[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 

Oxidative Stress Induced by Air Pollution, 2nd Edition

A special issue of Antioxidants (ISSN 2076-3921). This special issue belongs to the section "Health Outcomes of Antioxidants and Oxidative Stress".

Deadline for manuscript submissions: 15 March 2025 | Viewed by 3789

Special Issue Editor


E-Mail Website
Guest Editor
Department of Immunology and Parasitology, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan
Interests: IL-1 signaling; NF-κB; particulate matter; neutrophil
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Over the recent decades, rapid industrialization and urbanization have significantly increased the levels of air pollution, presenting a multifaceted challenge to global public health. One of the critical consequences of exposure to various air pollutants is the induction of oxidative stress, a complex imbalance between the production of reactive oxygen species (ROS) and the body's ability to detoxify or repair the resulting damage. This phenomenon has emerged as a pivotal link between air pollution and a myriad of adverse health outcomes, ranging from respiratory diseases to cardiovascular disorders and beyond.

Building upon the fruitful first edition, this second edition will further unravel the complex interplay between air pollution and oxidative stress. By assembling a collection of cutting-edge studies, this Special Issue seeks to deepen our understanding of the intricate pathways through which air pollutants contribute to oxidative stress and, consequently, influence the development and progression of various diseases.

Dr. Yasuhiro Yoshida
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Antioxidants is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • particulate matter
  • air pollution
  • oxidative stress
  • reactive oxygen species
  • inflammation

Benefits of Publishing in a Special Issue

  • Ease of navigation: Grouping papers by topic helps scholars navigate broad scope journals more efficiently.
  • Greater discoverability: Special Issues support the reach and impact of scientific research. Articles in Special Issues are more discoverable and cited more frequently.
  • Expansion of research network: Special Issues facilitate connections among authors, fostering scientific collaborations.
  • External promotion: Articles in Special Issues are often promoted through the journal's social media, increasing their visibility.
  • e-Book format: Special Issues with more than 10 articles can be published as dedicated e-books, ensuring wide and rapid dissemination.

Further information on MDPI's Special Issue polices can be found here.

Published Papers (4 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

21 pages, 4339 KiB  
Article
A Combined Extract from Dioscorea bulbifera and Zingiber officinale Mitigates PM2.5-Induced Respiratory Damage by NF-κB/TGF-β1 Pathway
by In Young Kim, Hyo Lim Lee, Hye Ji Choi, Yeong Hyeon Ju, Yu Mi Heo, Hwa Rang Na, Dong Yeol Lee, Won Min Jeong and Ho Jin Heo
Antioxidants 2024, 13(12), 1572; https://doi.org/10.3390/antiox13121572 - 20 Dec 2024
Viewed by 514
Abstract
This research evaluated the protective role of a combined extract of Dioscorea bulbifera and Zingiber officinale (DBZO) against respiratory dysfunction caused by particulate matter (PM2.5) exposure in BALB/c mice. The bioactive compounds identified in the DBZO are catechin, astragalin, 6-gingerol, 8-gingerol, [...] Read more.
This research evaluated the protective role of a combined extract of Dioscorea bulbifera and Zingiber officinale (DBZO) against respiratory dysfunction caused by particulate matter (PM2.5) exposure in BALB/c mice. The bioactive compounds identified in the DBZO are catechin, astragalin, 6-gingerol, 8-gingerol, and 6-shogaol. DBZO ameliorated cell viability and reactive oxygen species (ROS) production in PM2.5-stimulated A549 and RPMI 2650 cells. In addition, it significantly alleviated respiratory dysfunction in BALB/c mice exposed to PM2.5. DBZO improved the antioxidant systems in lung tissues by modulating malondialdehyde (MDA) content, as well as levels of reduced glutathione (GSH) and superoxide dismutase (SOD). Likewise, DBZO restored mitochondrial dysfunction by improving ROS levels, mitochondrial membrane potential, and ATP production. Moreover, DBZO modulated the levels of neutrophils, eosinophils, monocytes, and lymphocytes (specifically CD4+, CD8+, and CD4+IL-4+ T cells) in blood and IgE levels in serum. DBZO was shown to regulate the c-Jun N-terminal kinase (JNK) pathway, nuclear factor kappa B (NF-κB) pathway, and transforming growth factor β (TGF-β)/suppressor of mothers against decapentaplegic (Smad) pathway. Histopathological observation indicated that DBZO mitigates the increase in alveolar septal thickness. These findings indicate that DBZO is a promising natural agent for improving respiratory health. Full article
(This article belongs to the Special Issue Oxidative Stress Induced by Air Pollution, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS) chromatogram of a combined extract from <span class="html-italic">Dioscorea bulbifera</span> and <span class="html-italic">Zingiber officinale</span> (DBZO).</p>
Full article ">Figure 2
<p>Effects of DBZO of particulate matter (PM<sub>2.5</sub>)-stimulated A549 and RPMI 2650 cells. Cell viability in (<b>a</b>) A549 and (<b>b</b>) RPMI 2650 cells and intracellular oxidative stress levels in (<b>c</b>) A549 and (<b>d</b>) RPMI 2650 cells. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 3
<p>Effects of DBZO on antioxidant system in the lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Malondialdehyde (MDA) contents, (<b>b</b>) reduced glutathione (GSH) levels, and (<b>c</b>) superoxide dismutase (SOD) levels. The results are presented as mean ± SD (<span class="html-italic">n</span> = 5). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 4
<p>Effects of DBZO on mitochondrial function in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Mitochondrial reactive oxygen species (ROS) production, (<b>b</b>) mitochondrial membrane potential, and (<b>c</b>) mitochondrial ATP content. The results are presented as mean ± SD (<span class="html-italic">n</span> = 4). Data are statistically represented with * = significantly different from the NC group, and # = significantly different from the PM<sub>2.5</sub> group; * and # = <span class="html-italic">p</span> &lt; 0.05 and ** and ## = <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Effects of DBZO on inflammatory cells in whole blood and immunoglobulin E (IgE) levels in serum of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Flow cytometry plots, frequency of (<b>b</b>) CD3<sup>+</sup>CD4<sup>+</sup> T cells, (<b>c</b>) CD3<sup>+</sup>CD8<sup>+</sup> T cells, (<b>d</b>) CD3<sup>+</sup>CD4<sup>+</sup>IL-4<sup>+</sup> T cells in whole blood, and (<b>e</b>) IgE levels in serum. The results are presented as mean ± SD (<b>b</b>–<b>d</b>, <span class="html-italic">n</span> = 5; e, <span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 6
<p>Effects of DBZO on alveolar size in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Histopathological sections and (<b>b</b>) alveolar area. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 7
<p>Effects of DBZO on inflammation-related protein expression levels in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Western blot images, protein expression levels of (<b>b</b>) IL-33, (<b>c</b>) MyD88, (<b>d</b>) p-IκB-α, (<b>e</b>) p-NF-κB, (<b>f</b>) IL-1β, and (<b>g</b>) TNF-α. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 8
<p>Effects of DBZO on apoptosis-related protein expression levels in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Western blot images, protein expression levels of (<b>b</b>) p-JNK, (<b>c</b>) BCl-2, (<b>d</b>) BAX, (<b>e</b>) BAX/BCl-2 ratio, and (<b>f</b>) caspase-3. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 8 Cont.
<p>Effects of DBZO on apoptosis-related protein expression levels in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Western blot images, protein expression levels of (<b>b</b>) p-JNK, (<b>c</b>) BCl-2, (<b>d</b>) BAX, (<b>e</b>) BAX/BCl-2 ratio, and (<b>f</b>) caspase-3. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">Figure 9
<p>Effects of DBZO on pulmonary fibrosis-related protein expression levels in lung tissues of PM<sub>2.5</sub>-exposed BALB/c mice. (<b>a</b>) Western blot images, protein expression levels of (<b>b</b>) TGF-β1, (<b>c</b>) p-Smad2, (<b>d</b>) p-Samd3, (<b>e</b>) MMP-2, and (<b>f</b>) MMP-9. The results are presented as mean ± SD (<span class="html-italic">n</span> = 3). Data were statistically considered at <span class="html-italic">p</span> &lt; 0.05, and different small letters represent the statistical differences.</p>
Full article ">
14 pages, 1240 KiB  
Article
Association Between Oxidative Potential of Particulate Matter Collected by Personal Samplers and Systemic Inflammation Among Asthmatic and Non-Asthmatic Adults
by Miguel Santibáñez, Juan José Ruiz-Cubillán, Andrea Expósito, Juan Agüero, Juan Luis García-Rivero, Beatriz Abascal, Carlos Antonio Amado, Laura Ruiz-Azcona, Marcos Lopez-Hoyos, Juan Irure, Yolanda Robles, Ana Berja, Esther Barreiro, Adriana Núñez-Robainas, José Manuel Cifrián and Ignacio Fernandez-Olmo
Antioxidants 2024, 13(12), 1464; https://doi.org/10.3390/antiox13121464 - 28 Nov 2024
Viewed by 621
Abstract
With the rationale that the oxidative potential of particulate matter (PM-OP) may induce oxidative stress and inflammation, we conducted the ASTHMA-FENOP study in which 44 asthmatic patients and 37 matched controls wore a personal sampler for 24 h, allowing the collection of fine [...] Read more.
With the rationale that the oxidative potential of particulate matter (PM-OP) may induce oxidative stress and inflammation, we conducted the ASTHMA-FENOP study in which 44 asthmatic patients and 37 matched controls wore a personal sampler for 24 h, allowing the collection of fine and coarse PM fractions separately, to determine PM-OP by the dithiothreitol (DTT) and ascorbic acid (AA) methods. The levels of Interleukin 6 (IL-6) and the IL-6/IL-10 ratio, as indicators of pro- and anti-inflammatory statuses, were determined by calculating the mean differences (MDs), odds ratios (ORs) and p-trends adjusted for sex, age, study level and body mass index. Positive associations for IL-6 levels in the form of adjusted MDs and ORs were obtained for all PM-OP metrics, reaching statistical significance for both OP-DTT and OP-AA in the fine fraction, with adjusted OR = 5.66; 95%CI (1.46 to 21.92) and 3.32; 95%CI (1.07 to 10.35), respectively, along with statistically significant dose–response patterns when restricting to asthma and adjusted also for clinical variables (adjusted p-trend = 0.029 and 0.01). Similar or stronger associations and dose–response patterns were found for the IL-6/IL-10 ratio. In conclusion, our findings on the effect of PM-OP on systemic inflammation support that asthma is a heterogeneous disease at the molecular level, with PM-OP potentially playing an important role. Full article
(This article belongs to the Special Issue Oxidative Stress Induced by Air Pollution, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Forest plot of crude and adjusted odds ratios (ORs) between higher values of OP-DTT and OP-AA (for the fine and coarse PM fractions); and higher IL-6 levels (on the <b>left</b>) and higher IL-6/IL-10 ratio levels (on the <b>right</b>). ORs adjusted for age, sex, educational level, BMI according to WHO classification, and FeNO levels.</p>
Full article ">Figure 2
<p>Forest plot of crude and adjusted mean differences (aMDs) between higher values of OP-DTT and OP-AA (for the fine and coarse PM fractions); and IL-6 (on the <b>left</b>) and the IL-6/IL-10 ratio levels (on the <b>right</b>). MDs adjusted for age, sex, educational level, BMI according to WHO classification, and FeNO levels.</p>
Full article ">
17 pages, 7392 KiB  
Article
Photobiomodulation Mitigates PM2.5-Exacerbated Pathologies in a Mouse Model of Allergic Asthma
by Jisu Park, Bo-Young Kim, Eun Jung Park, Yong-Il Shin and Ji Hyeon Ryu
Antioxidants 2024, 13(8), 1003; https://doi.org/10.3390/antiox13081003 - 19 Aug 2024
Cited by 2 | Viewed by 1546
Abstract
Exposure to particulate matter (PM), especially PM2.5, is known to exacerbate asthma, posing a significant public health risk. This study investigated the asthma-reducing effects of photobiomodulation (PBM) in a mice model mimicking allergic airway inflammation exacerbated by PM2.5 exposure. The [...] Read more.
Exposure to particulate matter (PM), especially PM2.5, is known to exacerbate asthma, posing a significant public health risk. This study investigated the asthma-reducing effects of photobiomodulation (PBM) in a mice model mimicking allergic airway inflammation exacerbated by PM2.5 exposure. The mice received sensitization with ovalbumin (OVA) and were subsequently treated with PM2.5 at a dose of 0.1 mg/kg every 3 days, for 9 times over 3 weeks during the challenge. PBM, using a 610 nm wavelength LED, was applied at 1.7 mW/cm2 to the respiratory tract via direct skin contact for 20 min daily for 19 days. Results showed that PBM significantly reduced airway hyperresponsiveness, plasma immunoglobulin E (IgE) and OVA-specific IgE, airway inflammation, T-helper type 2 cytokine, histamine and tryptase in bronchoalveolar lavage fluid (BALF), and goblet cell hyperplasia in PM2.5-exposed asthmatic mice. Moreover, PBM alleviated subepithelial fibrosis by reducing collagen deposition, airway smooth muscle mass, and expression of fibrosis-related genes. It mitigated reactive oxygen species generation, oxidative stress, endoplasmic reticulum stress, apoptotic cell death, ferroptosis, and modulated autophagic signals in the asthmatic mice exposed to PM2.5. These findings suggest that PBM could be a promising intervention for PM2.5-induced respiratory complications in patients with allergic asthma. Full article
(This article belongs to the Special Issue Oxidative Stress Induced by Air Pollution, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Inhibitory effects of PBM on the induction of airway hyperresponsiveness (AHR) and plasma IgE in a PM<sub>2.5</sub>-exposed asthma exacerbation model. (<b>A</b>) Establishment of an allergic asthma exacerbation mouse model induced by PM<sub>2.5</sub> exposure. A timeline describing the asthma exacerbation model induction and PBM treatment. (<b>B</b>) Measurement of body weight, thymus-to-body-weight ratio, and spleen-to-body-weight ratio on the final day of the experiment. (<b>C</b>) Assessment of AHR to methacholine (MCh) at concentrations of 25 and 50 mg/mL. (<b>D</b>) Measurement of total immunoglobulin E (IgE) and ovalbumin (OVA)-specific IgE in plasma. Data are shown as the mean ± SEM (<span class="html-italic">n</span> = 8). <span class="html-italic">* p</span> &lt; 0.05 compared with control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA. i.n., intranasal injection; i.p., intraperitoneal injection; BW, body weight; PM, particulate matter; OVA, ovalbumin; PBM, photobiomodulation; DEX, dexamethasone.</p>
Full article ">Figure 2
<p>Inhibitory effects of PBM on the elevation of allergic airway inflammation and goblet cell metaplasia in a PM<sub>2.5</sub>-exposed asthma exacerbation model. (<b>A</b>) Measurement of total and differential inflammatory cell counts (macrophage, neutrophils, lymphocytes, and eosinophils) in bronchoalveolar lavage fluid (BALF). (<b>B</b>) Evaluation of Th2 cytokines including interleukin (IL)-4, IL-5, and IL-13 in BALF. (<b>C</b>) Assessment of histamine and mast cell tryptase in BALF. (<b>D</b>) Representative images of H&amp;E staining revealed the infiltration of inflammatory cells in lung tissues. Scale bar represents 200 µm (up) and 100 µm (down). The bar graphs represent the summarized score of inflammation. (<b>E</b>) Goblet cells secreting mucus in lung tissues were identified using PAS staining. The bar graphs represent the number of PAS-reactive airway epithelial cells. Scale bar represents 50 µm. The bar graphs represent the summarized scores of PAS-positive mucus-producing cells. Data are shown as the mean ± SEM (<span class="html-italic">n</span> = 8). <span class="html-italic">* p</span> &lt; 0.05 compared with control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA. <sup>‡</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA + PBM.</p>
Full article ">Figure 3
<p>Inhibitory effects of PBM on the elevation of subepithelial fibrosis in a PM<sub>2.5</sub>-exposed asthma exacerbation model. Representative histological images showing (<b>A</b>) lung collagen fiber (Masson’s trichrome staining) and (<b>B</b>) collagen deposition (Sirius Red staining) in the lung tissues. Scale bar represents 100 µm. The bar graphs represent the summarized scores of collagen fiber deposition (<span class="html-italic">n</span> = 8). (<b>C</b>) Representative images of α-smooth muscle actin (α-SMA) and FITC expression, as determined by immunohistochemistry, in bronchioles of similar size. Scale bar represents 20 µm. The bar graphs represent the area of α-SMA staining per micrometer length of the bronchiolar basement membrane (µm<sup>2</sup>/µm; <span class="html-italic">n</span> = 6). (<b>D</b>) Detection of the mRNA levels of <span class="html-italic">Acta2</span>, <span class="html-italic">Tgfb1</span>, <span class="html-italic">Col1a1</span>, and <span class="html-italic">Col3a1</span> in lung tissues using qRT-PCR (<span class="html-italic">n</span> = 4). Data are shown as the mean ± SEM. <span class="html-italic">* p</span> &lt; 0.05 compared with control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA.</p>
Full article ">Figure 4
<p>Inhibitory effects of PBM on the ROS-mediated ER stress in a PM<sub>2.5</sub>-exposed asthma exacerbation model. (<b>A</b>) Representative lung sections were stained with antibody specific for 8-hydroxy-2′-deoxyguanosine (8-OHdG). Bar graphs represent the quantification of positive areas of 8-OHdG in each experimental group (<span class="html-italic">n</span> = 4). Scale bar represents 100 µm. (<b>B</b>) ROS levels in lung tissue were measured in relative fluorescence units (RFU). (<b>C</b>) Protein expression of superoxide dismutase 1 (SOD1) and peroxiredoxin 4 (PRDX4). (<b>D</b>) ER stress markers (PERK, eIF2α, ATF4, and CHOP) in lung tissues by Western blotting. β-actin was used as a loading control. Bar graphs represent the quantification protein expression (<span class="html-italic">n</span> = 3). Data are shown as the mean ± SEM. <span class="html-italic">* p</span> &lt; 0.05 compared with control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA.</p>
Full article ">Figure 5
<p>Inhibitory effects of PBM on the cell death in a PM<sub>2.5</sub>-exposed asthma exacerbation model. (<b>A</b>) Representative immunofluorescence for TUNEL (green) and DAPI (blue) staining. Scale bar represents 50 µm. Bar graphs represent TUNEL (+)/DAPI (+) cells in the lung tissues (<span class="html-italic">n</span> = 3). (<b>B</b>) Apoptotic markers in lung tissues. Bar graphs represent the quantification protein expression (<span class="html-italic">n</span> = 3). Bar graphs represent the quantification protein expression (<span class="html-italic">n</span> = 3). Data are shown as the mean ± SEM. <span class="html-italic">* p</span> &lt; 0.05 compared with control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared with PM + OVA.</p>
Full article ">Figure 6
<p>Inhibitory effects of PBM on the ferroptosis and autophagic signals in a PM<sub>2.5</sub>-exposed asthma exacerbation model. (<b>A</b>) Deposition of iron in lung tissue using Perls Prussian blue staining in lung tissues (<span class="html-italic">n</span> = 6). Scale bar represents 20 µm. (<b>B</b>) Malondialdehyde (MDA) concentration in lung tissue (<span class="html-italic">n</span> = 6). (<b>C</b>) Glutathione (GSH) concentration in lung tissue (<span class="html-italic">n</span> = 6). (<b>D</b>) Ca<sup>2+</sup> levels in lung tissue (<span class="html-italic">n</span> = 5). (<b>E</b>) 4-Hydroxynonenal (4-HNE) levels in lung tissue (<span class="html-italic">n</span> = 6). (<b>F</b>) Ferroptosis markers in lung tissue (<span class="html-italic">n</span> = 3). (<b>G</b>) Autophagy markers in lung tissues. Bar graphs represent the quantification protein expression (<span class="html-italic">n</span> = 3). Data are shown as the mean ± SEM. <span class="html-italic">* p</span> &lt; 0.05 compared to control. <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05 compared to PM + OVA.</p>
Full article ">Figure 7
<p>Schematic representation of the anti-asthmatic effects of photobiomodulation (PBM) therapy on PM<sub>2.5</sub> exposure-induced allergic asthma in a mouse model. PBM therapy reduces AHR, inflammation, Th2 cytokines, goblet cell hyperplasia, and subepithelial fibrosis in a PM<sub>2.5</sub>-exacerbated allergic asthma mouse model. PBM therapy also decreases oxidative and ER stress, apoptosis, and ferroptosis, while modulating autophagy in asthmatic mice exposed to PM<sub>2.5</sub>. These findings suggest PBM’s potential as an adjunct to asthma treatment in patients exposed to environmental pollutants. Abbreviations: DEX, dexamethasone; OVA, ovalbumin; PBM, photobiomodulation; PM, particulate matter, PM<sub>2.5</sub>, PM with a diameter &lt; 2.5 μm.</p>
Full article ">

Review

Jump to: Research

15 pages, 782 KiB  
Review
Deciphering the Liaison Between Fine Particulate Matter Pollution, Oxidative Stress, and Prostate Cancer: Where Are We Now?
by Chiang-Wen Lee, Yao-Chang Chiang, Thi Thuy Tien Vo, Zih-Chan Lin, Miao-Ching Chi, Mei-Ling Fang, Kuo-Ti Peng, Ming-Horng Tsai and I-Ta Lee
Antioxidants 2024, 13(12), 1505; https://doi.org/10.3390/antiox13121505 - 10 Dec 2024
Viewed by 493
Abstract
Prostate cancer (PCa), a highly prevalent cancer in men worldwide, is projected to rise in the coming years. As emerging data indicate the carcinogenic effects of fine particulate matter (PM2.5) in lung cancer and other site-specific cancers, there is an urgent need to [...] Read more.
Prostate cancer (PCa), a highly prevalent cancer in men worldwide, is projected to rise in the coming years. As emerging data indicate the carcinogenic effects of fine particulate matter (PM2.5) in lung cancer and other site-specific cancers, there is an urgent need to evaluate the relationship between this environmental risk factor and PCa as a potential target for intervention. The present review provides up-to-date evidence about the impact of airborne PM2.5 pollution on the initiation and progression of PCa. Examining the composition and characteristics of PM2.5 reveals its ability to induce toxic effects, inflammatory injuries, and oxidative damages. Additionally, PM2.5 can attach to endocrine-disrupting chemicals implicated in prostatic carcinogenesis. Considering the potential significance of oxidative stress in the risk of the disease, our review underlines the protective strategies, such as antioxidant-based approaches, for individuals exposed to increased PM2.5 levels. Moreover, the findings call for further research to understand the associations and mechanisms linking PM2.5 exposure to PCa risk as well as to suggest appropriate measures by policymakers, scientific researchers, and healthcare professionals in order to address this global health issue. Full article
(This article belongs to the Special Issue Oxidative Stress Induced by Air Pollution, 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>The potential mechanisms through which PM2.5 increases PCa risk with an emphasis on redox biology. PM2.5 can generate ROS both directly and indirectly, which, if not properly counteracted by antioxidant capacity, can lead to oxidative stress. As second messengers, ROS can dysregulate varying redox-sensitive signaling transduction pathways, including the mitogen-activated protein kinases (MAPKs) and phosphoinositide 3-kinase/protein kinase B (PI3K/Akt), that are involved in multistage carcinogenesis. Redox states also play an important role in immunity and T-cell activity, in which ROS levels determine immune responses. ROS overproduction may enhance the release of proinflammatory cytokines orchestrated and regulated by many redox-sensitive transcription factors, such as the NF-κB and Nrf-2. In addition, some genetic alterations during PCa progression may contribute to the activity of the androgen receptor (AR), whose regulation exhibits a reciprocal negative feedback mechanism with PI3K/Akt signaling. Interestingly, PM2.5 may have an endocrine-disrupting potential, presenting another exposure source to endocrine disruptors implicated in prostatic carcinogenesis.</p>
Full article ">
Back to TopTop