1
|
Perez L, Ambroise J, Bearzatto B, Froidure A, Pilette C, Yakoub Y, Palmai-Pallag M, Bouzin C, Ryelandt L, Pavan C, Huaux F, Lison D. Unique transcriptomic responses of rat and human alveolar macrophages in an in vitro model of overload with TiO 2 and carbon black. Part Fibre Toxicol 2025; 22:8. [PMID: 40281615 PMCID: PMC12023592 DOI: 10.1186/s12989-025-00624-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Chronic inhalation of titanium dioxide or carbon black can lead, at high exposure, to lung overload, and can induce chronic inflammation and lung cancer in rats. Whether this rat adverse response is predictive for humans has been questioned for more than 40 years. Currently, these particles are conservatively considered as possible human carcinogens. OBJECTIVE To clarify the mechanisms of the adverse rat response to lung overload and its human relevance. METHODS Primary rat and human alveolar macrophages were exposed in vitro to control, non-overload or overload doses of titanium dioxide (P25) or carbon black (Printex 90) particles, and their activation profile was examined by untargeted transcriptomics. RESULTS Rat macrophages were largely the most responsive to particle overload. In particular, eighteen genes were identified as robust markers of P25 and Printex 90 overload in rat cells. The known functions of these genes can be related to the potential mechanisms of the adverse outcomes recorded in rats in vivo. Most of these 18 genes were similarly modulated in human macrophages, but with a markedly lower magnitude. In addition, a 16 gene signature was observed upon overload in human macrophages, but not in rat macrophages. CONCLUSIONS These findings provide insights into the mechanisms of lung overload and inflammation in rats, and highlight similarities and differences in transcriptomic responses of rat and human alveolar macrophages.
Collapse
Affiliation(s)
- Laeticia Perez
- Louvain Centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium.
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées , Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Bertrand Bearzatto
- Centre de Technologies Moléculaires Appliquées , Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Antoine Froidure
- Pôle Pneumologie, ORL (Airways) et dermatologie (Skin), Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Charles Pilette
- Pôle Pneumologie, ORL (Airways) et dermatologie (Skin), Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Mihaly Palmai-Pallag
- Secteur des Sciences de la santé , Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform (2IP; RRID:SCR_023378), Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Laurence Ryelandt
- Institute of Mechanics, Materials and Civil Engineering, Université Catholique de Louvain, Louvain-la- Neuve, Belgium
| | - Cristina Pavan
- Department of Chemistry, University of Turin, Turin, Italy
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
2
|
Nagar E, Singh N, Saini N, Arora N. Glutathione attenuates diesel exhaust-induced lung epithelial injury via NF-κB/Nrf2/GPX4-mediated ferroptosis. Toxicology 2025; 515:154154. [PMID: 40239913 DOI: 10.1016/j.tox.2025.154154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/06/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Diesel exhaust (DE) emissions pose a significant threat to public health. This study linked DE-mediated reactive oxygen species (ROS) and ferroptosis with lung epithelial disruption, also the protective potential of exogenous glutathione (GSH) administration was investigated. C57BL/6 mice were divided into three groups: filtered air (control), DE exposed, and DE+GSH (administered intranasally on alternate days). Airway hyperresponsiveness (AHR), lung tissues, and bronchoalveolar lavage fluid (BALF) were used for analysis. DE exposure significantly impaired lung function parameters as shown by AHR. Elevated ROS depleted the GSH/GSSG ratio and suppressed Nrf2 activity, disrupting antioxidant defense mechanisms, which were restored by GSH administration. DE-induced ROS acted as a key driver of ferroptosis, characterized by suppressed SLC7411 expression thereby decreased GSH synthesis and GPX-4 activity, inducing lipid peroxidation. Ferroptosis was significantly mitigated by increased GSH pool, which restored GPX-4 levels and reduced lipid peroxidation. Concurrently, DE induced ROS promoted DNA damage and apoptosis in lung epithelial cells wherein GSH treatment preserved cell survival in DE exposed mice. The heightened DE-associated ROS further amplified inflammation, as shown by increased cytokines (TNF-α, IL-6, TSLP, IL-33) and P-NF-κB activation. Activated inflammatory cascade disrupted tight junction proteins (claudins, occludin), resulted in weakened epithelial barrier and increased permeability. Lung barrier disruption was evidenced by transmission electron microscopy and immunohistochemistry, corroborated with elevated albumin levels. GSH effectively restored tight junction integrity and preserved barrier function in DE+GSH mice lungs. In conclusion, DE-induced oxidative stress and ferroptosis-triggered inflammation compromised epithelial barrier promoting lung injury. Exogenous GSH administration showed potential in restoring DE-associated lung damage.
Collapse
Affiliation(s)
- Ekta Nagar
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Naresh Singh
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Neeru Saini
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Naveen Arora
- CSIR-Institute of Genomics and Integrative Biology, Delhi 110007, India.
| |
Collapse
|
3
|
Park JW, Kang M, Kim G, Hyun SY, Shin J, Kim SY, Lee JH, Choi WS, Lee JH, Lee K, Kim SH, Cho WS, Kim HS. The impact of atmospheric ultrafine particulate matter on IgE-mediated type 1 hypersensitivity reaction. JOURNAL OF HAZARDOUS MATERIALS 2025; 484:136705. [PMID: 39637818 DOI: 10.1016/j.jhazmat.2024.136705] [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: 05/23/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
The effect of atmospheric ultrafine particulate matter (UPM) on respiratory allergic diseases has been investigated for decades; however, the precise molecular mechanisms underlying these effects remain poorly understood. In this study, we used a simulated UPM (sUPM) generated via the spark discharge method to refine black carbon, a core particle that closely mimics real-world UPM, including the size (i.e., size of agglomerates: 165 nm) and organic carbon/elemental carbon ratio (i.e., 2.62). When 25 μg/mouse of dispersed sUPM was instilled into the lungs of mice, it promoted the infiltration and degranulation response of pulmonary mast cells, and exposure to sUPM in an immunoglobulin E (IgE)-mediated passive anaphylaxis model intensified the degranulation response of peripheral mast cells. These effects of sUPM were demonstrated to amplify the downstream signaling mechanism of the high-affinity IgE receptor (FcεRI) mediated by IgE when tested using rat basophil leukemia (RBL)-2H3 and mouse bone marrow-derived mast cells (BMMCs) collected from the bone marrow of BALB/c mice. These results indicate that airborne UPM can exacerbate type 1 hypersensitivity reactions by enhancing the IgE-mediated signaling pathways within mast cells. Furthermore, this study provided mechanistic evidence on exacerbated allergic pulmonary diseases induced by UPM inhalation.
Collapse
Affiliation(s)
- Jeong Won Park
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Minseong Kang
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Gyuri Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seung Yeun Hyun
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Juhyun Shin
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Seon Young Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jun Ho Lee
- Department of Korean Medicine, College of Korean Medicine, Woosuk University, Jeonju 54986, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology, College of Medicine, Konkuk University, Chungju 27478, Republic of Korea
| | - Jong-Ho Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do 56212, Republic of Korea
| | - Seok-Ho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Wan-Seob Cho
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea.
| | - Hyuk Soon Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea; Department of Biomedical Sciences, College of Natural Science, Dong-A University, Busan 49315, Republic of Korea.
| |
Collapse
|
4
|
Smyth TR, Brocke S, Kim YH, Christianson C, Kovalcik KD, Pancras JP, Hays MD, Wu W, An Z, Jaspers I. Human Monocyte-Derived Macrophages Demonstrate Distinct Responses to Ambient Particulate Matter in a Polarization State- and Particle Seasonality-Specific Manner. Chem Res Toxicol 2025; 38:73-90. [PMID: 39704336 DOI: 10.1021/acs.chemrestox.4c00291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Macrophages are professional phagocytic immune cells that, following activation, polarize on a spectrum between the proinflammatory M1 and the proresolution M2 states. Macrophages have further been demonstrated to retain plasticity, allowing for the reprogramming of their polarization states following exposure to new stimuli. Particulate matter (PM) has been repeatedly shown to modify macrophage function and polarization while also inducing worsening respiratory infection morbidity and mortality. However, limited work has considered the impact of the initial macrophage polarization state on subsequent responses to PM exposure. PM composition can demonstrate seasonality-specific compositional changes based on differences in seasonal weather patterns and energy needs, introducing the need to consider the seasonality-specific effects of airborne PM when investigating its impact on human health. This study sought to determine the impact of airborne PM collected during different seasons of the year in Xinxiang, China, on macrophage function in a polarization state-dependent manner. Macrophages were differentiated using the macrophage colony-stimulating factor (M-CSF) on CD14+CD16- monocytes isolated from the blood of healthy human volunteers. The resulting macrophages were polarized into indicated states using well-characterized polarization methods and assessed for phagocytic function, bioenergetic properties, and secretory profile following exposure to PM collected during a single day during each season of the year. Macrophages demonstrated clear polarization state-dependent phagocytic, bioenergetic, and secretory properties at the baseline and following PM exposure. Specific PM seasonality had a minimal impact on phagocytic function and a minor effect on bioenergetic properties but had clear impacts on the secretory profile as demonstrated by the enriched secretion of well-characterized mediator clusters by particle season. Together, these data suggest that both particle seasonality and macrophage polarization state must be considered when investigating the impact of PM on macrophage function. These factors may contribute to the negative outcomes linked to PM exposure during respiratory infections.
Collapse
Affiliation(s)
- Timothy R Smyth
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
| | - Stephanie Brocke
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
| | - Yong Ho Kim
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States of America
| | - Cara Christianson
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States of America
| | - Kasey D Kovalcik
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States of America
| | - Joseph Patrick Pancras
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States of America
| | - Michael D Hays
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27709, United States of America
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang 453004, China
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang 453004, China
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States of America
| |
Collapse
|
5
|
Pardo M, Li C, Jabali A, Rudich Y. Cellular and metabolic impacts of repeated sub-acute exposures to biomass-burning extracts in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117491. [PMID: 39657377 DOI: 10.1016/j.ecoenv.2024.117491] [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: 09/16/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The increasing exposure to biomass-burning emissions underscores the need to understand their toxicological impacts on human health. In this study, we developed a laboratory model to evaluate the effects of single and repeated sub-acute exposures to water-soluble wood tar (WT) extracts, a product of biomass burning, on human lung, liver, and immune cells. Using representative cell lines for different tissues, we examined the cytotoxic effects under conditions mimicking sub-acute environmental exposure levels relevant to humans. Our findings indicate that repeated sub-acute exposures to water-soluble WT extracts significantly enhance the inflammatory response, evidenced by increased IL6, IL8, and TNFa cytokine levels, compared to a single exposure. Additionally, oxidative stress responses were more pronounced with increased lipid peroxidation and HMOX1, GCLC and CYP1A1 gene expression following repeated exposures. Metabolomics analyses of polar and lipid metabolites revealed changes related to energy production and consumption that emerge even after a single exposure at sub-acute levels and vary across different cell types representing the different tissues. Impaired cellular respiration, measured by oxygen consumption rate, corroborates the observed changes. These results provide important insights into the cellular mechanisms driving the response to biomass-burning exposure and highlight the potential health risks associated with sub-acute exposure to environmental pollutants.
Collapse
Affiliation(s)
- Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel.
| | - Chunlin Li
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
| | - Amani Jabali
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot 7610001, Israel
| |
Collapse
|
6
|
Ahmad S, Nasser W, Ahmad A. Epigenetic mechanisms of alveolar macrophage activation in chemical-induced acute lung injury. Front Immunol 2024; 15:1488913. [PMID: 39582870 PMCID: PMC11581858 DOI: 10.3389/fimmu.2024.1488913] [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: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 11/26/2024] Open
Abstract
Airways, alveoli and the pulmonary tissues are the most vulnerable to the external environment including occasional deliberate or accidental exposure to highly toxic chemical gases. However, there are many effective protective mechanisms that maintain the integrity of the pulmonary tissues and preserve lung function. Alveolar macrophages form the first line of defense against any pathogen or chemical/reactant that crosses the airway mucociliary barrier and reaches the alveolar region. Resident alveolar macrophages are activated or circulating monocytes infiltrate the airspace to contribute towards inflammatory or reparative responses. Studies on response of alveolar macrophages to noxious stimuli are rapidly emerging and alveolar macrophage are also being sought as therapeutic target. Here such studies have been reviewed and put together for a better understanding of the role pulmonary macrophages in general and alveolar macrophage in particular play in the pathogenesis of disease caused by chemical induced acute lung injury.
Collapse
Affiliation(s)
- Shama Ahmad
- Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | | |
Collapse
|
7
|
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
|
8
|
Preedalikit W, Chittasupho C, Leelapornpisid P, Duangnin N, Kiattisin K. Potential of Coffee Cherry Pulp Extract against Polycyclic Aromatic Hydrocarbons in Air Pollution Induced Inflammation and Oxidative Stress for Topical Applications. Int J Mol Sci 2024; 25:9416. [PMID: 39273362 PMCID: PMC11395326 DOI: 10.3390/ijms25179416] [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: 07/18/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Airborne particulate matter (PM) contains polycyclic aromatic hydrocarbons (PAHs) as primary toxic components, causing oxidative damage and being associated with various inflammatory skin pathologies such as premature aging, atopic dermatitis, and psoriasis. Coffee cherry pulp (CCS) extract, rich in chlorogenic acid, caffeine, and theophylline, has demonstrated strong antioxidant properties. However, its specific anti-inflammatory effects and ability to protect macrophages against PAH-induced inflammation remain unexplored. Thus, this study aimed to evaluate the anti-inflammatory properties of CCS extract on RAW 264.7 macrophage cells exposed to atmospheric PAHs, compared to chlorogenic acid (CGA), caffeine (CAF), and theophylline (THP) standards. The CCS extract was assessed for its impact on the production of nitric oxide (NO) and expression of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Results showed that CCS extract exhibited significant antioxidant activities and effectively inhibited protease and lipoxygenase (LOX) activities. The PAH induced the increase in intracellular reactive oxygen species, NO, TNF-α, IL-6, iNOS, and COX-2, which were markedly suppressed by CCS extract in a dose-dependent manner, comparable to the effects of chlorogenic acid, caffeine, and theophylline. In conclusion, CCS extract inhibits PAH-induced inflammation by reducing pro-inflammatory cytokines and reactive oxygen species (ROS) production in RAW 264.7 cells. This effect is likely due to the synergistic effects of its bioactive compounds. Chlorogenic acid showed strong antioxidant and anti-inflammatory activities, while caffeine and theophylline enhanced anti-inflammatory activity. CCS extract did not irritate the hen's egg chorioallantoic membrane. Therefore, CCS extract shows its potential as a promising cosmeceutical ingredient for safely alleviating inflammatory skin diseases caused by air pollution.
Collapse
Affiliation(s)
- Weeraya Preedalikit
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | | | - Kanokwan Kiattisin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
9
|
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
|
10
|
Wardhani K, Yazzie S, McVeigh C, Edeh O, Grimes M, Jacquez Q, Dixson C, Barr E, Liu R, Bolt AM, Feng C, Zychowski KE. Systemic immunological responses are dependent on sex and ovarian hormone presence following acute inhaled woodsmoke exposure. Part Fibre Toxicol 2024; 21:27. [PMID: 38797836 PMCID: PMC11129474 DOI: 10.1186/s12989-024-00587-5] [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: 12/01/2023] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Rural regions of the western United States have experienced a noticeable surge in both the frequency and severity of acute wildfire events, which brings significant challenges to both public safety and environmental conservation efforts, with impacts felt globally. Identifying factors contributing to immune dysfunction, including endocrinological phenotypes, is essential to understanding how hormones may influence toxicological susceptibility. METHODS This exploratory study utilized male and female C57BL/6 mice as in vivo models to investigate distinct responses to acute woodsmoke (WS) exposure with a focus on sex-based differences. In a second set of investigations, two groups were established within the female mouse cohort. In one group, mice experienced ovariectomy (OVX) to simulate an ovarian hormone-deficient state similar to surgical menopause, while the other group received Sham surgery as controls, to investigate the mechanistic role of ovarian hormone presence in driving immune dysregulation following acute WS exposure. Each experimental cohort followed a consecutive 2-day protocol with daily 4-h exposure intervals under two conditions: control HEPA-filtered air (FA) and acute WS to simulate an acute wildfire episode. RESULTS Metals analysis of WS particulate matter (PM) revealed significantly increased levels of 63Cu, 182W, 208Pb, and 238U, compared to filtered air (FA) controls, providing insights into the specific metal components most impacted by the changing dynamics of wildfire occurrences in the region. Male and female mice exhibited diverse patterns in lung mRNA cytokine expression following WS exposure, with males showing downregulation and females displaying upregulation, notably for IL-1β, TNF-α, CXCL-1, CCL-5, TGF-β, and IL-6. After acute WS exposure, there were notable differences in the responses of macrophages, neutrophils, and bronchoalveolar lavage (BAL) cytokines IL-10, IL-6, IL-1β, and TNF-α. Significant diverse alterations were observed in BAL cytokines, specifically IL-1β, IL-10, IL-6, and TNF-α, as well as in the populations of immune cells, such as macrophages and polymorphonuclear leukocytes, in both Sham and OVX mice, following acute WS exposure. These findings elucidated the profound influence of hormonal changes on inflammatory outcomes, delineating substantial sex-related differences in immune activation and revealing altered immune responses in OVX mice due to ovarian hormone deficiency. In addition, the flow cytometry analysis highlighted the complex interaction between OVX surgery, acute WS exposure, and their collective impact on immune cell populations within the hematopoietic bone marrow niche. CONCLUSIONS In summary, both male and female mice, alongside females subjected to OVX and those who had sham surgery, exhibit significant variations in the expression of proinflammatory cytokines, chemokines, lung mRNA gene expression, and related functional networks linked to signaling pathways. These differences potentially act as mediators of sex-specific and hormonal influences in the systemic inflammatory response to acute WS exposure during a wildfire event. Understanding the regulatory roles of genes expressed differentially under environmental stressors holds considerable implications, aiding in identifying sex-specific therapeutic targets for addressing acute lung inflammation and injury.
Collapse
Affiliation(s)
- Kartika Wardhani
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
- Biochemistry and Biotechnology Group (B-TEK), Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Sydnee Yazzie
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Charlotte McVeigh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Onamma Edeh
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Martha Grimes
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Quiteria Jacquez
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Connor Dixson
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Edward Barr
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Alicia M Bolt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Changjian Feng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA
| | - Katherine E Zychowski
- College of Nursing, University of New Mexico-Health Sciences Center, Albuquerque, New Mexico, USA.
| |
Collapse
|
11
|
Khraishah H, Chen Z, Rajagopalan S. Understanding the Cardiovascular and Metabolic Health Effects of Air Pollution in the Context of Cumulative Exposomic Impacts. Circ Res 2024; 134:1083-1097. [PMID: 38662860 PMCID: PMC11253082 DOI: 10.1161/circresaha.124.323673] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Poor air quality accounts for more than 9 million deaths a year globally according to recent estimates. A large portion of these deaths are attributable to cardiovascular causes, with evidence indicating that air pollution may also play an important role in the genesis of key cardiometabolic risk factors. Air pollution is not experienced in isolation but is part of a complex system, influenced by a host of other external environmental exposures, and interacting with intrinsic biologic factors and susceptibility to ultimately determine cardiovascular and metabolic outcomes. Given that the same fossil fuel emission sources that cause climate change also result in air pollution, there is a need for robust approaches that can not only limit climate change but also eliminate air pollution health effects, with an emphasis of protecting the most susceptible but also targeting interventions at the most vulnerable populations. In this review, we summarize the current state of epidemiologic and mechanistic evidence underpinning the association of air pollution with cardiometabolic disease and how complex interactions with other exposures and individual characteristics may modify these associations. We identify gaps in the current literature and suggest emerging approaches for policy makers to holistically approach cardiometabolic health risk and impact assessment.
Collapse
Affiliation(s)
- Haitham Khraishah
- Division of Cardiovascular Medicine, University of Maryland Medical Center, Baltimore (H.K.)
| | - Zhuo Chen
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH (Z.C., S.R.)
- Case Western Reserve University School of Medicine, Cleveland, OH (Z.C., S.R.)
| | - Sanjay Rajagopalan
- Harrington Heart and Vascular Institute, University Hospitals, Cleveland, OH (Z.C., S.R.)
- Case Western Reserve University School of Medicine, Cleveland, OH (Z.C., S.R.)
| |
Collapse
|
12
|
White AR. The firestorm within: A narrative review of extreme heat and wildfire smoke effects on brain health. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 922:171239. [PMID: 38417511 DOI: 10.1016/j.scitotenv.2024.171239] [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: 11/20/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/01/2024]
Abstract
Climate change is generating increased heatwaves and wildfires across much of the world. With these escalating environmental changes comes greater impacts on human health leading to increased numbers of people suffering from heat- and wildfire smoke-associated respiratory and cardiovascular impairment. One area of health impact of climate change that has received far less attention is the effects of extreme heat and wildfire smoke exposure on human brain health. As elevated temperatures, and wildfire-associated smoke, are increasingly experienced simultaneously over summer periods, understanding this combined impact is critical to management of human health especially in the elderly, and people with dementia, and other neurological disorders. Both extreme heat and wildfire smoke air pollution (especially particulate matter, PM) induce neuroinflammatory and cerebrovascular effects, oxidative stress, and cognitive impairment, however the combined effect of these impacts are not well understood. In this narrative review, a comprehensive examination of extreme heat and wildfire smoke impact on human brain health is presented, with a focus on how these factors contribute to cognitive impairment, and dementia, one of the leading health issues today. Also discussed is the potential impact of combined heat and wildfire smoke on brain health, and where future efforts should be applied to help advance knowledge in this rapidly growing and critical field of health research.
Collapse
Affiliation(s)
- Anthony R White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, 70211 Kuopio, Finland; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD, Australia.
| |
Collapse
|
13
|
Wang J, He W, Yue H, Zhao P, Li J. Effective-components combination alleviates PM2.5-induced inflammation by evoking macrophage autophagy in COPD. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117537. [PMID: 38043756 DOI: 10.1016/j.jep.2023.117537] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufei Yishen formula (BYF) is clinically used to treat chronic obstructive pulmonary disease (COPD). Effective-component compatibility (ECC) is a combination of five active components derived from BYF, which has an equal effect on COPD to BYF. Our previous study has also demonstrated that ECC can protect COPD rats against PM2.5 exposure. However, the precise mechanisms remain to be elucidated. AIM OF THE STUDY To explore the mechanism underlying the anti-inflammatory effects of ECC-BYF against PM2.5-accelerated COPD. MATERIALS AND METHODS MH-S macrophages were stimulated by PM2.5 suspension to establish an in vitro model. Western blotting and immunofluorescent staining were used to measure the protein levels of autophagy markers. ELISA and quantitative PCR were used to detect the levels of inflammatory cytokines. In vivo, an established PM2.5-accelerated COPD rat model was used to determine the protective effect of ECC-BYF. Lung function, pathology, autophagy, and inflammatory mediators were detected. RESULTS Firstly, we observed a significantly increased number of macrophages in the lungs upon PM2.5 exposure. Then, decreased autophagy flux while elevated inflammation was detected in PM2.5-exposed rats and MH-S cells. In MH-S cells, ECC-BYF significantly suppressed the PM2.5-increased inflammatory cytokines production, which was accompanied by the enhancement of autophagy flux. An autophagy inhibitor counteracted the anti-inflammatory effect elicited by ECC-BYF. In addition, ECC-BYF stimulated Foxo3 nuclear translocation and upregulated Foxo3 expression, whereas Foxo3 knockdown abrogated the inhibitory effect of ECC-BYF on inflammation. In PM2.5-accelerated COPD rats, ECC-BYF also attenuated the autophagy disruption and increased Foxo3 in the lungs, finally resulting in a suppression of pulmonary inflammation and an enhancement of lung function. CONCLUSION ECC-BYF can ameliorate PM2.5-aggravated inflammation in COPD, which might be associated with the enhancement of autophagy flux in alveolar macrophages through the activation of Foxo3 signals.
Collapse
Affiliation(s)
- Jing Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Weijing He
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China
| | - Huiyu Yue
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, Henan Province, 450046, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
14
|
Noh M, Sim JY, Kim J, Ahn JH, Min HY, Lee JU, Park JS, Jeong JY, Lee JY, Lee SY, Lee HJ, Park CS, Lee HY. Particulate matter-induced metabolic recoding of epigenetics in macrophages drives pathogenesis of chronic obstructive pulmonary disease. JOURNAL OF HAZARDOUS MATERIALS 2024; 464:132932. [PMID: 37988864 DOI: 10.1016/j.jhazmat.2023.132932] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/19/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a group of illnesses associated with unresolved inflammation in response to toxic environmental stimuli. Persistent exposure to PM is a major risk factor for COPD, but the underlying mechanism remains unclear. Using our established mouse model of PM-induced COPD, we find that repeated PM exposure provokes macrophage-centered chronic inflammation and COPD development. Mechanistically, chronic PM exposure induces transcriptional downregulation of HAAO, KMO, KYNU, and QPRT in macrophages, which are the enzymes of de novo NAD+ synthesis pathway (kynurenine pathway; KP), via elevated chromatin binding of the CCCTC-binding factor (CTCF) near the transcriptional regulatory regions of the enzymes. Subsequent reduction of NAD+ and SIRT1 function increases histone acetylation, resulting in elevated expression of pro-inflammatory genes in PM-exposed macrophages. Activation of SIRT1 by nutraceutical resveratrol mitigated PM-induced chronic inflammation and COPD development. In agreement, increased levels of histone acetylation and decreased expression of KP enzymes were observed in pulmonary macrophages of COPD patients. We newly provide an evidence that dysregulated NAD+ metabolism and consecutive SIRT1 deficiency significantly contribute to the pathological activation of macrophages during PM-mediated COPD pathogenesis. Additionally, targeting PM-induced intertwined metabolic and epigenetic reprogramming in macrophages is an effective strategy for COPD treatment.
Collapse
Affiliation(s)
- Myungkyung Noh
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jeong Yeon Sim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jisung Kim
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Jee Hwan Ahn
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Young Min
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Natural Products Research Institute, Seoul National University, Seoul 08826, South Korea
| | - Jong-Uk Lee
- Department of Medical Bioscience, Graduate School, Soonchunhyang University, 22, Soonchunhyang-ro, Asan 31538, South Korea
| | - Jong-Sook Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Jae Young Lee
- Department of Environmental and Safety Engineering, Ajou University, Suwon 16499, South Korea
| | - Shin Yup Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Lung Cancer Center, Kyungpook National University Chilgok Hospital, Daegu 41944, South Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Gyeonggi do, South Korea
| | - Choon-Sik Park
- Soonchunhyang University Bucheon Hospital, Bucheon-si, Gyeonggi-do 14584, South Korea
| | - Ho-Young Lee
- Creative Research Initiative Center for Concurrent Control of Emphysema and Lung Cancer, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology and College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
15
|
Shin HJ, Yang WK, Lee YC, Kim S, Moon SO, Kwon YJ, Noh HJ, Kim KH, Kim BK, Shin CH, Chae MY, Yun SH, Kim SH. Protective effect of the mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt extract on respiratory disorders. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115856. [PMID: 38134637 DOI: 10.1016/j.ecoenv.2023.115856] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/07/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Air pollutants, such as particulate matter (PM) and diesel exhaust particles (DEP), are associated with respiratory diseases. Therefore, preventive and therapeutic strategies against PM-and DEP (PM10D)-induced respiratory diseases are needed. Herein, we evaluate the protective effects of a mixture of Lactiplantibacillus plantarum KC3 and Leonurus Japonicas Houtt (LJH) extract against airway inflammation associated with exposure to PM10D. To determine the anti-inflammatory effects of the LJH extract, reactive oxygen species (ROS) production and the expression of inflammatory pathways were determined in PM10-induced MH-S cells. For the respiratory protective effects, BALB/c mice were exposed to PM10D via intranasal injection, and a mixture of L. plantarum KC3 and LJH extract was administered orally for 12 days. LJH extract inhibited ROS production and the phosphorylation of downstream factors of NF-κB in PM10-stimulated MH-S cells. The mixture of L. plantarum KC3 and LJH repressed the infiltration of neutrophils, reduced the immune cells number, and suppressed the proinflammatory mediators and cyclooxygenase (COX)-2 expressions in PM10D-induced airway inflammation with reduced phosphorylation of downstream factors of NF-κB. In addition, these effects were not observed in an alveolar macrophage depleted PM10D-induced mouse model using clodronate liposomes. The extract mixture also regulated gut microbiota in feces and upregulated the mRNA expression of Foxp3, transforming growth factor (TGF)-β1, and interleukin (IL)-10 in the colon. The L. plantarum KC3 and LJH extract mixture may inhibit alveolar macrophage- and neutrophil-mediated inflammatory responses and regulate gut microbiota and immune response in PM10D-induced airway inflammation, suggesting it is a potential remedy to prevent and cure airway inflammation and respiratory disorders.
Collapse
Affiliation(s)
- Han Jae Shin
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Won-Kyung Yang
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Young Chul Lee
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Soeun Kim
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Sung Ok Moon
- KT&G Research Institute, Daejeon 34128, the Republic of Korea
| | - Yoo Jin Kwon
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Hye-Ji Noh
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Kyung Hwan Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Byoung Kook Kim
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Chang Hun Shin
- Chong Kun Dang Bio Research Institute (CKDBiO), Seoul 03722, the Republic of Korea
| | - Min-Young Chae
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Su-Hyeon Yun
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, the Republic of Korea.
| |
Collapse
|
16
|
Priyadarshini NP, Gopamma D, Srinivas N, Malla RR, Kumar KS. Particulate Matter and Its Impact on Macrophages: Unraveling the Cellular Response for Environmental Health. Crit Rev Oncog 2024; 29:33-42. [PMID: 38989736 DOI: 10.1615/critrevoncog.2024053305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Particulate matter (PM) imposes a significant impact to environmental health with deleterious effects on the human pulmonary and cardiovascular systems. Macrophages (Mφ), key immune cells in lung tissues, have a prominent role in responding to inhaled cells, accommodating inflammation, and influencing tissue repair processes. Elucidating the critical cellular responses of Mφ to PM exposure is essential to understand the mechanisms underlying PM-induced health effects. The present review aims to give a glimpse on literature about the PM interaction with Mφ, triggering the cellular events causing the inflammation, oxidative stress (OS) and tissue damage. The present paper reviews the different pathways involved in Mφ activation upon PM exposure, including phagocytosis, intracellular signaling cascades, and the release of pro-inflammatory mediators. Potential therapeutic strategies targeting Mφ-mediated responses to reduce PM-induced health effects are also discussed. Overall, unraveling the complex interplay between PM and Mφ sheds light on new avenues for environmental health research and promises to develop targeted interventions to reduce the burden of PM-related diseases on global health.
Collapse
Affiliation(s)
- Nyayapathi Priyanka Priyadarshini
- Department of Environmental Science, GITAM School of Science, GITAM Deemed to be University, Visakhapatnam, Andhra Pradesh 530045, India
| | - Daka Gopamma
- Department of Environmental Science, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Namuduri Srinivas
- Department of Environmental Science, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, Gandhi Institute of Technology and Management (GITAM) (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| | - Kolli Suresh Kumar
- Department of Environmental Science, GITAM School of Science, GITAM Deemed to be University, Visakhapatnam, Andhra Pradesh 530045, India
| |
Collapse
|
17
|
Smyth T, Jaspers I. Diesel exhaust particles induce polarization state-dependent functional and transcriptional changes in human monocyte-derived macrophages. Am J Physiol Lung Cell Mol Physiol 2024; 326:L83-L97. [PMID: 38084400 PMCID: PMC11279754 DOI: 10.1152/ajplung.00085.2023] [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: 04/17/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 01/10/2024] Open
Abstract
Macrophage populations exist on a spectrum between the proinflammatory M1 and proresolution M2 states and have demonstrated the ability to reprogram between them after exposure to opposing polarization stimuli. Particulate matter (PM) has been repeatedly linked to worsening morbidity and mortality following respiratory infections and has been demonstrated to modify macrophage function and polarization. The purpose of this study was to determine whether diesel exhaust particles (DEP), a key component of airborne PM, would demonstrate polarization state-dependent effects on human monocyte-derived macrophages (hMDMs) and whether DEP would modify macrophage reprogramming. CD14+CD16- monocytes were isolated from the blood of healthy human volunteers and differentiated into macrophages with macrophage colony-stimulating factor (M-CSF). Resulting macrophages were left unpolarized or polarized into the proresolution M2 state before being exposed to DEP, M1-polarizing conditions (IFN-γ and LPS), or both and tested for phagocytic function, secretory profile, gene expression patterns, and bioenergetic properties. Contrary to previous reports, we observed a mixed M1/M2 phenotype in reprogrammed M2 cells when considering the broader range of functional readouts. In addition, we determined that DEP exposure dampens phagocytic function in all polarization states while modifying bioenergetic properties in M1 macrophages preferentially. Together, these data suggest that DEP exposure of reprogrammed M2 macrophages results in a highly inflammatory, highly energetic subpopulation of macrophages that may contribute to the poor health outcomes following PM exposure during respiratory infections.NEW & NOTEWORTHY We determined that reprogramming M2 macrophages in the presence of diesel exhaust particles (DEP) results in a highly inflammatory mixed M1/M2 phenotype. We also demonstrated that M1 macrophages are particularly vulnerable to particulate matter (PM) exposure as seen by dampened phagocytic function and modified bioenergetics. Our study suggests that PM causes reprogrammed M2 macrophages to become a highly energetic, highly secretory subpopulation of macrophages that may contribute to negative health outcomes observed in humans after PM exposure.
Collapse
Affiliation(s)
- Timothy Smyth
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Ilona Jaspers
- Curriculum in Toxicology & Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Center for Environmental Medicine, Asthma, and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| |
Collapse
|
18
|
Kamanzi C, Becker M, Jacobs M, Konečný P, Von Holdt J, Broadhurst J. The impact of coal mine dust characteristics on pathways to respiratory harm: investigating the pneumoconiotic potency of coals. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2023; 45:7363-7388. [PMID: 37131112 PMCID: PMC10517901 DOI: 10.1007/s10653-023-01583-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 04/19/2023] [Indexed: 05/04/2023]
Abstract
Exposure to dust from the mining environment has historically resulted in epidemic levels of mortality and morbidity from pneumoconiotic diseases such as silicosis, coal workers' pneumoconiosis (CWP), and asbestosis. Studies have shown that CWP remains a critical issue at collieries across the globe, with some countries facing resurgent patterns of the disease and additional pathologies from long-term exposure. Compliance measures to reduce dust exposure rely primarily on the assumption that all "fine" particles are equally toxic irrespective of source or chemical composition. For several ore types, but more specifically coal, such an assumption is not practical due to the complex and highly variable nature of the material. Additionally, several studies have identified possible mechanisms of pathogenesis from the minerals and deleterious metals in coal. The purpose of this review was to provide a reassessment of the perspectives and strategies used to evaluate the pneumoconiotic potency of coal mine dust. Emphasis is on the physicochemical characteristics of coal mine dust such as mineralogy/mineral chemistry, particle shape, size, specific surface area, and free surface area-all of which have been highlighted as contributing factors to the expression of pro-inflammatory responses in the lung. The review also highlights the potential opportunity for more holistic risk characterisation strategies for coal mine dust, which consider the mineralogical and physicochemical aspects of the dust as variables relevant to the current proposed mechanisms for CWP pathogenesis.
Collapse
Affiliation(s)
- Conchita Kamanzi
- Department of Chemical Engineering, Minerals to Metals Initiative, University of Cape Town, Cape Town, South Africa.
- Department of Chemical Engineering, Centre for Minerals Research, University of Cape Town, Cape Town, South Africa.
| | - Megan Becker
- Department of Chemical Engineering, Minerals to Metals Initiative, University of Cape Town, Cape Town, South Africa
- Department of Chemical Engineering, Centre for Minerals Research, University of Cape Town, Cape Town, South Africa
| | - Muazzam Jacobs
- Division of Immunology, Department of Pathology, Institute for Infectious Diseases and Molecular Medicine, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Johannesburg, South Africa
| | - Petr Konečný
- Division of Immunology, Department of Pathology, Institute for Infectious Diseases and Molecular Medicine, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Johanna Von Holdt
- Department of Environmental and Geographical Science, University of Cape Town, Cape Town, South Africa
| | - Jennifer Broadhurst
- Department of Chemical Engineering, Minerals to Metals Initiative, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
19
|
Jassim A, Rahrmann EP, Simons BD, Gilbertson RJ. Cancers make their own luck: theories of cancer origins. Nat Rev Cancer 2023; 23:710-724. [PMID: 37488363 DOI: 10.1038/s41568-023-00602-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/26/2023]
Abstract
Cancer has been a leading cause of death for decades. This dismal statistic has increased efforts to prevent the disease or to detect it early, when treatment is less invasive, relatively inexpensive and more likely to cure. But precisely how tissues are transformed continues to provoke controversy and debate, hindering cancer prevention and early intervention strategies. Various theories of cancer origins have emerged, including the suggestion that it is 'bad luck': the inevitable consequence of random mutations in proliferating stem cells. In this Review, we discuss the principal theories of cancer origins and the relative importance of the factors that underpin them. The body of available evidence suggests that developing and ageing tissues 'walk a tightrope', retaining adequate levels of cell plasticity to generate and maintain tissues while avoiding overstepping into transformation. Rather than viewing cancer as 'bad luck', understanding the complex choreography of cell intrinsic and extrinsic factors that characterize transformation holds promise to discover effective new ways to prevent, detect and stop cancer before it becomes incurable.
Collapse
Affiliation(s)
- Amir Jassim
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eric P Rahrmann
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ben D Simons
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - Richard J Gilbertson
- CRUK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Kjerulff B, Thisted Horsdal H, Kaspersen K, Mikkelsen S, Manh Dinh K, Hørup Larsen M, Rye Ostrowski S, Ullum H, Sørensen E, Birger Pedersen O, Topholm Bruun M, René Nielsen K, Brandt J, Geels C, Frohn LM, Christensen JH, Sigsgaard T, Eric Sabel C, Bøcker Pedersen C, Erikstrup C. Medium term moderate to low-level air pollution exposure is associated with higher C-reactive protein among healthy Danish blood donors. ENVIRONMENTAL RESEARCH 2023; 233:116426. [PMID: 37336432 DOI: 10.1016/j.envres.2023.116426] [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: 12/27/2022] [Revised: 05/10/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
Air pollution is a significant contributor to the global burden of disease with a plethora of associated health effects such as pulmonary and systemic inflammation. C-reactive protein (CRP) is associated with a wide range of diseases and is associated with several exposures. Studies on the effect of air pollution exposure on CRP levels in low to moderate pollution settings have shown inconsistent results. In this cross-sectional study high sensitivity CRP measurements on 18,463 Danish blood donors were linked to modelled air pollution data for NOx, NO2, O3, CO, SO2, NH3, mineral dust, black carbon, organic carbon, sea salt, secondary inorganic aerosols and its components, primary PM2.5, secondary organic aerosols, total PM2.5, and total PM10 at their residential address over the previous month. Associations were analysed using ordered logistic regression with CRP quartile as individuals outcome and air pollution exposure as scaled deciles. Analyses were adjusted for health related and socioeconomic covariates using health questionnaires and Danish register data. Exposure to different air pollution components was generally associated with higher CRP (odds ratio estimates ranging from 1.11 to 1.67), while exposure to a few air pollution components was associated with lower CRP. For example, exposure to NO2 increased the odds of high CRP 1.32-fold (95%CI 1.16-1.49), while exposure to NH3 decreased the odds of high CRP 0.81-fold (95%CI 0.73-0.89). This large study among healthy individuals found air pollution exposure to be associated with increased levels of CRP even in a setting with low to moderate air pollution levels.
Collapse
Affiliation(s)
- Bertram Kjerulff
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; Department of Clinical Medicine, Aarhus University, Denmark.
| | - Henriette Thisted Horsdal
- Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; National Centre for Register-based Research, Aarhus BSS, Aarhus University, DK-8210, Aarhus V, Denmark
| | - Kathrine Kaspersen
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark
| | - Susan Mikkelsen
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark
| | - Khoa Manh Dinh
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark
| | - Margit Hørup Larsen
- Dept. of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Sisse Rye Ostrowski
- Dept. of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Denmark
| | | | - Erik Sørensen
- Dept. of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Denmark; Dept. of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | | | | | - Jørgen Brandt
- Department of Environmental Science, Aarhus University, DK-4000, Roskilde, Denmark; IClimate - Interdisciplinary Centre for Climate Change, Aarhus University, DK-4000, Denmark
| | - Camilla Geels
- Department of Environmental Science, Aarhus University, DK-4000, Roskilde, Denmark; IClimate - Interdisciplinary Centre for Climate Change, Aarhus University, DK-4000, Denmark
| | - Lise M Frohn
- Department of Environmental Science, Aarhus University, DK-4000, Roskilde, Denmark; IClimate - Interdisciplinary Centre for Climate Change, Aarhus University, DK-4000, Denmark
| | - Jesper H Christensen
- Department of Environmental Science, Aarhus University, DK-4000, Roskilde, Denmark; IClimate - Interdisciplinary Centre for Climate Change, Aarhus University, DK-4000, Denmark
| | - Torben Sigsgaard
- Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; Department of Public Health, Aarhus University, DK-8000, Aarhus, Denmark
| | - Clive Eric Sabel
- Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; Department of Public Health, Aarhus University, DK-8000, Aarhus, Denmark
| | - Carsten Bøcker Pedersen
- Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; National Centre for Register-based Research, Aarhus BSS, Aarhus University, DK-8210, Aarhus V, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, DK-8200, Aarhus N, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| |
Collapse
|
21
|
He C, Xie L, Gu L, Yan H, Feng S, Zeng C, Danzhen W, Zhang X, Han M, Li Z, Duoji Z, Guo B, Zhang J, Hong F, Zhao X. Anemia is associated with long-term exposure to PM 2.5 and its components: a large population-based study in Southwest China. Ther Adv Hematol 2023; 14:20406207231189922. [PMID: 37654523 PMCID: PMC10467225 DOI: 10.1177/20406207231189922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/07/2023] [Indexed: 09/02/2023] Open
Abstract
Background Anemia is linked to PM2.5 (particulate matter with aerodynamic diameters of ⩽2.5 μm) exposure, which can increase the risk of various negative health outcomes. It remains unclear which PM2.5 components are associated with anemia and the respective contribution of each component to this association. Objective This study aimed at investigating the association between PM2.5 and anemia in the general population and to identify the most critical PM2.5 toxic components in this association. Design Cross-sectional study. Methods Our study involved a large cohort of 73,511 individuals aged 30-79 from China's multi-ethnic population. We employed satellite observations and the chemical transport model (GEOS-Chem)to estimate the long-term exposure to PM2.5 and its components. Anemia was defined, according to WHO guidelines, as Hb levels below 130 g/L for men and below 120 g/L for women. Through logistic regression, we investigated the association between PM2.5 components and anemia. By utilizing weighted quantile sum (WQS) analysis, we identified key components and gained insights into their combined impact on anemia. Overall, our study sheds light on the relationship between PM2.5 exposure, its constituents, and the risk of anemia in a large cohort. Results PM2.5 and three components, nitrate (NIT), organic matter (OM), and soil particles (SOIL), were associated with anemia. Per-standard deviation increase in the 3-year average concentrations of PM2.5 [odds ratio (OR): 1.14, 95% confidence interval (CI): 1.01, 1.28], NIT (1.20, 1.06, 1.35), OM (1.17, 1.04, 1.32), and SOIL (1.22, 1.11, 1.33) were associated with higher odds of anemia. In WQS regression analysis, the WQS index was associated with anemia (OR: 1.29, 95% CI: 1.13, 1.47). SOIL has the highest weight among all PM2.5 components. Conclusions Long-term exposure to PM2.5 and its constituents is associated with anemia. Moreover, SOIL might be the most critical component of the relationship between PM2.5 and anemia. Our research increases the evidence of the association between PM2.5 and anemia in the general population, and targeted emission control measures should be taken into consideration to mitigate the adverse effects of PM2.5-related anemia.
Collapse
Affiliation(s)
- Congyuan He
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingxi Gu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongyu Yan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyu Feng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunmei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wangjiu Danzhen
- Tibet Center for Disease Control and Prevention CN, Lhasa, Tibet, China
| | - Xuehui Zhang
- School of Public Health, Kunming Medical University, Kunming, Yunnan, China
| | - Mingming Han
- Chengdu Center for Disease Control & Prevention, Chengdu, Sichuan, China
| | - Zhifeng Li
- Chongqing Municipal Center for Disease Control and Prevention, Chongqing, China
| | | | - Bing Guo
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Juying Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Chengdu, Sichuan 610041, China
| | - Feng Hong
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, University Town, Guian New Area, Guiyang, Guizhou 550025, China
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University, No. 16, Section 3, Renmin South Road, Chengdu, Sichuan 610041, China
| |
Collapse
|
22
|
Hasegawa Y, Okamura T, Nakajima H, Kitagawa N, Majima S, Okada H, Senmaru T, Ushigome E, Nakanishi N, Hamaguchi M, Takano H, Fukui M. Metabolic outcomes and changes in innate immunity induced by diesel exhaust particles airway exposure and high-fat high-sucrose diet. Life Sci 2023; 326:121794. [PMID: 37224953 DOI: 10.1016/j.lfs.2023.121794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023]
Abstract
AIMS Epidemiological studies have shown that exposure to diesel exhaust particles (DEP) is associated with metabolic diseases. We used mice with nonalcoholic fatty liver disease (NAFLD) caused by a high-fat, high-sucrose diet (HFHSD), which mimics a Western diet, to investigate the mechanism of NAFLD exacerbation via changes in innate immunity in the lungs by airway exposure to DEP. MAIN METHODS Six-week-old C57BL6/J male mice were fed HFHSD, and DEP was administered endotracheally once a week for eight weeks. The histology, gene expression, innate immunity cells in the lung and liver, and the serum inflammatory cytokine levels, were investigated. KEY FINDINGS Under the HFHSD, DEP increased blood glucose levels, serum lipid levels, and NAFLD activity scores, and also the expression of genes associated with inflammation in the lungs and liver. DEP caused an increase in ILC1s, ILC2s, ILC3s, and M1 macrophages in the lungs and a marked increase in ILC1s, ILC3s, M1 macrophages, and natural killer cells in the liver, while ILC2 levels were not changed. Furthermore, DEP caused high levels of inflammatory cytokines in the serum. SIGNIFICANCE Chronic exposure to DEP in HFHSD-fed mice increased inflammatory cells involved in innate immunity in the lungs and raised local inflammatory cytokine levels. This inflammation spread throughout the body, suggesting the association with the progression of NAFLD via increased inflammatory cells involved in innate immunity and inflammatory cytokine levels in the liver. These findings contribute to a better understanding of the role of innate immunity in air pollution-related systemic diseases, especially metabolic diseases.
Collapse
Affiliation(s)
- Yuka Hasegawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Hanako Nakajima
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Nobuko Kitagawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Saori Majima
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan.
| | - Hirohisa Takano
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, 615-8530, Japan; Institute for International Academic Research, Kyoto University of Advanced Science, Kyoto 615-8577, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto 602-8566, Japan
| |
Collapse
|
23
|
Kao CM, Chen YM, Huang WN, Chen YH, Chen HH. Association between air pollutants and initiation of biological therapy in patients with ankylosing spondylitis: a nationwide, population-based, nested case-control study. Arthritis Res Ther 2023; 25:75. [PMID: 37147678 PMCID: PMC10161550 DOI: 10.1186/s13075-023-03060-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/29/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Outdoor air pollution has been found to trigger systemic inflammatory responses and aggravate the activity of certain rheumatic diseases. However, few studies have explored the influence of air pollution on the activity of ankylosing spondylitis (AS). As patients with active AS in Taiwan can be reimbursed through the National Health Insurance programme for biological therapy, we investigated the association between air pollutants and the initiation of reimbursed biologics for active AS. METHODS Since 2011, hourly concentrations of ambient air pollutants, including PM2.5, PM10, NO2, CO, SO2, and O3, have been estimated in Taiwan. Using Taiwanese National Health Insurance Research Database, we identified patients with newly diagnosed AS from 2003 to 2013. We selected 584 patients initiating biologics from 2012 to 2013 and 2336 gender-, age at biologic initiation-, year of AS diagnosis- and disease duration-matched controls. We examined the associations of biologics initiation with air pollutants exposure within 1 year prior to biologic use whilst adjusting for potential confounders, including disease duration, urbanisation level, monthly income, Charlson comorbidity index (CCI), uveitis, psoriasis and the use of medications for AS. Results are shown as adjusted odds ratio (aOR) with 95% confidence intervals (CIs). RESULTS The initiation of biologics was associated with exposure to CO (per 1 ppm) (aOR, 8.57; 95% CI, 2.02-36.32) and NO2 (per 10 ppb) (aOR, 0.23; 95% CI, 0.11-0.50). Other independent predictors included disease duration (incremental year, aOR, 8.95), CCI (aOR, 1.31), psoriasis (aOR, 25.19), use of non-steroidal anti-inflammatory drugs (aOR, 23.66), methotrexate use (aOR, 4.50; 95% CI, 2.93-7.00), sulfasalazine use (aOR, 12.16; 95% CI, 8.98-15.45) and prednisolone equivalent dosages (mg/day, aOR, 1.12). CONCLUSIONS This nationwide, population-based study revealed the initiation of reimbursed biologics was positively associated with CO levels, but negatively associated with NO2 levels. Major limitations included lack of information on individual smoking status and multicollinearity amongst air pollutants.
Collapse
Affiliation(s)
- Chung-Mao Kao
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, No.1650, Sec.4, Taiwan Boulevard, Taichung, 40705, Taiwan
- Division of Translational Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Ming Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, No.1650, Sec.4, Taiwan Boulevard, Taichung, 40705, Taiwan
- Division of Translational Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Nan Huang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, No.1650, Sec.4, Taiwan Boulevard, Taichung, 40705, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Business Administration, Ling-Tung University, Taichung, Taiwan
| | - Yi-Hsing Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, No.1650, Sec.4, Taiwan Boulevard, Taichung, 40705, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsin-Hua Chen
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, No.1650, Sec.4, Taiwan Boulevard, Taichung, 40705, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
- Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung, Taiwan.
- Institute of Public Health and Community Medicine Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Big Data Center, National Chung Hsing University, Taichung, Taiwan.
- Division of General Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.
| |
Collapse
|
24
|
Hill W, Lim EL, Weeden CE, Lee C, Augustine M, Chen K, Kuan FC, Marongiu F, Evans EJ, Moore DA, Rodrigues FS, Pich O, Bakker B, Cha H, Myers R, van Maldegem F, Boumelha J, Veeriah S, Rowan A, Naceur-Lombardelli C, Karasaki T, Sivakumar M, De S, Caswell DR, Nagano A, Black JRM, Martínez-Ruiz C, Ryu MH, Huff RD, Li S, Favé MJ, Magness A, Suárez-Bonnet A, Priestnall SL, Lüchtenborg M, Lavelle K, Pethick J, Hardy S, McRonald FE, Lin MH, Troccoli CI, Ghosh M, Miller YE, Merrick DT, Keith RL, Al Bakir M, Bailey C, Hill MS, Saal LH, Chen Y, George AM, Abbosh C, Kanu N, Lee SH, McGranahan N, Berg CD, Sasieni P, Houlston R, Turnbull C, Lam S, Awadalla P, Grönroos E, Downward J, Jacks T, Carlsten C, Malanchi I, Hackshaw A, Litchfield K, DeGregori J, Jamal-Hanjani M, Swanton C. Lung adenocarcinoma promotion by air pollutants. Nature 2023; 616:159-167. [PMID: 37020004 PMCID: PMC7614604 DOI: 10.1038/s41586-023-05874-3] [Citation(s) in RCA: 309] [Impact Index Per Article: 154.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 02/21/2023] [Indexed: 04/07/2023]
Abstract
A complete understanding of how exposure to environmental substances promotes cancer formation is lacking. More than 70 years ago, tumorigenesis was proposed to occur in a two-step process: an initiating step that induces mutations in healthy cells, followed by a promoter step that triggers cancer development1. Here we propose that environmental particulate matter measuring ≤2.5 μm (PM2.5), known to be associated with lung cancer risk, promotes lung cancer by acting on cells that harbour pre-existing oncogenic mutations in healthy lung tissue. Focusing on EGFR-driven lung cancer, which is more common in never-smokers or light smokers, we found a significant association between PM2.5 levels and the incidence of lung cancer for 32,957 EGFR-driven lung cancer cases in four within-country cohorts. Functional mouse models revealed that air pollutants cause an influx of macrophages into the lung and release of interleukin-1β. This process results in a progenitor-like cell state within EGFR mutant lung alveolar type II epithelial cells that fuels tumorigenesis. Ultradeep mutational profiling of histologically normal lung tissue from 295 individuals across 3 clinical cohorts revealed oncogenic EGFR and KRAS driver mutations in 18% and 53% of healthy tissue samples, respectively. These findings collectively support a tumour-promoting role for PM2.5 air pollutants and provide impetus for public health policy initiatives to address air pollution to reduce disease burden.
Collapse
Affiliation(s)
- William Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Emilia L Lim
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare E Weeden
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Claudia Lee
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Division of Medicine, University College London, London, UK
| | - Marcellus Augustine
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Division of Medicine, University College London, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - Kezhong Chen
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery and Thoracic Oncology Institute, Peking University People's Hospital, Beijing, China
| | - Feng-Che Kuan
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan, Taiwan
| | - Fabio Marongiu
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Edward J Evans
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David A Moore
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Felipe S Rodrigues
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Bjorn Bakker
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Hongui Cha
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Renelle Myers
- BC Cancer Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Febe van Maldegem
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jesse Boumelha
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Takahiro Karasaki
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Swapnanil De
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Deborah R Caswell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Ai Nagano
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Min Hyung Ryu
- Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, British Columbia, Canada
| | - Ryan D Huff
- Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, British Columbia, Canada
| | - Shijia Li
- Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, British Columbia, Canada
| | | | - Alastair Magness
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Alejandro Suárez-Bonnet
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
- Experimental Histopathology, The Francis Crick Institute, London, UK
| | - Simon L Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
- Experimental Histopathology, The Francis Crick Institute, London, UK
| | - Margreet Lüchtenborg
- National Disease Registration Service (NDRS), NHS England, Leeds, UK
- Centre for Cancer, Society and Public Health, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Katrina Lavelle
- National Disease Registration Service (NDRS), NHS England, Leeds, UK
| | - Joanna Pethick
- National Disease Registration Service (NDRS), NHS England, Leeds, UK
| | - Steven Hardy
- National Disease Registration Service (NDRS), NHS England, Leeds, UK
| | - Fiona E McRonald
- National Disease Registration Service (NDRS), NHS England, Leeds, UK
| | - Meng-Hung Lin
- Health Information and Epidemiology Laboratory, Chang-Gung Memorial Hospital, Chiayi, Taiwan
| | - Clara I Troccoli
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Flagship Biosciences, Boulder, CO, USA
| | - Moumita Ghosh
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - York E Miller
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Veterans Affairs Eastern Colorado Healthcare System, Aurora, CO, USA
| | - Daniel T Merrick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Robert L Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Veterans Affairs Eastern Colorado Healthcare System, Aurora, CO, USA
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Chris Bailey
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mark S Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Lao H Saal
- SAGA Diagnostics, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Yilun Chen
- SAGA Diagnostics, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Anthony M George
- SAGA Diagnostics, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Christopher Abbosh
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Nnennaya Kanu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Peter Sasieni
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Richard Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Clare Turnbull
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Stephen Lam
- BC Cancer Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip Awadalla
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Eva Grönroos
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Julian Downward
- Oncogene Biology Laboratory, The Francis Crick Institute, London, UK
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher Carlsten
- Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, British Columbia, Canada
| | - Ilaria Malanchi
- Tumour-Host Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Allan Hackshaw
- Cancer Research UK and UCL Cancer Trials Centre, London, UK
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
25
|
Bosch AJT, Rohm TV, AlAsfoor S, Low AJY, Keller L, Baumann Z, Parayil N, Stawiski M, Rachid L, Dervos T, Mitrovic S, Meier DT, Cavelti-Weder C. Lung versus gut exposure to air pollution particles differentially affect metabolic health in mice. Part Fibre Toxicol 2023; 20:7. [PMID: 36895000 PMCID: PMC9996885 DOI: 10.1186/s12989-023-00518-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Air pollution has emerged as an unexpected risk factor for diabetes. However, the mechanism behind remains ill-defined. So far, the lung has been considered as the main target organ of air pollution. In contrast, the gut has received little scientific attention. Since air pollution particles can reach the gut after mucociliary clearance from the lungs and through contaminated food, our aim was to assess whether exposure deposition of air pollution particles in the lung or the gut drive metabolic dysfunction in mice. METHODS To study the effects of gut versus lung exposure, we exposed mice on standard diet to diesel exhaust particles (DEP; NIST 1650b), particulate matter (PM; NIST 1649b) or phosphate-buffered saline by either intratracheal instillation (30 µg 2 days/week) or gavage (12 µg 5 days/week) over at least 3 months (total dose of 60 µg/week for both administration routes, equivalent to a daily inhalation exposure in humans of 160 µg/m3 PM2.5) and monitored metabolic parameters and tissue changes. Additionally, we tested the impact of the exposure route in a "prestressed" condition (high-fat diet (HFD) and streptozotocin (STZ)). RESULTS Mice on standard diet exposed to particulate air pollutants by intratracheal instillation developed lung inflammation. While both lung and gut exposure resulted in increased liver lipids, glucose intolerance and impaired insulin secretion was only observed in mice exposed to particles by gavage. Gavage with DEP created an inflammatory milieu in the gut as shown by up-regulated gene expression of pro-inflammatory cytokines and monocyte/macrophage markers. In contrast, liver and adipose inflammation markers were not increased. Beta-cell secretory capacity was impaired on a functional level, most likely induced by the inflammatory milieu in the gut, and not due to beta-cell loss. The differential metabolic effects of lung and gut exposures were confirmed in a "prestressed" HFD/STZ model. CONCLUSIONS We conclude that separate lung and gut exposures to air pollution particles lead to distinct metabolic outcomes in mice. Both exposure routes elevate liver lipids, while gut exposure to particulate air pollutants specifically impairs beta-cell secretory capacity, potentially instigated by an inflammatory milieu in the gut.
Collapse
Affiliation(s)
- Angela J T Bosch
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Theresa V Rohm
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Shefaa AlAsfoor
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Andy J Y Low
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Lena Keller
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Zora Baumann
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Neena Parayil
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Marc Stawiski
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Leila Rachid
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Thomas Dervos
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Sandra Mitrovic
- Department of Laboratory Medicine, University Hospital Basel, 4031, Basel, Switzerland
| | - Daniel T Meier
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Claudia Cavelti-Weder
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland. .,Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, 4031, Basel, Switzerland. .,Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), Rämistrasse 100, 8009, Zurich, Switzerland.
| |
Collapse
|
26
|
Alewel DI, Henriquez AR, Schladweiler MC, Grindstaff R, Fisher AA, Snow SJ, Jackson TW, Kodavanti UP. Intratracheal instillation of respirable particulate matter elicits neuroendocrine activation. Inhal Toxicol 2023; 35:59-75. [PMID: 35867597 PMCID: PMC10590194 DOI: 10.1080/08958378.2022.2100019] [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: 02/18/2022] [Accepted: 04/19/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Inhalation of ozone activates central sympathetic-adrenal-medullary and hypothalamic-pituitary-adrenal stress axes. While airway neural networks are known to communicate noxious stimuli to higher brain centers, it is not known to what extent responses generated from pulmonary airways contribute to neuroendocrine activation. MATERIALS AND METHODS Unlike inhalational exposures that involve the entire respiratory tract, we employed intratracheal (IT) instillations to expose only pulmonary airways to either soluble metal-rich residual oil fly ash (ROFA) or compressor-generated diesel exhaust particles (C-DEP). Male Wistar-Kyoto rats (12-13 weeks) were IT instilled with either saline, C-DEP or ROFA (5 mg/kg) and necropsied at 4 or 24 hr to assess temporal effects. RESULTS IT-instillation of particulate matter (PM) induced hyperglycemia as early as 30-min and glucose intolerance when measured at 2 hr post-exposure. We observed PM- and time-specific effects on markers of pulmonary injury/inflammation (ROFA>C-DEP; 24 hr>4hr) as corroborated by increases in lavage fluid injury markers, neutrophils (ROFA>C-DEP), and lymphocytes (ROFA). Increases in lavage fluid pro-inflammatory cytokines differed between C-DEP and ROFA in that C-DEP caused larger increases in TNF-α whereas ROFA caused larger increases in IL-6. No increases in circulating cytokines occurred. At 4 hr, PM impacts on neuroendocrine activation were observed through depletion of circulating leukocytes, increases in adrenaline (ROFA), and decreases in thyroid-stimulating-hormone, T3, prolactin, luteinizing-hormone, and testosterone. C-DEP and ROFA both increased lung expression of genes involved in acute stress and inflammatory processes. Moreover, small increases occurred in hypothalamic Fkbp5, a glucocorticoid-sensitive gene. CONCLUSION Respiratory alterations differed between C-DEP and ROFA, with ROFA inducing greater overall lung injury/inflammation; however, both PM induced a similar degree of neuroendocrine activation. These findings demonstrate neuroendocrine activation after pulmonary-only PM exposure, and suggest the involvement of pituitary- and adrenal-derived hormones.
Collapse
Affiliation(s)
- Devin I. Alewel
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States of America
| | - Andres R. Henriquez
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States of America
| | - Mette C. Schladweiler
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Rachel Grindstaff
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Anna A. Fisher
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Samantha J. Snow
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| | - Thomas W. Jackson
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States of America
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
27
|
Ishida Y, Kuninaka Y, Mukaida N, Kondo T. Immune Mechanisms of Pulmonary Fibrosis with Bleomycin. Int J Mol Sci 2023; 24:3149. [PMID: 36834561 PMCID: PMC9958859 DOI: 10.3390/ijms24043149] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Fibrosis and structural remodeling of the lung tissue can significantly impair lung function, often with fatal consequences. The etiology of pulmonary fibrosis (PF) is diverse and includes different triggers such as allergens, chemicals, radiation, and environmental particles. However, the cause of idiopathic PF (IPF), one of the most common forms of PF, remains unknown. Experimental models have been developed to study the mechanisms of PF, and the murine bleomycin (BLM) model has received the most attention. Epithelial injury, inflammation, epithelial-mesenchymal transition (EMT), myofibroblast activation, and repeated tissue injury are important initiators of fibrosis. In this review, we examined the common mechanisms of lung wound-healing responses after BLM-induced lung injury as well as the pathogenesis of the most common PF. A three-stage model of wound repair involving injury, inflammation, and repair is outlined. Dysregulation of one or more of these three phases has been reported in many cases of PF. We reviewed the literature investigating PF pathogenesis, and the role of cytokines, chemokines, growth factors, and matrix feeding in an animal model of BLM-induced PF.
Collapse
Affiliation(s)
| | | | | | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| |
Collapse
|
28
|
Lee MK, Kim HD, Lee SH, Lee JH. Curcumin Ameliorates Particulate Matter-Induced Pulmonary Injury through Bimodal Regulation of Macrophage Inflammation via NF-κB and Nrf2. Int J Mol Sci 2023; 24:ijms24031858. [PMID: 36768180 PMCID: PMC9915121 DOI: 10.3390/ijms24031858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/12/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023] Open
Abstract
The direct effects of particulate matter (PM) on lung injury and its specific molecular mechanisms are unclear. However, experimental evidence has shown that oxidative stress-mediated inflammation in macrophages is the main pathological outcome of PM exposure. Curcumin has been reported to protect organs against the disturbance of homeostasis caused by various toxic agents through anti-inflammatory and antioxidative effects. However, the protective action of curcumin against PM-induced pulmonary inflammation and the underlying mechanism have not been thoroughly investigated. In this study, we established a PM-induced pulmonary inflammation mouse model using the intratracheal instillation method to investigate the protective ability of curcumin against PM-induced pulmonary inflammation. Compared to the mice treated with PM only, the curcumin-treated mice showed alleviated alveolar damage, decreased immune cell infiltration, and reduced proinflammatory cytokine production in both lung tissue and BALF. To evaluate the underlying mechanism, the mouse macrophage cell line RAW264.7 was used. Pretreatment with curcumin prevented the production of PM-induced proinflammatory cytokines by deactivating NF-κB through the suppression of MAPK signaling pathways. Furthermore, curcumin appears to attenuate PM-induced oxidative stress through the activation of Nrf2 and downstream antioxidant signaling. Our findings demonstrate that curcumin protects against PM-induced lung injury by suppressing oxidative stress and inflammatory activation in macrophages.
Collapse
Affiliation(s)
- Min Kook Lee
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
- BK21 FOUR Research Group for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
| | - Hyo Dam Kim
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
- BK21 FOUR Research Group for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
| | - Suk Hee Lee
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
- BK21 FOUR Research Group for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
- Biological Clock-Based Anti-Aging Convergence RLRC, Korea University, Sejong 30019, Republic of Korea
- Correspondence: (S.H.L.); (J.H.L.); Tel.: +82-044-860-1764 (J.H.L.)
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea
- BK21 FOUR Research Group for Omics-Based Bio-Health in Food Industry, Korea University, Sejong 30019, Republic of Korea
- Biological Clock-Based Anti-Aging Convergence RLRC, Korea University, Sejong 30019, Republic of Korea
- Correspondence: (S.H.L.); (J.H.L.); Tel.: +82-044-860-1764 (J.H.L.)
| |
Collapse
|
29
|
Mechanisms of Lung Damage and Development of COPD Due to Household Biomass-Smoke Exposure: Inflammation, Oxidative Stress, MicroRNAs, and Gene Polymorphisms. Cells 2022; 12:cells12010067. [PMID: 36611860 PMCID: PMC9818405 DOI: 10.3390/cells12010067] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic exposure to indoor biomass smoke from the combustion of solid organic fuels is a major cause of disease burden worldwide. Almost 3 billion people use solid fuels such as wood, charcoal, and crop residues for indoor cooking and heating, accounting for approximately 50% of all households and 90% of rural households globally. Biomass smoke contains many hazardous pollutants, resulting in household air pollution (HAP) exposure that often exceeds international standards. Long-term biomass-smoke exposure is associated with Chronic Obstructive Pulmonary Disease (COPD) in adults, a leading cause of morbidity and mortality worldwide, chronic bronchitis, and other lung conditions. Biomass smoke-associated COPD differs from the best-known cigarette smoke-induced COPD in several aspects, such as a slower decline in lung function, greater airway involvement, and less emphysema, which suggests a different phenotype and pathophysiology. Despite the high burden of biomass-associated COPD, the molecular, genetic, and epigenetic mechanisms underlying its pathogenesis are poorly understood. This review describes the pathogenic mechanisms potentially involved in lung damage, the development of COPD associated with wood-derived smoke exposure, and the influence of genetic and epigenetic factors on the development of this disease.
Collapse
|
30
|
Li T, Yu Y, Sun Z, Duan J. A comprehensive understanding of ambient particulate matter and its components on the adverse health effects based from epidemiological and laboratory evidence. Part Fibre Toxicol 2022; 19:67. [PMID: 36447278 PMCID: PMC9707232 DOI: 10.1186/s12989-022-00507-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022] Open
Abstract
The impacts of air pollution on public health have become a great concern worldwide. Ambient particulate matter (PM) is a major air pollution that comprises a heterogeneous mixture of different particle sizes and chemical components. The chemical composition and physicochemical properties of PM change with space and time, which may cause different impairments. However, the mechanisms of the adverse effects of PM on various systems have not been fully elucidated and systematically integrated. The Adverse Outcome Pathway (AOP) framework was used to comprehensively illustrate the molecular mechanism of adverse effects of PM and its components, so as to clarify the causal mechanistic relationships of PM-triggered toxicity on various systems. The main conclusions and new insights of the correlation between public health and PM were discussed, especially at low concentrations, which points out the direction for further research in the future. With the deepening of the study on its toxicity mechanism, it was found that PM can still induce adverse health effects with low-dose exposure. And the recommended Air Quality Guideline level of PM2.5 was adjusted to 5 μg/m3 by World Health Organization, which meant that deeper and more complex mechanisms needed to be explored. Traditionally, oxidative stress, inflammation, autophagy and apoptosis were considered the main mechanisms of harmful effects of PM. However, recent studies have identified several emerging mechanisms involved in the toxicity of PM, including pyroptosis, ferroptosis and epigenetic modifications. This review summarized the comprehensive evidence on the health effects of PM and the chemical components of it, as well as the combined toxicity of PM with other air pollutants. Based on the AOP Wiki and the mechanisms of PM-induced toxicity at different levels, we first constructed the PM-related AOP frameworks on various systems.
Collapse
Affiliation(s)
- Tianyu Li
- grid.24696.3f0000 0004 0369 153XDepartment of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Yang Yu
- grid.24696.3f0000 0004 0369 153XDepartment of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Zhiwei Sun
- grid.24696.3f0000 0004 0369 153XDepartment of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| | - Junchao Duan
- grid.24696.3f0000 0004 0369 153XDepartment of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069 People’s Republic of China ,grid.24696.3f0000 0004 0369 153XBeijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069 People’s Republic of China
| |
Collapse
|
31
|
Arteannuin-B and (3-Chlorophenyl)-2-Spiroisoxazoline Derivative Exhibit Anti-Inflammatory Effects in LPS-Activated RAW 264.7 Macrophages and BALB/c Mice-Induced Proinflammatory Responses via Downregulation of NF-κB/P38 MAPK Signaling. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27228068. [PMID: 36432169 PMCID: PMC9699497 DOI: 10.3390/molecules27228068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Host inflammatory responses are key to protection against injury; however, persistent inflammation is detrimental and contributes to morbidity and mortality. Herein, we demonstrated the anti-inflammatory role of Arteannuin-B (1) and its new spirocyclic-2-isoxazoline derivative JR-9 and their side effects in acute inflammatory condition in vivo using LPS-induced cytokines assay, carrageenan-induced paw edema, acetic acid-induced writhing and tail immersion. The results show that the spirocyclic-2-isoxazoline derivative is a potent anti-inflammatory agent with minimal cell toxicity as compared to Arteannuin-B. In addition, the efficacies of these compounds were also validated by flow cytometric, computational, and histopathological analysis. Our results show that the anti-inflammatory response of JR-9 significantly reduces the ability of mouse macrophages to produce NO, TNF-α, and IL-6 following LPS stimulation. Therefore, JR-9 is a prospective candidate for the development of anti-inflammatory drugs and its molecular mechanism is likely related to the regulation of NF-κB and MAPK signaling pathway.
Collapse
|
32
|
Li CH, Tsai ML, Chiou HY(C, Lin YC, Liao WT, Hung CH. Role of Macrophages in Air Pollution Exposure Related Asthma. Int J Mol Sci 2022; 23:ijms232012337. [PMID: 36293195 PMCID: PMC9603963 DOI: 10.3390/ijms232012337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/30/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction, bronchial hyper-responsiveness, and airway inflammation. The chronic inflammation of the airway is mediated by many cell types, cytokines, chemokines, and inflammatory mediators. Research suggests that exposure to air pollution has a negative impact on asthma outcomes in adult and pediatric populations. Air pollution is one of the greatest environmental risks to health, and it impacts the lungs' innate and adaptive defense systems. A major pollutant in the air is particulate matter (PM), a complex component composed of elemental carbon and heavy metals. According to the WHO, 99% of people live in air pollution where air quality levels are lower than the WHO air quality guidelines. This suggests that the effect of air pollution exposure on asthma is a crucial health issue worldwide. Macrophages are essential in recognizing and processing any inhaled foreign material, such as PM. Alveolar macrophages are one of the predominant cell types that process and remove inhaled PM by secreting proinflammatory mediators from the lung. This review focuses on macrophages and their role in orchestrating the inflammatory responses induced by exposure to air pollutants in asthma.
Collapse
Affiliation(s)
- Chung-Hsiang Li
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Mei-Lan Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Faculty of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hsin-Ying (Clair) Chiou
- Teaching and Research Center of Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| | - Yi-Ching Lin
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Doctoral Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wei-Ting Liao
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (W.-T.L.); or (C.-H.H.); Tel.: +886-7-312-1101 (ext. 2791) (W.-T.L.); +886-7-311-5140 (C.-H.H.); Fax: +886-7-312-5339 (W.-T.L.); +886-7-321-3931 (C.-H.H.)
| | - Chih-Hsing Hung
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
- Correspondence: (W.-T.L.); or (C.-H.H.); Tel.: +886-7-312-1101 (ext. 2791) (W.-T.L.); +886-7-311-5140 (C.-H.H.); Fax: +886-7-312-5339 (W.-T.L.); +886-7-321-3931 (C.-H.H.)
| |
Collapse
|
33
|
Li K, Zhang Q, Wang T, Rong R, Hu X, Zhang Y. Laboratory investigation of pollutant emissions and PM 2.5 toxicity of underground coal fires. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155537. [PMID: 35489495 DOI: 10.1016/j.scitotenv.2022.155537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 06/14/2023]
Abstract
Widespread underground coal fires (UCFs) release large amounts of pollutants, thus leading to air pollution and health impacts. However, this topic has not been widely investigated, especially regarding the potential health hazards. We quantified the pollutant emissions and analyzed the physicochemical properties of UCF PM2.5 in a laboratory study of coal smoldering under a simulated UCF background. The emission factors of CO2, CO, and PM2.5 were 2489 ± 35, 122 ± 9, 12.90 ± 1.79 g/kg, respectively. UCF PM2.5 are carbonaceous particles with varied morphology and complex composition, including heavy metals, silica and polycyclic aromatic hydrocarbons (PAHs). The main PAHs components were those with 2-4 rings. Benzoapyrene (BaP) and indeno[1,2, 3-cd]pyrene (IcdP) were important contributors to the carcinogenesis of these PAHs. We quantitatively evaluate the toxicity of inhaled UCF PM2.5 using a nasal inhalation exposure system. The target organs of UCF PM2.5 are lungs, liver, and kidneys. UCF PM2.5 presented an enriched chemical composition and induced inflammation and oxidative stress, which together mediated multiple organ injury. Long-term PM2.5 metabolism is the main cause of persistent toxicity, which might lead to long-term chronic diseases. Therefore, local authorities should recognize the importance and effects of UCF emissions, especially PM2.5, to establish control and mitigation measures.
Collapse
Affiliation(s)
- Kaili Li
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China
| | - Qixing Zhang
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China.
| | - Tong Wang
- Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory (HFIPS), Chinese Academy of Science, Hefei 230031, China; University of Science and Technology of China, Hefei 230026, China
| | - Rui Rong
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, China
| | - Xiaowen Hu
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yongming Zhang
- State Key Laboratory of Fire Science (SKLFS), University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
34
|
Ren H, Xia X, Dai X, Dai Y. The role of neuroplastin65 in macrophage against E. coli infection in mice. Mol Immunol 2022; 150:78-89. [PMID: 36007354 DOI: 10.1016/j.molimm.2022.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/20/2022] [Accepted: 08/04/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Innate immune response constitutes the first line of defense against pathogens. Inflammatory responses involve close contact between different populations of cells. These adhesive interactions mediate migration of cells to sites of infection leading the effective action of cells within the lesions. Cell adhesion molecules are critical to controlling immune response mediating cell adhesion or chemotaxis, as well as coordinating actin-based cell motility during phagocytosis and chemotaxis. Recently, a newly discovered neuroplastin (Np) adhesion molecule is found to play an important role in the nervous system. However, there is limited information on Np functions in immune response. To understand how Np is involved in innate immune response, a mouse model of intraperitoneal infection was established to investigate the effect of Np on macrophage-mediated clearance of E. coli infection and its possible molecular mechanisms. METHODS Specific deficiency mice with Nptn gene controlling Np65 isoform were employed in this study. The expression levels of mRNA and proteins were detected by qPCR and western blot, or evaluated by flow cytometry. The expression level of NO and ROS were measured with their specific indicators. Cell cycle and apoptosis were detected by specific detection kits. Acid phosphatase activity was measured by flow cytometry after labelling with LysoRed fluorescent probe. Bone marrow derived macrophages (BMDMs) were isolated from bone marrow of mice hind legs. Cell proliferation was detected by CCK8 assay. Cell migration was measured by wound healing assay or transwell assay. RESULTS The lethal dose of E. coli infection in Np65-/- mice dropped to the half of lethal dose in WT mice. The bacterial load in the spleen, kidney and liver from Np65-/- mice were significantly higher than that from WT mice, which were due to the dramatic reduction of NO and ROS production in phagocytes from Np65-/- mice. Np65 gene deficiency remarkably impaired phagocytosis and function of lysosome in macrophage. Furthermore, Np65 molecule was involved in maturation and proliferation, even in migration and chemotaxis of BMDM in vitro. CONCLUSION This study for the first time demonstrates that Np is involved in multi-function of phagocytes during bacterial infection, proposing that Np adhesion molecule plays a critical role in clearing pathogen infection in innate immunity.
Collapse
Affiliation(s)
- Huan Ren
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Xiaoxue Xia
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Xueting Dai
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China
| | - Yalei Dai
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, and Department of Immunology and Microbiology, Tongji University School of Medicine, 1239 Siping Road, Shanghai 200092, China.
| |
Collapse
|
35
|
Gene Expression Changes Induced by Exposure of RAW 264.7 Macrophages to Particulate Matter of Air Pollution: The Role of Endotoxins. Biomolecules 2022; 12:biom12081100. [PMID: 36008994 PMCID: PMC9405577 DOI: 10.3390/biom12081100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/05/2022] [Accepted: 08/07/2022] [Indexed: 11/17/2022] Open
Abstract
Despite the variable chemical and physical characteristics of particulate air pollutants, inflammation and oxidative stress have been identified as common mechanisms for cell damage and negative health influences. These effects are produced by organic components, especially by endotoxins. This study analyzed the gene expression profile after exposure of RAW 264.7 cells to the standard particulate matter (PM) material, NIST1648a, and PM with a reduced organic matter content, LAp120, in comparison to the effects of lipopolysaccharide (LPS). The selected parameters of cell viability, cell cycle progression, and metabolic and inflammatory activity were also investigated. Both forms of PM negatively influenced the parameters of cell activity. These results were generally reflected in the gene expression profile. Only NIST1648a, excluding LAp120, contained endotoxins and showed small but statistically significant pro-inflammatory activity. However, the gene expression profiling revealed strong pro-inflammatory cell activation induced by NIST1648a that was close to the effects of LPS. Changes in gene expression triggered by LAp120 were relatively small. The observed differences in the effects of NIST1648a and LAp120 were related to the content of organic matter in which bacterial endotoxins play an important role. However, other organic compounds and their interactions with other PM components also appear to be of significant importance.
Collapse
|
36
|
Zarnke A, Oliver C, Dorman S. McIntyre Powder and its potential contributions to cardiovascular disease risk: A literature review through the McIntyre Powder historical lens. Am J Ind Med 2022; 65:813-821. [PMID: 35863903 PMCID: PMC9541914 DOI: 10.1002/ajim.23415] [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: 12/22/2021] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022]
Abstract
McIntyre Powder (MP) is a fine aluminum powder that was developed to prevent silicosis in gold and uranium mine workers in Ontario, Canada, and was administered to miners there from 1943 to 1979. Mine workers were exposed to high concentrations (35.6 mg/m3) of MP for approximately 10 min before every work shift. Contemporary physical and chemical characterizations of this powder have revealed that 12% of the powder is in the ultrafine particle size‐range (nanoparticles); and the remaining 88%, in the fine particulate size range (below 2.5 µm in diameter). The confluence of ultrafine particulate (UFP) composition and high airborne concentration of MP would be expected to overwhelm the defense mechanisms of the lung and increase the lung dust burden of the mine worker exposed to respirable dust in the mine. Published studies revealing associations between air pollution particulates and increased risk for cardiovascular disease (CVD) shown a dose–response relationship with ambient PM2.5 and UFP and suggest that miners exposed to MP may also be at increased risk of CVD. The historical perspective of the use of MP in northern Ontario hard‐rock mines and its potential implications for CVD in exposed mine workers are discussed.
Collapse
Affiliation(s)
- Andrew Zarnke
- Laurentian University, Sudbury, Ontario, Canada.,The Occupational Health Clinics for Ontario Workers, Sudbury, Ontario, Canada.,The Centre for Research in Occupational Safety and Health (CROSH), Laurentian University, Sudbury, Ontario, Canada
| | - Christine Oliver
- The Occupational Health Clinics for Ontario Workers, Sudbury, Ontario, Canada.,Dalla Lana School of Public Health, Division of Occupational and Environmental Health, University of Toronto, Toronto, Ontario, Canada
| | - Sandra Dorman
- Laurentian University, Sudbury, Ontario, Canada.,The Centre for Research in Occupational Safety and Health (CROSH), Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
37
|
Effects of Particulate Matter on Inflammation and Thrombosis: Past Evidence for Future Prevention. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148771. [PMID: 35886623 PMCID: PMC9317970 DOI: 10.3390/ijerph19148771] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 02/04/2023]
Abstract
Ambient air pollution has become a common problem worldwide. Exposure to pollutant particles causes many health conditions, having a particular impact on pulmonary and cardiovascular disease. Increased understanding of the pathological processes related to these conditions may facilitate the prevention of the adverse impact of air pollution on our physical health. Evidence from in vitro, in vivo, and clinical studies has consistently shown that exposure to particulate matter could induce the inflammatory responses such as IL-6, TNF-α, IL-1β, as well as enhancing the oxidative stress. These result in vascular injury, adhesion molecule release, platelet activation, and thrombin generation, ultimately leading to a prothrombotic state. In this review, evidence on the effects of particulate matter on inflammation, oxidative stress, adhesion molecules, and coagulation pathways in enhancing the risk of thrombosis is comprehensively summarized and discussed. The currently available outcomes of interventional studies at a cellular level and clinical reports are also presented and discussed.
Collapse
|
38
|
Amin A, Hossen MJ, Fu XQ, Chou JY, Wu JY, Wang XQ, Chen YJ, Wu Y, Yin CL, Dou XB, Liang C, Chou GX, Yu ZL. Inhibition of the Akt/NF-κB pathway is involved in the anti-gastritis effects of an ethanolic extract of the rhizome of Atractylodes macrocephala. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115251. [PMID: 35381310 DOI: 10.1016/j.jep.2022.115251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gastritis can lead to ulcers and the development of gastric cancer. The rhizome of Atractylodes macrocephala Koidz. (Asteraceae), a traditional Chinese medicinal herb, is prescribed for the treatment of gastric disorders, hepatitis and rheumatism. Its bio-active compounds are considered to be particularly effective in this regard. However, the molecular processes of the herb's anti-inflammatory activity remain obscure. This study elucidates a mechanism upon which an ethanolic extract of this herb (Am-EE) exerts anti-inflammation effects in RAW264.7 macrophage cells (RAW cells) stimulated by lipopolysaccharide (LPS) treatment and HCl Ethanol-stimulated gastritis rats. AIM OF THE STUDY To investigate the anti-gastritis activities of Am-EE and explore the mode of action. MATERIALS AND METHODS Ethanol (95%) was used to prepare Am-EE. The quality of the extract was monitored by HPLC analysis. The in vivo effects of this extract were examined in an HCl Ethanol-stimulated gastritis rat model, while LPS-stimulated RAW cells were used for in vitro assays. Cell viability and nitric oxide (NO) production were observed by MTT and Griess assays. Real-time PCR was used to examine mRNA expression. The PGE2 ELISA kit was employed to detect prostaglandin E2 (PGE2). Enzyme activities and protein contents were examined by immunoblotting. Luciferase reporter gene assays (LRA) were employed to observe nuclear transcription factor (NF)-κB activity. The SPSS (SPSS Inc., Chicago, Illinois, United States) application was used for statistical examination. RESULTS HPLC analysis indicates that Am-EE contains atractylenolide-1 (AT-1, 1.33%, w/w) and atractylenolide-2 (AT-2, 1.25%, w/w) (Additional Figure. A1). Gastric tissue damage (induced by HCl Ethanol) was significantly decreased in SD rats following intra-gastric application of 35 mg/kg Am-EE. Indistinguishable to the anti-inflammation effects of 35 mg/kg ranitidine (gastric medication). Am-EE treatment also reduced LPS-mediated nitric oxide (NO) and prostaglandin E2 (PGE2) production. The mRNA and protein synthesis of inducible cyclooxygenase (COX)-2 and NO synthase (iNOS) was down-regulated following treatment in RAW cells. Am-EE decreased NF-κB (p50) nuclear protein levels and inhibited NF-κB-stimulated LRA activity in RAW cells. Lastly, Am-EE decreased the up-regulated levels of phosphorylated IκBα and Akt proteins in rat stomach lysates and in LPS challenged RAW cell samples. CONCLUSION Our study illustrates that Am-EE suppresses the Akt/IκBα/NF-κB pathway and exerts an anti-inflammatory effect. These novel conclusions provide a pharmacological basis for the clinical use of the A. macrocephala rhizome in the treatment and prevention of gastritis and gastric cancer.
Collapse
Affiliation(s)
- Aftab Amin
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Muhammad Jahangir Hossen
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Department of Animal Science, Patuakhali Science and Technology University, Dumki, Patuakhali, 8602, Bangladesh.
| | - Xiu-Qiong Fu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Ji-Yao Chou
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Jia-Ying Wu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Xiao-Qi Wang
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Ying-Jie Chen
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Ying Wu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Cheng-Le Yin
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Xiao-Bing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Chun Liang
- Division of Life Science, Center for Cancer Research and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China; EnKang Pharmaceuticals, Limited, Guangzhou, China.
| | - Gui-Xin Chou
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhi-Ling Yu
- Center for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China; Research and Development Center for Natural Health Products, HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
39
|
Polygonatum Polysaccharide Regulates Macrophage Polarization and Improves LPS-Induced Acute Lung Injury through TLR4-MAPK/NF-κB Pathway. Can Respir J 2022; 2022:2686992. [PMID: 35874106 PMCID: PMC9303503 DOI: 10.1155/2022/2686992] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
Objective To investigate the effects of polygonatum sibiricum polysaccharides (PSPs) on the polarization of macrophages to M1 and M2 phenotypes and their potential mechanism. Methods PSPs samples were prepared through water extraction and alcohol precipitation assay. The properties of PSPs were identified and analyzed by high-performance liquid chromatography, FT-IR, and NMR assay. Then, the effects of PSPs on mouse macrophage RAW264.7 viability were measured by CCK-8 assay. The cells were randomly divided into the control group, PSPs group, LPS group, and LPS + PSPs group. M1 phenotype polarization of RAW264.7 cells was induced by LPS treatment. The effects of various treatments on expression of M2 phenotype CD206, activation of TLR4-MAPK/NF-κB signal pathway, and translocation of NF-κB into the nucleus were determined by ELISA, western blot, and immunofluorescence assay, respectively. TLR4 inhibitor, TAK-242, and MAPK inhibitor, BIRB 796, were used to verify the effects of PSPs on the TLR4-MAPK/NF-κB pathway. The mice model of acute lung injury (ALI) was established and randomly divided into control group, PSPs group, LPS group, and LPS + PSPs group. Bronchoalveolar lavage fluid (BALF) and lung tissue were collected to measure protein, inflammatory cells, neutrophil and macrophage cells number, and the levels of IL-6 and TNF-α in BALF. Flow cytometry and western blot assay measured the phenotypic changes of macrophages and the activation of the TLR4-MAPK/NF-κB signaling pathway. Results The concentrations of PSPs lower than 100 μg/mL showed no toxicity to RAW264.7 cells. PSPs treatment could significantly reverse the reduction of CD206 protein expression (P < 0.05) and the increase of the expression of inflammatory factor TNF-α, IL-1β, and IL-6 (all P < 0.05), TLR4-MAPK/NF-κB signaling pathway activation (all P < 0.05), and NF-κB translocation into the nucleus induced by LPS. The effect of inhibitors TAK-242 and BIRB 796 was consistent with that of PSPs. In the mice model of ALI, PSPs treatment could reduce the total protein levels of BALF and the number of inflammatory cells level, reverse the number changes of neutrophils and macrophages, and downregulate the proinflammatory factors IL-6 and TNF-α caused by LPS (all P < 0.05). In addition, PSPs treatment could also significantly reverse the increase in the number of iNOS expressing macrophages in alveolar lavage fluid induced by LPS (P < 0.05). In contrast, CD206-expressed cells decreased (P < 0.05). PSPs could also reverse LPS-induced TLR4-MAPK/NF-κB signal pathway protein activation (all P < 0.05). Conclusion PSPs could suppress TLR4-MAPK/NF-κB activation induced by LPS, inhibit M1 phenotypic polarization of macrophages, and promote M2 phenotypic polarization, thus playing an anti-inflammatory role.
Collapse
|
40
|
Kumari S, Singh R. Protective effects of intranasal curcumin on silica-induced lung damage. Cytokine 2022; 157:155949. [PMID: 35764024 DOI: 10.1016/j.cyto.2022.155949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 06/09/2022] [Accepted: 06/18/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE Being anti-inflammatory and an antioxidant in nature, curcumin has been studied for its anti-asthmatic effects, but its impact on silicosis has not been investigated before. It is a form of occupational lung illness caused by inhaling crystalline silica. It is particularly common among those who work in construction-related sectors. Therefore, present study has been undertaken to investigate impact of intranasal curcumin on silica induced lung damage in mice model of silicosis. MATERIALS AND METHODS Mice model of silicosis was developed by intranasal silica instillation (2.5 mg/mice) for different durations mainly 7, 14 and 21 days, where the longest duration of silica exposure (21 days) mimics chronic occupational exposure of silica dust leading to silicosis. Curcumin (5 mg/kg,i.n) and /or dexamethasone, a known corticosteroid (10 mg/kg,i.p) was administered an hour prior to silica administration. RESULTS Present study revealed silica induced lung damage in the mice model of silicosis characterized by airway inflammation, collagen deposition and enhanced expression of fibrosis markers (MMP-9, α-SMA, Hydroxyproline), which were significantly reduced in curcumin treatment groups. Inhibitory effects of curcumin were compared with standard drug, dexamethasone, a corticosteroid and was found better in protecting structural alterations in the lung. Damaged and abnormal mitochondria (enlarged and irregular shapes) were observed in silicosis group which were reduced in curcumin and dexamethasone treatment groups as revealed in transmission electron microscopic studies. CONCLUSIONS Present study shows protective effects of intranasal curcumin on silica-induced airway inflammation and structural changes thereby lung damage. Hence, it can be considered as an alternative and complementary medication for silicosis.
Collapse
Affiliation(s)
- Sneha Kumari
- Department of Zoology, MMV Unit, Banaras Hindu University, Varanasi 221005, India
| | - Rashmi Singh
- Department of Zoology, MMV Unit, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
41
|
Lin S, Fu X, Luo M, Zhong WH. Tailoring bimodal protein fabrics for enhanced air filtration performance. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2022.120913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Upadhyay S, Chakraborty A, Thimraj TA, Baldi M, Steneholm A, Ganguly K, Gerde P, Ernstgård L, Palmberg L. Establishment of Repeated In Vitro Exposure System for Evaluating Pulmonary Toxicity of Representative Criteria Air Pollutants Using Advanced Bronchial Mucosa Models. TOXICS 2022; 10:toxics10060277. [PMID: 35736886 PMCID: PMC9228979 DOI: 10.3390/toxics10060277] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022]
Abstract
There is mounting evidence that shows the association between chronic exposure to air pollutants (particulate matter and gaseous) and onset of various respiratory impairments. However, the corresponding toxicological mechanisms of mixed exposure are poorly understood. Therefore, in this study, we aimed to establish a repeated exposure setting for evaluating the pulmonary toxicological effects of diesel exhaust particles (DEP), nitrogen dioxide (NO2), and sulfur dioxide (SO2) as representative criterial air pollutants. Single, combined (DEP with NO2 and SO2), and repeated exposures were performed using physiologically relevant human bronchial mucosa models developed at the air−liquid interface (bro-ALI). The bro-ALI models were generated using human primary bronchial epithelial cells (3−4 donors; 2 replicates per donor). The exposure regime included the following: 1. DEP (12.5 µg/cm2; 3 min/day, 3 days); 2. low gaseous (NO2: 0.1 ppm + SO2: 0.2 ppm); (30 min/day, 3 days); 3. high gaseous (NO2: 0.2 ppm + SO2: 0.4 ppm) (30 min/day, 3 days); and 4. single combined (DEP + low gaseous for 1 day). The markers for pro-inflammatory (IL8, IL6, NFKB, TNF), oxidative stress (HMOX1, GSTA1, SOD3,) and tissue injury/repair (MMP9, TIMP1) responses were assessed at transcriptional and/ or secreted protein levels following exposure. The corresponding sham-exposed samples under identical conditions served as the control. A non-parametric statistical analysis was performed and p < 0.05 was considered as significant. Repeated exposure to DEP and single combined (DEP + low gaseous) exposure showed significant alteration in the pro-inflammatory, oxidative stress and tissue injury responses compared to repeated exposures to gaseous air pollutants. The study demonstrates that it is feasible to predict the long-term effects of air pollutants using the above explained exposure system.
Collapse
Affiliation(s)
- Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
- Correspondence: (S.U.); (L.P.); Tel.:+46-85-2487930 (S.U.); +46-8-524-822-10 (L.P.)
| | - Ashesh Chakraborty
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Tania A. Thimraj
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Marialuisa Baldi
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | | | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Per Gerde
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Lena Ernstgård
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
| | - Lena Palmberg
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden; (A.C.); (T.A.T.); (M.B.); (K.G.); (P.G.); (L.E.)
- Correspondence: (S.U.); (L.P.); Tel.:+46-85-2487930 (S.U.); +46-8-524-822-10 (L.P.)
| |
Collapse
|
43
|
Chen X, Kim DI, Moon HG, Chu M, Lee K. Coconut Oil Alleviates the Oxidative Stress-Mediated Inflammatory Response via Regulating the MAPK Pathway in Particulate Matter-Stimulated Alveolar Macrophages. Molecules 2022; 27:molecules27092898. [PMID: 35566249 PMCID: PMC9105152 DOI: 10.3390/molecules27092898] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 11/16/2022] Open
Abstract
Exposure to particulate matter (PM) is related to various respiratory diseases, and this affects the respiratory immune system. Alveolar macrophages (AMs), which are defenders against pathogens, play a key role in respiratory inflammation through cytokine production and cellular interactions. Coconut oil demonstrates antioxidant and anti-inflammatory properties, and it is consumed worldwide for improved health. However, reports on the protective effects of coconut oil on the PM-induced respiratory immune system, especially in AMs, are limited. In this study, we generated artificial PM (APM) with a diameter approximately of 30 nm by controlling the temperature, and compared its cytotoxicity with diesel exhaust particles (DEP). We also investigated the antioxidant and anti-inflammatory effects of coconut oil in APM− and DEP−stimulated AMs, and the underlying molecular mechanisms. Our results showed that APM and DEP had high cytotoxicity in a dose-dependent manner in AMs. In particular, APM or DEP at 100 μg/mL significantly decreased cell viability (p < 0.05) and significantly increased oxidative stress markers such as reactive oxygen species (p < 0.01); the GSSH/GSH ratio (p < 0.01); and cytokine production, such as tumor necrosis factor-α (p < 0.001), interleukin (IL)-1β (p < 0.001), and IL-6 (p < 0.001). The expression of the genes for chemokine (C-X-C motif) ligand-1 (p < 0.05) and monocyte chemoattractant protein-1 (p < 0.001); and the proteins toll-like receptor (TLR) 4 (p < 0.01), mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (p < 0.001), p38 (p < 0.001); and extracellular receptor-activated kinase (p < 0.001), were also upregulated by PM. These parameters were reversed upon treatment with coconut oil in APM− or DEP−stimulated AMs. In conclusion, coconut oil can reduce APM− or DEP−induced inflammation by regulating the TLR4/MAPK pathway in AMs, and it may protect against adverse respiratory effects caused by PM exposure.
Collapse
Affiliation(s)
- Xinyu Chen
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup-si 56212, Korea; (X.C.); (D.I.K.); (H.-G.M.)
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Korea
| | - Dong Im Kim
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup-si 56212, Korea; (X.C.); (D.I.K.); (H.-G.M.)
| | - Hi-Gyu Moon
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup-si 56212, Korea; (X.C.); (D.I.K.); (H.-G.M.)
| | - Minchul Chu
- Greensol Co., Ltd., 89-26, Jimok-ro, Paju-si 10880, Korea;
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup-si 56212, Korea; (X.C.); (D.I.K.); (H.-G.M.)
- Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon 34113, Korea
- Correspondence: or ; Tel.: +82-63-570-8740
| |
Collapse
|
44
|
Xie G, Yue J, Yang W, Yang L, Xu M, Sun L, Zhang B, Guo L, Chung MC. Effects of PM 2.5 and its constituents on hemoglobin during the third trimester in pregnant women. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:35193-35203. [PMID: 35060058 PMCID: PMC9076737 DOI: 10.1007/s11356-022-18693-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
Anemia has been a public health issue evoking global concern, and the low hemoglobin (Hb) concentration links to adverse pregnancy outcomes. However, the associations of PM2.5 and its constituents with Hb and anemia in pregnant women remain unclear. In this retrospective birth cohort study, 7932 pregnant women who delivered in the First Affiliated Hospital of Xi'an Jiaotong University from 2015 to 2018 were included. The Hb during the third trimester in pregnant women was assessed before delivery. PM2.5 and its constituents (BC, NH4+, NO3-, OM, SO42-, and Dust) during pregnancy were retrieved from the V4.CH.03 product constructed by the Atmospheric Composition Analysis Group. Generalized linear regression model was applied to investigate the effects of PM2.5 and its constituents on Hb and anemia during the third trimester in pregnant women. The means and standard deviations of PM2.5, BC, NH4+, NO3-, OM, SO42-, and Dust were 69.56 (15.24), 10.02 (2.72), 8.11 (1.77), 14.96 (5.42), 15.36 (4.11), 10.08 (1.20), and 10.98 (1.85) μg/m3, respectively. Per IQR increase (μg/m3) of PM2.5, BC, NO3-, and OM linked to - 0.75 (- 1.50, - 0.01), - 0.85 (- 1.65, - 0.04), - 0.79 (- 1.56, - 0.03), and - 0.73 (- 1.44, - 0.03) g/L decrease of Hb during the third trimester in multiparous pregnant women, but not for NH4+, SO42-, Dust, and primiparous pregnant women. PM2.5 and its constituents had no significant association with anemia, except for Dust (OR: 0.90, 95% CI: 0.82, 0.99, per IQR increase) in primiparous pregnant women. Besides, SO42- was of lag effects on Hb and anemia in multiparous pregnant women. Moreover, non-linear associations were found among PM2.5 and its constituents, Hb, and anemia. Therefore, exposure to PM2.5 and some constituents of PM2.5 was associated with reduced Hb level during the third trimester in multiparous pregnant women. Related departments and pregnant women should take targeted actions to eliminate the detrimental effects of PM2.5 and its constituents on pregnancy outcomes.
Collapse
Affiliation(s)
- Guilan Xie
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, People's Republic of China
| | - Jie Yue
- Department of Pediatrics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Wenfang Yang
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China.
| | - Liren Yang
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, People's Republic of China
| | - Mengmeng Xu
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
| | - Landi Sun
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, People's Republic of China
| | - Boxing Zhang
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, People's Republic of China
| | - Leqian Guo
- Department of Obstetrics and Gynecology, Maternal & Child Health Center, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Shaanxi Province, 710061, Xi'an, People's Republic of China
| | - Mei Chun Chung
- Division of Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
45
|
Gandomani EA, Mosaffa N, Zendehdel R, Kohneshahri MH, Vahabi M, Sabour S. Release of Interleukin-1β evaluation among mineral oil mist–exposed workers. Toxicol Ind Health 2022; 38:270-276. [DOI: 10.1177/07482337221090708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure to aerosols has been found to be linked to respiratory impairment. Although the effects of both indoor and outdoor exposures to particulates have been extensively reported, exposures to mists are less studied. Herein, we reported a survey of mineral oil mist toxicity in an occupational exposure scenario. For the purpose of this study, 65 lathe workers of the metal processing industry, as mineral oil mist–exposed population, were studied. Thereafter, the participants’ age, smoking habits and work experience were matched with those of the control workers ( n = 65) who were not occupationally exposed to mist. Thereafter, air samples were evaluated from the breathing zone of the workers using NIOSH method 5026. Plasma Interleukin-1β as a pro-inflammatory indicator was assessed in all the studied subjects. Mean ± standard deviation of mineral oil mist time-weighted average exposure in lathe workers was 7.10± 3.49 mg/m3. IL-1β cytokine levels were significantly higher in the lathe groups compared to the control group. The mean level of Interleukin-1β in the control subjects (2922 pg/L) was selected as the cut-off point of the inflammation effect. Based on this pro-inflammatory point, the results of monitoring showed that 60% of the exposed were affected. A Spearman correlation was also found between mineral oil mist exposure and inflammation in the affected subjects. Our findings highlighted the immunological potential of mineral oil mist in occupational exposure. Overall, the results of this study suggested that Interleukin-1β evaluation in mineral oil mist exposure could be considered as both an acute and chronic inflammation marker.
Collapse
Affiliation(s)
- Elham A Gandomani
- Department of Occupational Health and Safety Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Zendehdel
- Department of Occupational Health and Safety Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad H Kohneshahri
- Department of Occupational Health and Safety Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoomeh Vahabi
- Department of Occupational Health and Safety Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Sabour
- Department of Epidemiology, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Exploring the Potential Effects and Mechanisms of Asarum sieboldii Radix Essential Oil for Treatment of Asthma. Pharmaceutics 2022; 14:pharmaceutics14030558. [PMID: 35335934 PMCID: PMC8953372 DOI: 10.3390/pharmaceutics14030558] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Asthma, a common chronic pulmonary disorder characterized by airway remodeling, hyperresponsiveness and obstruction, can be aggravated by repeated exposure to particulate matter (PM). The potential effect and mechanisms of Asarum sieboldii Radix essential oil (AEO) against asthma were explored based on network pharmacology. AEO was pre-treated using a nebulizer for 3 weeks and the mice were sensitized to ovalbumin (OVA) and PM10 with the co-treatment of AEO for 4 weeks. In addition, A549 lung epithelial cells were sensitized with PM10 to investigate the underlying mechanisms of AEO regarding the lung-fibrosis-related mediators. The target genes of methyl eugenol, a main compound of AEO, were highly matched by 48% with the gene set of asthma. AEO markedly inhibited the increase in epithelial thickness through the accumulation of goblet cells in the airways. Collagen deposition in the lung tissues of OVA+PM10-challenged asthmatic mice was significantly decreased by AEO. AEO also inhibited the influx of inflammatory cells in the bronchoalveolar lavage fluid, as well as the increases in serum IgE and IgG2a and cytokines in the lung tissues. Furthermore, AEO regulated the expressions of fibrotic mediators, especially POSTN and TGF-β. In conclusion, we expect that AEO can be one of the effective alternative therapeutics to relieve asthma.
Collapse
|
47
|
Marchini T, Magnani N, Garces M, Kelly J, Paz M, Caceres L, Calabro V, Lasagni Vitar R, Caltana L, Contin M, Reynoso S, Lago N, Vico T, Vanasco V, Wolf D, Tripodi V, Gonzalez Maglio D, Alvarez S, Buchholz B, Berra A, Gelpi R, Evelson P. Chronic exposure to polluted urban air aggravates myocardial infarction by impaired cardiac mitochondrial function and dynamics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 295:118677. [PMID: 34906594 DOI: 10.1016/j.envpol.2021.118677] [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: 07/17/2021] [Revised: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
Air pollution exposure positively correlates with increased cardiovascular morbidity and mortality rates, mainly due to myocardial infarction (MI). Herein, we aimed to study the metabolic mechanisms underlying this association, focusing on the evaluation of cardiac mitochondrial function and dynamics, together with its impact over MI progression. An initial time course study was performed in BALB/c mice breathing filtered air (FA) or urban air (UA) in whole-body exposure chambers located in Buenos Aires City downtown for up to 16 weeks (n = 8 per group and time point). After 12 weeks, lung inflammatory cell recruitment was evident in UA-exposed mice. Interestingly, impaired redox metabolism, characterized by decreased lung SOD activity and increased GSSG levels and NOX activity, precede local inflammation in this group. At this selected time point, additional mice were exposed to FA or UA (n = 12 per group) and alveolar macrophage PM uptake and nitric oxide (NO) production was observed in UA-exposed mice, together with increased pro-inflammatory cytokine levels (TNF-α and IL-6) in BAL and plasma. Consequently, impaired heart tissue oxygen metabolism and altered mitochondrial ultrastructure and function were observed in UA-exposed mice after 12 weeks, characterized by decreased active state respiration and ATP production rates, and enhanced mitochondrial H2O2 production. Moreover, disturbed cardiac mitochondrial dynamics was detected in this group. This scenario led to a significant increase in the area of infarcted tissue following myocardial ischemia reperfusion injury in vivo, from 43 ± 3% of the area at risk in mice breathing FA to 66 ± 4% in UA-exposed mice (n = 6 per group, p < 0.01), together with a sustained increase in LVEDP during myocardial reperfusion. Taken together, our data unravel cardiac mitochondrial mechanisms that contribute to the understanding of the adverse health effects of urban air pollution exposure, and ultimately highlight the importance of considering environmental factors in the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Mariana Garces
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Jazmin Kelly
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Mariela Paz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Lourdes Caceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Valeria Calabro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Romina Lasagni Vitar
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Laura Caltana
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias (IBCN), Buenos Aires, C1121ABG, Argentina
| | - Mario Contin
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Sofia Reynoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Nestor Lago
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Tamara Vico
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Dennis Wolf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Valeria Tripodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Daniel Gonzalez Maglio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Silvia Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Bruno Buchholz
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Alejandro Berra
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Ricardo Gelpi
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina.
| |
Collapse
|
48
|
Firdous P, Nissar K, Bashir H, Hussain QA, Masoodi SR, Ganai BA. Environmental Factors as Diabetic Mediators: A Mechanistic Approach. Curr Diabetes Rev 2022; 18:e301221199656. [PMID: 34967298 DOI: 10.2174/1573399818666211230104327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/13/2021] [Accepted: 09/29/2021] [Indexed: 11/22/2022]
Abstract
Despite substantial investment in research and treatment options, diabetes mellitus remains a pressing public health concern with potential epidemic proportions globally. There are reports that by the end of 2040, 642 million people will be suffering from diabetes. Also, according to an estimation, 1.6 million deaths were caused directly by diabetes in 2016. Diabetes is a metabolic disorder characterized by impaired glucose regulation in the body due to the destruction of pancreatic β-cells or insulin resistance. Genetic propensity, unhealthy and imbalanced diet, obesity and increasing urbanization are the common risk factors for diabetes. Besides this, it has been reported that environmental pollutants like organic pesticides, heavy metals, and air pollutants act as strong predisposing factors for diabetes owing to their highly bio-accumulative nature. These pollutants disturb glucose homeostasis either by up-regulating or down-regulating the expression of diabetic marker genes like insulin (INS) and glucokinase (GCK). Unfortunately, the molecular mechanism of the role of pollutants in causing diabetes is not very clear. This mechanistic review provides evidence of different environmental determinants, including persistent organic pollutants (POPs), air pollutants, toxic metals, etc., in inducing diabetes and proposes a framework for the possible mechanisms involved. It also illuminates the current status and future challenges, which will not only broaden our understanding but can also be a reasonable platform for further investigation.
Collapse
Affiliation(s)
- Parveena Firdous
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Kamran Nissar
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
- Department of Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Humayra Bashir
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| | - Qazi A Hussain
- P.G. Department of Environmental Science, Sri Pratap College Campus, Cluster University Srinagar, Jammu and Kashmir 190001, India
| | | | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, Jammu and Kashmir 190006, India
| |
Collapse
|
49
|
Berman R, Rose CS, Downey GP, Day BJ, Chu HW. Role of Particulate Matter from Afghanistan and Iraq in Deployment-Related Lung Disease. Chem Res Toxicol 2021; 34:2408-2423. [PMID: 34808040 DOI: 10.1021/acs.chemrestox.1c00090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Approximately 3 million United States military personnel and contractors were deployed to Southwest Asia and Afghanistan over the past two decades. After returning to the United States, many developed persistent respiratory symptoms, including those due to asthma, rhinosinusitis, bronchiolitis, and others, which we collectively refer to as deployment-related lung diseases (DRLD). The mechanisms of different DRLD have not been well defined. Limited studies from us and others suggest that multiple factors and biological signaling pathways contribute to the onset of DRLD. These include, but are not limited to, exposures to high levels of particulate matter (PM) from sandstorms, burn pit combustion products, improvised explosive devices, and diesel exhaust particles. Once inhaled, these hazardous substances can activate lung immune and structural cells to initiate numerous cell-signaling pathways such as oxidative stress, Toll-like receptors, and cytokine-driven cell injury (e.g., interleukin-33). These biological events may lead to a pro-inflammatory response and airway hyperresponsiveness. Additionally, exposures to PM and other environmental hazards may predispose military personnel and contractors to more severe disease due to the interactions of those hazardous materials with subsequent exposures to allergens and cigarette smoke. Understanding how airborne exposures during deployment contribute to DRLD may identify effective targets to alleviate respiratory diseases and improve quality of life in veterans and active duty military personnel.
Collapse
Affiliation(s)
- Reena Berman
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Cecile S Rose
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Gregory P Downey
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Brian J Day
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, 1400 Jackson Street, Denver, Colorado 80206, United States
| |
Collapse
|
50
|
Lin CC, Law BF, Hettick JM. MicroRNA-mediated calcineurin signaling activation induces CCL2, CCL3, CCL5, IL8, and chemotactic activities in 4,4'-methylene diphenyl diisocyanate exposed macrophages. Xenobiotica 2021; 51:1436-1452. [PMID: 34775880 DOI: 10.1080/00498254.2021.2005851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Occupational exposure to 4,4'-methylene diphenyl diisocyanate (MDI), the most widely used monomeric diisocyanate, is one of the leading causes of occupational asthma (OA). Previously, we identified microRNA (miR)-206-3p/miR-381-3p-mediated PPP3CA/calcineurin signalling regulated iNOS transcription in macrophages and bronchoalveolar lavage cells (BALCs) after acute MDI exposure; however, whether PPP3CA/calcineurin signalling participates in regulation of other asthma-associated mediators secreted by macrophages/BALCs after MDI exposure is unknown.Several asthma-associated, macrophage-secreted mediator mRNAs from MDI exposed murine BALCs and MDI-glutathione (GSH) conjugate treated differentiated THP-1 macrophages were analysed using RT-qPCR.Endogenous IL1B, TNF, CCL2, CCL3, CCL5, and TGFB1 were upregulated in MDI or MDI-GSH conjugate exposed BALCs and macrophages, respectively. Calcineurin inhibitor tacrolimus (FK506) attenuated the MDI-GSH conjugate-mediated induction of CCL2, CCL3, CCL5, and CXCL8/IL8 but not others. Transfection of either miR-inhibitor-206-3p or miR-inhibitor-381-3p in macrophages induced chemokine CCL2, CCL3, CCL5, and CXCL8 transcription, whereas FK506 attenuated the miR-inhibitor-206-3p or miR-inhibitor-381-3p-mediated effects. Finally, MDI-GSH conjugate treated macrophages showed increased chemotactic ability to various immune cells, which may be attenuated by FK506.In conclusion, these results indicate that MDI exposure to macrophages/BALCs may recruit immune cells into the airway via induction of chemokines by miR-206-3p and miR-381-3p-mediated calcineurin signalling activation.
Collapse
Affiliation(s)
- Chen-Chung Lin
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Brandon F Law
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Justin M Hettick
- Allergy and Clinical Immunology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|