Respiratory Viral Infections: A Spotlight on Climate Change and Air Pollution. J Asthma Allergy 2023; 16:183-194. [PMID: 36721739 PMCID: PMC9884560 DOI: 10.2147/jaa.s364845] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Climate change has both direct and indirect effects on human health, and some populations are more vulnerable to these effects than others. Viral respiratory infections are most common illnesses in humans, with estimated 17 billion incident infections globally in 2019. Anthropogenic drivers of climate change, chiefly the emission of greenhouse gases and toxic pollutants from burning of fossil fuels, and the consequential changes in temperature, precipitation, and frequency of extreme weather events have been linked with increased susceptibility to viral respiratory infections. Air pollutants like nitrogen dioxide, particulate matter, diesel exhaust particles, and ozone have been shown to impact susceptibility and immune responses to viral infections through various mechanisms, including exaggerated or impaired innate and adaptive immune responses, disruption of the airway epithelial barrier, altered cell surface receptor expression, and impaired cytotoxic function. An estimated 90% of the world's population is exposed to air pollution, making this a topic with high relevance to human health. This review summarizes the available epidemiologic and experimental evidence for an association between climate change, air pollution, and viral respiratory infection.
Collapse
Affiliation(s)
- Allison J Burbank
- Division of Pediatric Allergy and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Correspondence: Allison J Burbank, 5008B Mary Ellen Jones Building, 116 Manning Dr, CB#7231, Chapel Hill, NC, 27599, USA, Tel +1 919 962 5136, Fax +1 919 962 4421, Email
| |
Collapse
|
31
|
Jeong YJ, Kim CU, Lee KS, Kim JH, Park SY, Jeong AY, Lee JB, Kim DJ, Park YJ, Lee MS. Pseudomonas stutzeri PM101005 inhaled with atmospheric particulate matter induces lung damage through inflammatory responses. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 317:120741. [PMID: 36435285 DOI: 10.1016/j.envpol.2022.120741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 06/16/2023]
Abstract
Atmospheric particulate matter (PM) contains a mixture of chemical and biological elements that pose threat to human health by increasing susceptibility to respiratory diseases. Although the identification of the microorganisms composing the PM has been assessed, their immunological impacts are still questionable. Here, we examined the mechanisms responsible for the pathogenicity of Pseudomonas stutzeri PM101005 (PMPS), a bacterium isolated from fine dust, in lung epithelial cells, alveolar cells, and macrophages. Relative to its comparative strain Pseudomonas stutzeri (PS), infections with PMPS induced higher production of inflammatory cytokines and chemokines, mediated by the activation of NF-κB and MAPK signaling pathways. Additionally, with three-dimensional (3D) airway spheroids which mimic the human bronchial epithelium, we confirmed that PMPS infections lead to relatively higher induction of pro-inflammatory cytokines than PM infections. Consistent results were observed in murine models as the infections with PMPS provoked greater inflammatory responses than the infections with PS. These PMPS-induced responses were mediated by the signaling pathways of the Toll-like receptors (TLRs), which regulated PMPS infection and played an important role in the expression of the antibiotic peptide β-defensin 3 (BD3) that suppressed PMPS proliferation. Moreover, PM pretreatment enhanced inflammatory responses and tissue damage of PMPS, while reducing BD3 expression. Overall, these results indicate that PM-isolated PMPS induce TLR-mediated inflammatory responses in lung tissues, and contributes to the understanding of the etiology of PM-induced respiratory damage.
Collapse
Affiliation(s)
- Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Ji Hyung Kim
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120, Republic of Korea
| | - Seo Young Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Ahn Young Jeong
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea; Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Jun Bong Lee
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Kangwon, 24341, Republic of Korea
| | - Doo-Jin Kim
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea; Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Young-Jun Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
32
|
Effect of Quercetin on mitoBK Ca Channel and Mitochondrial Function in Human Bronchial Epithelial Cells Exposed to Particulate Matter. Int J Mol Sci 2022; 24:ijms24010638. [PMID: 36614079 PMCID: PMC9820441 DOI: 10.3390/ijms24010638] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Particulate matter (PM) exposure increases reactive oxygen species (ROS) levels. It can lead to inflammatory responses and damage of the mitochondria thus inducing cell death. Recently, it has been shown that potassium channels (mitoK) located in the inner mitochondrial membrane are involved in cytoprotection, and one of the mechanisms involves ROS. To verify the cytoprotective role of mitoBKCa, we performed a series of experiments using a patch-clamp, transepithelial electrical resistance assessment (TEER), mitochondrial respiration measurements, fluorescence methods for the ROS level and mitochondrial membrane potential assessment, and cell viability measurements. In the human bronchial epithelial cell model (16HBE14σ), PM < 4 μm in diameter (SRM-PM4.0) was used. We observed that PM decreased TEER of HBE cell monolayers. The effect was partially abolished by quercetin, a mitoBKCa opener. Consequently, quercetin decreased the mitochondrial membrane potential and increased mitochondrial respiration. The reduction of PM-induced ROS level occurs both on cellular and mitochondrial level. Additionally, quercetin restores HBE cell viability after PM administration. The incubation of cells with PM substantially reduced the mitochondrial function. Isorhamnetin had no effect on TEER, the mitoBKCa activity, respiratory rate, or mitochondrial membrane potential. Obtained results indicate that PM has an adverse effect on HBE cells at the cellular and mitochondrial level. Quercetin is able to limit the deleterious effect of PM on barrier function of airway epithelial cells. We show that the effect in HBE cells involves mitoBKCa channel-activation. However, quercetin’s mechanism of action is not exclusively determined by modulation of the channel activity.
Collapse
|
33
|
Qi Y, Chen Y, Yan X, Liu W, Ma L, Liu Y, Ma Q, Liu S. Co-Exposure of Ambient Particulate Matter and Airborne Transmission Pathogens: The Impairment of the Upper Respiratory Systems. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:15892-15901. [PMID: 36240448 PMCID: PMC9670849 DOI: 10.1021/acs.est.2c03856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Recent evidence has pinpointed the positive relevance between air particulate matter (PM) pollution and epidemic spread. However, there are still significant knowledge gaps in understanding the transmission and infection of pathogens loaded on PMs, for example, the interactions between pathogens and pre-existing atmospheric PM and the health effects of co-exposure on the inhalation systems. Here, we unraveled the interactions between fine particulate matter (FPM) and Pseudomonas aeruginosa (P. aeruginosa) and evaluated the infection and detrimental effects of co-exposure on the upper respiratory systems in both in vitro and in vivo models. We uncovered the higher accessibility and invasive ability of pathogens to epithelial cells after loading on FPMs, compared with the single exposure. Furthermore, we designed a novel laboratory exposure model to simulate a real co-exposure scenario. Intriguingly, the co-exposure induced more serious functional damage and longer inflammatory reactions to the upper respiratory tract, including the nasal cavity and trachea. Collectively, our results provide a new point of view on the transmission and infection of pathogens loaded on FPMs and uncover the in vivo systematic impairments of the inhalation tract under co-exposure through a novel laboratory exposure model. Hence, this study sheds light on further investigations of the detrimental effects of air pollution and epidemic spread.
Collapse
Affiliation(s)
- Yu Qi
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yucai Chen
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu Yan
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Liu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Ma
- Aerosol
and Haze Laboratory, Advanced Innovation Center for Soft Matter Science
and Engineering, Beijing University of Chemical
Technology, Beijing 100029, China
| | - Yongchun Liu
- Aerosol
and Haze Laboratory, Advanced Innovation Center for Soft Matter Science
and Engineering, Beijing University of Chemical
Technology, Beijing 100029, China
| | - Qingxin Ma
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Sijin Liu
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research
Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
34
|
Beentjes D, Shears RK, French N, Neill DR, Kadioglu A. Mechanistic Insights into the Impact of Air Pollution on Pneumococcal Pathogenesis and Transmission. Am J Respir Crit Care Med 2022; 206:1070-1080. [PMID: 35649181 PMCID: PMC9704843 DOI: 10.1164/rccm.202112-2668tr] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/01/2022] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is the leading cause of pneumonia and bacterial meningitis. A number of recent studies indicate an association between the incidence of pneumococcal disease and exposure to air pollution. Although the epidemiological evidence is substantial, the underlying mechanisms by which the various components of air pollution (particulate matter and gases such as NO2 and SO2) can increase susceptibility to pneumococcal infection are less well understood. In this review, we summarize the various effects air pollution components have on pneumococcal pathogenesis and transmission; exposure to air pollution can enhance host susceptibility to pneumococcal colonization by impairing the mucociliary activity of the airway mucosa, reducing the function and production of key antimicrobial peptides, and upregulating an important pneumococcal adherence factor on respiratory epithelial cells. Air pollutant exposure can also impair the phagocytic killing ability of macrophages, permitting increased replication of S. pneumoniae. In addition, particulate matter has been shown to activate various extra- and intracellular receptors of airway epithelial cells, which may lead to increased proinflammatory cytokine production. This increases recruitment of innate immune cells, including macrophages and neutrophils. The inflammatory response that ensues may result in significant tissue damage, thereby increasing susceptibility to invasive disease, because it allows S. pneumoniae access to the underlying tissues and blood. This review provides an in-depth understanding of the interaction between air pollution and the pneumococcus, which has the potential to aid the development of novel treatments or alternative strategies to prevent disease, especially in areas with high concentrations of air pollution.
Collapse
Affiliation(s)
- Daan Beentjes
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Rebecca K Shears
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Neil French
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Daniel R Neill
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Aras Kadioglu
- Department of Clinical Immunology, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
35
|
Zhi Y, Chen X, Cao G, Chen F, Seo HS, Li F. The effects of air pollutants exposure on the transmission and severity of invasive infection caused by an opportunistic pathogen Streptococcus pyogenes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 310:119826. [PMID: 35932897 DOI: 10.1016/j.envpol.2022.119826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/30/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Currently, urbanization is associated with an increase in air pollutants that contribute to invasive pathogen infections by altering the host's innate immunity and antimicrobial resistance capability. Streptococcus pyogenes, also known as Group A Streptococcus (GAS), is a gram-positive opportunistic pathogen that causes a wide range of diseases, especially in children and immunosuppressed individuals. Diesel exhaust particle (DEP), a significant constituent of particulate matter (PM), are considered a prominent risk factor for respiratory illness and circulatory diseases worldwide. Several clinical and epidemiological studies have identified a close association between PM and the prevalence of viral and bacterial infections. This study investigated the role of DEP exposure in increasing pulmonary and blood bacterial counts and mortality during GAS M1 strain infection in mice. Thus, we characterized the upregulation of reactive oxygen species production and disruption of tight junctions in the A549 lung epithelial cell line due to DEP exposure, leading to the upregulation of GAS adhesion and invasion. Furthermore, DEP exposure altered the leukocyte components of infiltrated cells in bronchoalveolar lavage fluid, as determined by Diff-Quik staining. The results highlighted the DEP-related macrophage dysfunction, neutrophil impairment, and imbalance in pro-inflammatory cytokine production via the toll-like receptor 4/mitogen-activated protein kinase signaling axis. Notably, the tolerance of the GAS biofilms toward potent antibiotics and bacterial resistance against environmental stresses was also significantly enhanced by DEP. This study aimed to provide a better understanding of the physiological and molecular interactions between exposure to invasive air pollutants and susceptibility to invasive GAS infections.
Collapse
Affiliation(s)
- Yong Zhi
- Department of Obstetrics and Gynecology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xinyu Chen
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, 34113, Republic of Korea
| | - Guangxu Cao
- Department of Obstetrics and Gynecology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fengjia Chen
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, 56212, Jeollabuk-do, Republic of Korea
| | - Ho Seong Seo
- Research Division for Radiation Science, Korea Atomic Energy Research Institute, Jeongeup, 56212, Jeollabuk-do, Republic of Korea; Department of Radiation Biotechnology and Applied Radioisotope Science, University of Science and Technology, Daejeon, Republic of Korea
| | - Fang Li
- Department of Obstetrics and Gynecology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
36
|
Purves J, Hussey SJK, Corscadden L, Purser L, Hall A, Misra R, Selley L, Monks PS, Ketley JM, Andrew PW, Morrissey JA. Air pollution induces Staphylococcus aureus USA300 respiratory tract colonization mediated by specific bacterial genetic responses involving the global virulence gene regulators Agr and Sae. Environ Microbiol 2022; 24:4449-4465. [PMID: 35642645 PMCID: PMC9796851 DOI: 10.1111/1462-2920.16076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/06/2022] [Accepted: 05/16/2022] [Indexed: 01/07/2023]
Abstract
Exposure to particulate matter (PM), a major component of air pollution, is associated with exacerbation of chronic respiratory disease, and infectious diseases such as community-acquired pneumonia. Although PM can cause adverse health effects through direct damage to host cells, our previous study showed that PM can also impact bacterial behaviour by promoting in vivo colonization. In this study we describe the genetic mechanisms involved in the bacterial response to exposure to black carbon (BC), a constituent of PM found in most sources of air pollution. We show that Staphylococcus aureus strain USA300 LAC grown in BC prior to inoculation showed increased murine respiratory tract colonization and pulmonary invasion in vivo, as well as adhesion and invasion of human epithelial cells in vitro. Global transcriptional analysis showed that BC has a widespread effect on S. aureus transcriptional responses, altering the regulation of the major virulence gene regulators Sae and Agr and causing increased expression of genes encoding toxins, proteases and immune evasion factors. Together these data describe a previously unrecognized causative mechanism of air pollution-associated infection, in that exposure to BC can increase bacterial colonization and virulence factor expression by acting directly on the bacterium rather than via the host.
Collapse
Affiliation(s)
- Jo Purves
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| | - Shane J. K. Hussey
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| | - Louise Corscadden
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| | - Lillie Purser
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| | - Andie Hall
- Molecular Biology, Core Research LaboratoriesNatural History MuseumCromwell Road, LondonSW7 5BDUK
| | - Raju Misra
- Molecular Biology, Core Research LaboratoriesNatural History MuseumCromwell Road, LondonSW7 5BDUK
| | - Liza Selley
- MRC Toxicology UnitUniversity of CambridgeCambridgeCB2 1QRUK
| | - Paul S. Monks
- Department of ChemistryUniversity of LeicesterUniversity RoadLeicesterLE1 7RHUK
| | - Julian M. Ketley
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| | - Peter W. Andrew
- Department of Respiratory SciencesUniversity of LeicesterUniversity Road, LeicesterLE1 9HNUK
| | - Julie A. Morrissey
- Department of GeneticsUniversity of Leicester, University RoadLeicesterLE1 7RHUK
| |
Collapse
|
37
|
Repeated exposure of bronchial epithelial cells to particular matter increases allergen-induced cytokine release and permeability. Cytokine 2022; 154:155878. [DOI: 10.1016/j.cyto.2022.155878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/03/2022] [Indexed: 11/22/2022]
|
38
|
Celebi Sozener Z, Ozdel Ozturk B, Cerci P, Turk M, Gorgulu Akin B, Akdis M, Altiner S, Ozbey U, Ogulur I, Mitamura Y, Yilmaz I, Nadeau K, Ozdemir C, Mungan D, Akdis CA. Epithelial barrier hypothesis: Effect of the external exposome on the microbiome and epithelial barriers in allergic disease. Allergy 2022; 77:1418-1449. [PMID: 35108405 PMCID: PMC9306534 DOI: 10.1111/all.15240] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/11/2022]
Abstract
Environmental exposure plays a major role in the development of allergic diseases. The exposome can be classified into internal (e.g., aging, hormones, and metabolic processes), specific external (e.g., chemical pollutants or lifestyle factors), and general external (e.g., broader socioeconomic and psychological contexts) domains, all of which are interrelated. All the factors we are exposed to, from the moment of conception to death, are part of the external exposome. Several hundreds of thousands of new chemicals have been introduced in modern life without our having a full understanding of their toxic health effects and ways to mitigate these effects. Climate change, air pollution, microplastics, tobacco smoke, changes and loss of biodiversity, alterations in dietary habits, and the microbiome due to modernization, urbanization, and globalization constitute our surrounding environment and external exposome. Some of these factors disrupt the epithelial barriers of the skin and mucosal surfaces, and these disruptions have been linked in the last few decades to the increasing prevalence and severity of allergic and inflammatory diseases such as atopic dermatitis, food allergy, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, and asthma. The epithelial barrier hypothesis provides a mechanistic explanation of how these factors can explain the rapid increase in allergic and autoimmune diseases. In this review, we discuss factors affecting the planet's health in the context of the 'epithelial barrier hypothesis,' including climate change, pollution, changes and loss of biodiversity, and emphasize the changes in the external exposome in the last few decades and their effects on allergic diseases. In addition, the roles of increased dietary fatty acid consumption and environmental substances (detergents, airborne pollen, ozone, microplastics, nanoparticles, and tobacco) affecting epithelial barriers are discussed. Considering the emerging data from recent studies, we suggest stringent governmental regulations, global policy adjustments, patient education, and the establishment of individualized control measures to mitigate environmental threats and decrease allergic disease.
Collapse
Affiliation(s)
| | - Betul Ozdel Ozturk
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic DiseasesEskisehir City HospitalEskisehirTurkey
| | - Murat Turk
- Clinic of Immunology and Allergic DiseasesKayseri City HospitalKayseriTurkey
| | - Begum Gorgulu Akin
- Clinic of Immunology and Allergic DiseasesAnkara City HospitalAnkaraTurkey
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Seda Altiner
- Clinic of Internal Medicine Division of Immunology and Allergic DiseasesKahramanmaras Necip Fazil City HospitalKahramanmarasTurkey
| | - Umus Ozbey
- Department of Nutrition and DietAnkara UniversityAnkaraTurkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Insu Yilmaz
- Department of Chest DiseasesDivision of Immunology and Allergic DiseasesErciyes UniversityKayseriTurkey
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University School of MedicineDivision of Pulmonary and Critical Care MedicineDepartment of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Cevdet Ozdemir
- Institute of Child HealthDepartment of Pediatric Basic SciencesIstanbul UniversityIstanbulTurkey
- Istanbul Faculty of MedicineDepartment of PediatricsDivision of Pediatric Allergy and ImmunologyIstanbul UniversityIstanbulTurkey
| | - Dilsad Mungan
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| |
Collapse
|
39
|
Shrestha D, Massey N, Bhat SM, Jelesijević T, Sahin O, Zhang Q, Bailey KL, Poole JA, Charavaryamath C. Nrf2 Activation Protects Against Organic Dust and Hydrogen Sulfide Exposure Induced Epithelial Barrier Loss and K. pneumoniae Invasion. Front Cell Infect Microbiol 2022; 12:848773. [PMID: 35521223 PMCID: PMC9062039 DOI: 10.3389/fcimb.2022.848773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/21/2022] [Indexed: 11/23/2022] Open
Abstract
Agriculture workers report various respiratory symptoms owing to occupational exposure to organic dust (OD) and various gases. Previously, we demonstrated that pre-exposure to hydrogen sulfide (H2S) alters the host response to OD and induces oxidative stress. Nrf2 is a master-regulator of host antioxidant response and exposures to toxicants is known to reduce Nrf2 activity. The OD exposure-induced lung inflammation is known to increase susceptibility to a secondary microbial infection. We tested the hypothesis that repeated exposure to OD or H2S leads to loss of Nrf2, loss of epithelial cell integrity and that activation of Nrf2 rescues this epithelial barrier dysfunction. Primary normal human bronchial epithelial (NHBE) cells or mouse precision cut-lung slices (PCLS) were treated with media, swine confinement facility organic dust extract (ODE) or H2S or ODE+H2S for one or five days. Cells were also pretreated with vehicle control (DMSO) or RTA-408, a Nrf2 activator. Acute exposure to H2S and ODE+H2S altered the cell morphology, decreased the viability as per the MTT assay, and reduced the Nrf2 expression as well as increased the keap1 levels in NHBE cells. Repeated exposure to ODE or H2S or ODE+H2S induced oxidative stress and cytokine production, decreased tight junction protein occludin and cytoskeletal protein ezrin expression, disrupted epithelial integrity and resulted in increased Klebsiella pneumoniae invasion. RTA-408 (pharmacological activator of Nrf2) activated Nrf2 by decreasing keap1 levels and reduced ODE+H2S-induced changes including reversing loss of barrier integrity, inflammatory cytokine production and microbial invasion in PCLS but not in NHBE cell model. We conclude that Nrf2 activation has a partial protective function against ODE and H2S.
Collapse
Affiliation(s)
- Denusha Shrestha
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Nyzil Massey
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Sanjana Mahadev Bhat
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Immunobiology Interdepartmental Graduate Program, Iowa State University, Ames, IA, United States
| | - Tomislav Jelesijević
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Orhan Sahin
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| | - Qijing Zhang
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Kristina L. Bailey
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Jill A. Poole
- Department of Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chandrashekhar Charavaryamath
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- *Correspondence: Chandrashekhar Charavaryamath,
| |
Collapse
|
40
|
LPS Response Is Impaired by Urban Fine Particulate Matter. Int J Mol Sci 2022; 23:ijms23073913. [PMID: 35409273 PMCID: PMC8998903 DOI: 10.3390/ijms23073913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Fine particulate matter (PM2.5) is a complex mixture of components with diverse chemical and physical characteristics associated with increased respiratory and cardiovascular diseases mortality. Our study aimed to investigate the effects of exposure to concentrated PM2.5 on LPS-induced lung injury onset. BALB/c male mice were exposed to either filtered air or ambient fine PM2.5 in an ambient particle concentrator for 5 weeks. Then, an acute lung injury was induced with nebulized LPS. The animals were euthanized 24 h after the nebulization to either LPS or saline. Inflammatory cells and cytokines (IL-1β, IL-4, IL-5, IL-6, IL-10, IL-17, TNF) were assessed in the blood, bronchoalveolar lavage fluid (BALF), and lung tissue. In addition, lung morphology was assessed by stereological methods. Our results showed that the PM+LPS group showed histological evidence of injury, leukocytosis with increased neutrophils and macrophages, and a mixed inflammatory response profile, with increased KC, IL-6, IL-1β, IL-4, and IL-17. Our analysis shows that there is an interaction between the LPS nebulization and PM2.5 exposure, differently modulating the inflammatory response, with a distinct response pattern as compared to LPS or PM2.5 exposure alone. Further studies are required to explain the mechanism of immune modulation caused by PM2.5 exposure.
Collapse
|
41
|
Aghapour M, Ubags ND, Bruder D, Hiemstra PS, Sidhaye V, Rezaee F, Heijink IH. Role of air pollutants in airway epithelial barrier dysfunction in asthma and COPD. Eur Respir Rev 2022; 31:31/163/210112. [PMID: 35321933 PMCID: PMC9128841 DOI: 10.1183/16000617.0112-2021] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic exposure to environmental pollutants is a major contributor to the development and progression of obstructive airway diseases, including asthma and COPD. Understanding the mechanisms underlying the development of obstructive lung diseases upon exposure to inhaled pollutants will lead to novel insights into the pathogenesis, prevention and treatment of these diseases. The respiratory epithelial lining forms a robust physicochemical barrier protecting the body from inhaled toxic particles and pathogens. Inhalation of airborne particles and gases may impair airway epithelial barrier function and subsequently lead to exaggerated inflammatory responses and airway remodelling, which are key features of asthma and COPD. In addition, air pollutant-induced airway epithelial barrier dysfunction may increase susceptibility to respiratory infections, thereby increasing the risk of exacerbations and thus triggering further inflammation. In this review, we discuss the molecular and immunological mechanisms involved in physical barrier disruption induced by major airborne pollutants and outline their implications in the pathogenesis of asthma and COPD. We further discuss the link between these pollutants and changes in the lung microbiome as a potential factor for aggravating airway diseases. Understanding these mechanisms may lead to identification of novel targets for therapeutic intervention to restore airway epithelial integrity in asthma and COPD. Exposure to air pollution induces airway epithelial barrier dysfunction through several mechanisms including increased oxidative stress, exaggerated cytokine responses and impaired host defence, which contributes to development of asthma and COPD. https://bit.ly/3DHL1CA
Collapse
Affiliation(s)
- Mahyar Aghapour
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Epalinges, Switzerland
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Venkataramana Sidhaye
- Pulmonary and Critical Care Medicine, Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Fariba Rezaee
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children's, Cleveland, OH, USA.,Dept of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Depts of Pathology and Medical Biology and Pulmonology, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| |
Collapse
|
42
|
Baron YM. Are there medium to short-term multifaceted effects of the airborne pollutant PM 2.5 determining the emergence of SARS-CoV-2 variants? Med Hypotheses 2021; 158:110718. [PMID: 34758423 PMCID: PMC8526108 DOI: 10.1016/j.mehy.2021.110718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/17/2021] [Indexed: 01/22/2023]
Abstract
The COVID-19 pandemic has been characterised by successiveoutbreaks effecting large swathes of the world's populations. These waves of infection have been mainly driven by a number of more transmissiblevariants which appear to evade the populations' immunity gained from previous outbreaks. There appears to be a link between COVID-19 and a ubiquitous airborne pollutant calledparticulate matter, PM2.5. Particulate matter through a number of mechanisms, including its anthropogenic effect, appears to be associated with the incidence and the mortality related to the COVID-19 pandemic. This paper poses a number of hypotheses on the short to medium-term mechanisms whereby PM2.5 may be party to the natural selection of SARS-CoV-2 virus, with the consequent emergence of variants.
Collapse
|
43
|
Yuksel H, Ocalan M, Yilmaz O. E-Cadherin: An Important Functional Molecule at Respiratory Barrier Between Defence and Dysfunction. Front Physiol 2021; 12:720227. [PMID: 34671272 PMCID: PMC8521047 DOI: 10.3389/fphys.2021.720227] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
While breathing, many microorganisms, harmful environmental particles, allergens, and environmental pollutants enter the human airways. The human respiratory tract is lined with epithelial cells that act as a functional barrier to these harmful factors and provide homeostasis between external and internal environment. Intercellular epithelial junctional proteins play a role in the formation of the barrier. E-cadherin is a calcium-dependent adhesion molecule and one of the most important molecules involved in intercellular epithelial barier formation. E-cadherin is not only physical barrier element but also regulates cell proliferation, differentiation and the immune response to environmental noxious agents through various transcription factors. In this study, we aimed to review the role of E-cadherin in the formation of airway epithelial barier, its status as a result of exposure to various environmental triggers, and respiratory diseases associated with its dysfunction. Moreover, the situations in which its abnormal activation can be noxious would be discussed.
Collapse
Affiliation(s)
- Hasan Yuksel
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Merve Ocalan
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| | - Ozge Yilmaz
- Department of Pediatric Allergy and Pulmonology, Faculty of Medicine, Celal Bayar University, Manisa, Turkey
| |
Collapse
|
44
|
Smyth T, Georas SN. Effects of ozone and particulate matter on airway epithelial barrier structure and function: a review of in vitro and in vivo studies. Inhal Toxicol 2021; 33:177-192. [PMID: 34346824 DOI: 10.1080/08958378.2021.1956021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The airway epithelium represents a crucial line of defense against the spread of inhaled pathogens. As the epithelium is the first part of the body to be exposed to the inhaled environment, it must act as both a barrier to and sentinel against any inhaled agents. Despite its vital role in limiting the spread of inhaled pathogens, the airway epithelium is also regularly exposed to air pollutants which disrupt its normal function. Here we review the current understanding of the structure and composition of the airway epithelial barrier, as well as the impact of inhaled pollutants, including the reactive gas ozone and particulate matter, on epithelial function. We discuss the current in vitro, rodent model, and human exposure findings surrounding the impact of various inhaled pollutants on epithelial barrier function, mucus production, and mucociliary clearance. Detailed information on how inhaled pollutants impact epithelial structure and function will further our understanding of the adverse health effects of air pollution exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steve N Georas
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
45
|
Rodriguez-Coira J, Villaseñor A, Izquierdo E, Huang M, Barker-Tejeda TC, Radzikowska U, Sokolowska M, Barber D. The Importance of Metabolism for Immune Homeostasis in Allergic Diseases. Front Immunol 2021; 12:692004. [PMID: 34394086 PMCID: PMC8355700 DOI: 10.3389/fimmu.2021.692004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/05/2021] [Indexed: 12/27/2022] Open
Abstract
There is increasing evidence that the metabolic status of T cells and macrophages is associated with severe phenotypes of chronic inflammation, including allergic inflammation. Metabolic changes in immune cells have a crucial role in their inflammatory or regulatory responses. This notion is reinforced by metabolic diseases influencing global energy metabolism, such as diabetes or obesity, which are known risk factors of severity in inflammatory conditions, due to the metabolic-associated inflammation present in these patients. Since several metabolic pathways are closely tied to T cell and macrophage differentiation, a better understanding of metabolic alterations in immune disorders could help to restore and modulate immune cell functions. This link between energy metabolism and inflammation can be studied employing animal, human or cellular models. Analytical approaches rank from classic immunological studies to integrated analysis of metabolomics, transcriptomics, and proteomics. This review summarizes the main metabolic pathways of the cells involved in the allergic reaction with a focus on T cells and macrophages and describes different models and platforms of analysis used to study the immune system and its relationship with metabolism.
Collapse
Affiliation(s)
- Juan Rodriguez-Coira
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Alma Villaseñor
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Elena Izquierdo
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Tomás Clive Barker-Tejeda
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain.,Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos Wolfgang, Switzerland
| | - Domingo Barber
- Departamento de Ciencias Medicas Basicas, Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla Del Monte, Madrid, Spain
| |
Collapse
|
46
|
Paplinska-Goryca M, Misiukiewicz-Stepien P, Proboszcz M, Nejman-Gryz P, Gorska K, Zajusz-Zubek E, Krenke R. Interactions of nasal epithelium with macrophages and dendritic cells variously alter urban PM-induced inflammation in healthy, asthma and COPD. Sci Rep 2021; 11:13259. [PMID: 34168212 PMCID: PMC8225888 DOI: 10.1038/s41598-021-92626-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/08/2021] [Indexed: 02/05/2023] Open
Abstract
Urban particulate matter (UPM) is an important trigger of airway inflammation. The cross-talk between the external and internal matrix in the respiratory tract occurs due to the transepithelial network of macrophages/dendritic cells. This study characterized the immune processes induced by the epithelium after UPM exposure in special regard to interactions with monocyte-derived dendritic cells (moDCs) and monocyte-derived macrophages (moMφs) in obstructive lung diseases. A triple-cell co-culture model (8 controls, 10 asthma, and 8 patients with COPD) utilized nasal epithelial cells, along with moMφs, and moDCs was exposed to UPM for 24 h. The inflammatory response of nasal epithelial cells to UPM stimulation is affected differently by cell-cell interactions in healthy people, asthma or COPD patients of which the interactions with DCs had the strongest impact on the inflammatory reaction of epithelial cells after UPM exposure. The epithelial remodeling and DCs dysfunction might accelerate the inflammation after air pollution exposure in asthma and COPD.
Collapse
Affiliation(s)
- Magdalena Paplinska-Goryca
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland.
| | - Paulina Misiukiewicz-Stepien
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Proboszcz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Patrycja Nejman-Gryz
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Katarzyna Gorska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| | - Elwira Zajusz-Zubek
- Faculty of Energy and Environmental Engineering, Department of Air Protection, Silesian University of Technology, Gliwice, Poland
| | - Rafal Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Banacha 1a, 02-097, Warsaw, Poland
| |
Collapse
|
47
|
Carlier FM, de Fays C, Pilette C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front Physiol 2021; 12:691227. [PMID: 34248677 PMCID: PMC8264588 DOI: 10.3389/fphys.2021.691227] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells, which provide a complex and adaptive module that ensures first-line defense against external toxics, irritants, antigens, and pathogens. The underlying mechanisms of host protection encompass multiple physical, chemical, and immune pathways. In the lung, inhaled agents continually challenge the airway epithelial barrier, which is altered in chronic diseases such as chronic obstructive pulmonary disease, asthma, cystic fibrosis, or pulmonary fibrosis. In this review, we describe the epithelial barrier abnormalities that are observed in such disorders and summarize current knowledge on the mechanisms driving impaired barrier function, which could represent targets of future therapeutic approaches.
Collapse
Affiliation(s)
- François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology and Lung Transplant, Centre Hospitalier Universitaire UCL Namur, Yvoir, Belgium
| | - Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| |
Collapse
|
48
|
Hamidou Soumana I, Carlsten C. Air pollution and the respiratory microbiome. J Allergy Clin Immunol 2021; 148:67-69. [PMID: 34048853 DOI: 10.1016/j.jaci.2021.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Illiassou Hamidou Soumana
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher Carlsten
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
49
|
Zhang S, Chen X, Wang J, Dai C, Gou Y, Wang H. Particulate air pollution and respiratory Haemophilus influenzae infection in Mianyang, southwest China. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:10.1007/s11356-021-13103-5. [PMID: 33638077 DOI: 10.1007/s11356-021-13103-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/18/2021] [Indexed: 02/05/2023]
Abstract
Particulate air pollution is correlated with many respiratory diseases. However, few studies have focused on the relationship between air particulate exposure and respiratory Heamophilus influenzae infection. Therefore, we detected respiratory Heamophilus influenzae infection by bacterial culture of sputum of patients, and we collected particulate air pollution data (including PM2.5 and PM10) from a national real-time urban air quality platform to analyze the relationship between particulate air pollution and respiratory Heamophilus influenzae infection. The mean concentrations of PM2.5 and PM10 were 37.58 μg/m3 and 58.44 μg/m3, respectively, showing particulate air pollution remains a severe issue in Mianyang. A total of 828 strains of Heamophilus influenzae were detected in sputum by bacterial culture. Multiple correspondence analysis suggested the heaviest particulate air pollution and the highest Heamophilus influenzae infection rates were all in winter, while the lowest particulate air pollution and the lowest Heamophilus influenzae infection rates were all in summer. In a single-pollutant model, each elevation of 10 μg/m3 of PM2.5, PM10, and PM2.5/10 (combined exposure level) increased the risk of respiratory Heamophilus influenzae infection by 34%, 23%, and 29%, respectively. Additionally, in the multiple-pollutant model, only PM2.5 was significantly associated with respiratory Heamophilus influenzae infection (B, 0.46; 95% confidence interval, 0.05-0.87), showing PM2.5 is an independent risk factor for respiratory Heamophilus influenzae infection. In summary, this study highlights air particulate exposure could increase the risk of respiratory Heamophilus influenzae infection, implying that stronger measures need to be taken to protect against respiratory infection induced by particulate air pollution.
Collapse
Affiliation(s)
- Shaocheng Zhang
- Department of Clinical Laboratory Medicine, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Xi Chen
- Department of Clinical Laboratory Medicine, Mianyang Central Hospital, 12 Changjia Lane, Jingzhong St, Mianyang, 621000, Sichuan, China.
| | - Jing Wang
- Department of Clinical Laboratory Medicine, Mianyang Central Hospital, 12 Changjia Lane, Jingzhong St, Mianyang, 621000, Sichuan, China
| | - Chunmei Dai
- Department of Clinical Laboratory Medicine, Mianyang Central Hospital, 12 Changjia Lane, Jingzhong St, Mianyang, 621000, Sichuan, China
| | - Yeran Gou
- Department of Respiratory and Critical Care Medicine, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Huanhuan Wang
- Department of Cell Biology and Genetics, Shantou University Medical College, 22 Xinling Rd, Shantou, 515041, Guangdong, China.
| |
Collapse
|
50
|
Celebi Sözener Z, Cevhertas L, Nadeau K, Akdis M, Akdis CA. Environmental factors in epithelial barrier dysfunction. J Allergy Clin Immunol 2021; 145:1517-1528. [PMID: 32507229 DOI: 10.1016/j.jaci.2020.04.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/11/2022]
Abstract
The main interfaces controlling and attempting to homeostatically balance communications between the host and the environment are the epithelial barriers of the skin, gastrointestinal system, and airways. The epithelial barrier constitutes the first line of physical, chemical, and immunologic defenses and provides a protective wall against environmental factors. Following the industrial revolution in the 19th century, urbanization and socioeconomic development have led to an increase in energy consumption, and waste discharge, leading to increased exposure to air pollution and chemical hazards. Particularly after the 1960s, biological and chemical insults from the surrounding environment-the exposome-have been disrupting the physical integrity of the barrier by degrading the intercellular barrier proteins at tight and adherens junctions, triggering epithelial alarmin cytokine responses such as IL-25, IL-33, and thymic stromal lymphopoietin, and increasing the epithelial barrier permeability. A typical type 2 immune response develops in affected organs in asthma, rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, food allergy, and atopic dermatitis. The aim of this article was to discuss the effects of environmental factors such as protease enzymes of allergens, detergents, tobacco, ozone, particulate matter, diesel exhaust, nanoparticles, and microplastic on the integrity of the epithelial barriers in the context of epithelial barrier hypothesis.
Collapse
Affiliation(s)
- Zeynep Celebi Sözener
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Department of Chest Diseases, Division of Allergy and Immunology, Ankara University School of Medicine, Ankara, Turkey
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Kari Nadeau
- the Naddisy Foundation, Sean Parker Asthma and Allergy Center, Stanford University, Stanford, Calif
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Herman-Burchard Strasse 9, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| |
Collapse
|