1
|
Meng X, Du W, Sun Z. Fine particulate matter‑induced cardiac developmental toxicity (Review). Exp Ther Med 2025; 29:6. [PMID: 39534282 PMCID: PMC11552469 DOI: 10.3892/etm.2024.12756] [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: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Fine particulate matter (PM2.5) has become an important risk factor threatening human health. Epidemiological and toxicological investigations have revealed that PM2.5 not only leads to cardiovascular dysfunction, but it also gives rise to various adverse health effects on the human body, such as cardiovascular and cerebrovascular diseases, cancers, neurodevelopmental disorders, depression and autism. PM2.5 is able to penetrate both respiratory and placental barriers, thereby resulting in negative effects on fetal development. A large body of epidemiological evidences has suggested that gestational exposure to PM2.5 increases the incidence of congenital diseases in offspring, including congenital heart defects. In addition, animal model studies have revealed that gestational exposure to PM2.5 can disrupt normal heart development in offspring, although the potential molecular mechanisms have yet to be fully elucidated. The aim of the present review was to provide a brief overview of what is currently known regarding the molecular mechanisms underlying cardiac developmental toxicity in offspring induced by gestational exposure to PM2.5.
Collapse
Affiliation(s)
- Xiangjiang Meng
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Weiyuan Du
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Zongli Sun
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| |
Collapse
|
2
|
Shilnikova K, Kang KA, Piao MJ, Herath HMUL, Fernando PDSM, Boo HJ, Yoon SP, Hyun JW. Shikonin protects skin cells against oxidative stress and cellular dysfunction induced by fine particulate matter. Cell Biol Int 2024; 48:1836-1848. [PMID: 39169545 DOI: 10.1002/cbin.12233] [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: 11/22/2023] [Revised: 07/02/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
Shikonin, an herbal naphthoquinone, demonstrates a broad spectrum of pharmacological properties. Owing to increasingly adverse environmental conditions, human skin is vulnerable to harmful influences from dust particles. This study explored the antioxidant capabilities of shikonin and its ability to protect human keratinocytes from oxidative stress induced by fine particulate matter (PM2.5). We found that shikonin at a concentration of 3 µM was nontoxic to human keratinocytes and effectively scavenged reactive oxygen species (ROS) while increasing the production of reduced glutathione (GSH). Furthermore, shikonin enhanced GSH level by upregulating glutamate-cysteine ligase catalytic subunit and glutathione synthetase mediated by nuclear factor-erythroid 2-related factor. Shikonin reduced ROS levels induced by PM2.5, leading to recovering PM2.5-impaired cellular biomolecules and cell viability. Shikonin restored the GSH level in PM2.5-exposed keratinocytes via enhancing the expression of GSH-synthesizing enzymes. Notably, buthionine sulphoximine, an inhibitor of GSH synthesis, diminished effect of shikonin against PM2.5-induced cell damage, confirming the role of GSH in shikonin-induced cytoprotection. Collectively, these findings indicated that shikonin could provide substantial cytoprotection against the adverse effects of PM2.5 through direct ROS scavenging and modulation of cellular antioxidant system.
Collapse
Affiliation(s)
- Kristina Shilnikova
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Herath Mudiyanselage Udari Lakmini Herath
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Pincha Devage Sameera Madushan Fernando
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Hye-Jin Boo
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Sang Pil Yoon
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, South Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, South Korea
| |
Collapse
|
3
|
Mady MS, Elsayed HE, Tawfik NF, Moharram FA. Volatiles extracted from Melaleuca Rugulosa (Link) Craven leaves: comparative profiling, bioactivity screening, and metabolomic analysis. BMC Complement Med Ther 2024; 24:394. [PMID: 39538246 PMCID: PMC11562704 DOI: 10.1186/s12906-024-04683-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Melaleuca species (family Myrtaceae) are characterized by their wide-ranging applications as antimicrobials and in skin-related conditions. Herein, we estimated the volatile profile and biological significance of M. rugulosa (Link) leaves for the first time supported by a dereplication protocol. MATERIALS AND METHODS Volatile components were extracted using hydrodistillation (HD), supercritical fluid (SF), and headspace (HS) techniques and identified using GC/MS. The variations among the three extracts were assessed using principal component analysis and orthogonal partial least square discriminant analysis (OPLS-DA). The extracted volatiles were tested for radical scavenging activity, anti-aging, and anti-hyperpigmentation potential. Finally, disc diffusion and broth microdilution assays were implemented to explore the antibacterial capacity against Streptococcus pyogenes, Staphylococcus aureus, Clostridium perfringens, and Pseudomonas aeruginosa. RESULTS The yield of the SF technique (0.8%) was three times higher than HD. GC/MS analysis revealed that the oxygenated compounds are the most proponents in the three extracts being 95.93% (HD), 80.94% (HS), and 48.4% (SF). Moreover, eucalyptol (1,8-cineol) represents the major component in the HD-EO (89.60%) and HS (73.13%) volatiles, while dl-α-tocopherol (16.27%) and α-terpineol (11.89%) represent the highest percentage in SF extract. Regarding the bioactivity profile, the HD-EO and SF-extract showed antioxidant potential in terms of oxygen radical absorbance capacity, and β- carotene assays, while exerting weak activity towards DPPH. In addition, they displayed potent anti-elastase and moderate anti-collagenase activities. The HD-EO exhibited potent anti-tyrosinase activity, while the SF extract showed a moderate level compared to tested controls. OPLS-DA and dereplication studies predicted that the selective antibacterial activity of HD-EO to S. aureus was related to eucalyptol, while SF extract to C. perfringens was related to α-tocopherol. CONCLUSIONS M. rugulosa leaves are considered a vital source of bioactive volatile components that are promoted for controlling skin aging and infection. However, further safety and clinical studies are recommended.
Collapse
Affiliation(s)
- Mohamed S Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt.
| | - Heba E Elsayed
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Nashwa F Tawfik
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| | - Fatma A Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
4
|
Lei Y, Lei TH, Lu C, Zhang X, Wang F. Wildfire Smoke: Health Effects, Mechanisms, and Mitigation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024. [PMID: 39516728 DOI: 10.1021/acs.est.4c06653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Wildfires are becoming more frequent and intense on a global scale, raising concerns about their acute and long-term effects on human health. We conducted a systematic review of the current epidemiological evidence on wildfire health risks and a meta-analysis to investigate the association between wildfire smoke exposure and various health outcomes. We discovered that wildfire smoke increases the risk of premature deaths and respiratory morbidity in the general population. Meta-analysis of cause-specific mortality and morbidity revealed that wildfire smoke had the strongest associations with cardiovascular mortality (RR: 1.018, 95% CI: 1.014-1.021), asthma hospitalization (RR: 1.054, 95% CI: 1.026-1.082), and asthma emergency department visits (RR: 1.117, 95% CI: 1.035-1.204) in the general population. Subgroup analyses of age found that adults and elderly adults were more susceptible to the cardiopulmonary effects of wildfire smoke. Next, we systematically addressed the toxicological mechanisms of wildfire smoke, including direct toxicity, oxidative stress, inflammatory reactions, immune dysregulation, genotoxicity and mutations, skin allergies, inflammation, and others. We discuss wildfire smoke risk mitigation strategies including public health interventions, regulatory measures, and personal actions. We conclude by highlighting current research limitations and future directions for wildfire research, such as elucidating the complex interactions of wildfire smoke components on human health, developing personalized risk assessment tools, and improving resilience and adaptation strategies to mitigate the health effects of wildfires in changing climate.
Collapse
Affiliation(s)
- Ying Lei
- Centre for Molecular Biosciences and Non-Communicable Diseases, School of Safety Science and Technology, Xi'an University of Science and Technology, Xi'an 710054, China
| | - Tze-Huan Lei
- Centre for Molecular Biosciences and Non-Communicable Diseases, School of Safety Science and Technology, Xi'an University of Science and Technology, Xi'an 710054, China
| | - Chan Lu
- XiangYa School of Public Health, Central South University, Changsha 410008, China
| | - Xue Zhang
- Centre for Molecular Biosciences and Non-Communicable Diseases, School of Safety Science and Technology, Xi'an University of Science and Technology, Xi'an 710054, China
| | - Faming Wang
- Centre for Molecular Biosciences and Non-Communicable Diseases, School of Safety Science and Technology, Xi'an University of Science and Technology, Xi'an 710054, China
- Division of Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, Leuven 3001, Belgium
| |
Collapse
|
5
|
Guillot AJ, Martínez-Navarrete M, Giner RM, Recio MC, Santos HA, Cordeiro AS, Melero A. Cyanocobalamin-loaded dissolving microneedles diminish skin inflammation in vivo. J Control Release 2024; 375:537-551. [PMID: 39299488 DOI: 10.1016/j.jconrel.2024.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/22/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Inflammatory diseases of the skin have a considerable high prevalence worldwide and negatively impact the patients' quality of life. First-line standard therapies for these conditions inherently entail important side effects when used long-term, particularly complicating the management of chronic cases. Therefore, there is a need to develop novel therapeutic strategies to offer reliable alternative treatments. Abnormally high reactive oxygen species (ROS) levels are characteristic of this kind of illnesses, and therefore a reasonable therapeutic goal. Cyanocobalamin, also known as Vitamin B12, possesses notable antioxidant and ROS-scavenging properties which could make it a possible therapeutic alternative. However, its considerable molecular weight restricts passive diffusion through the skin and forces the use of an advanced transdermal delivery system. Here, we present several prototypes of Cyanocobalamin-loaded Dissolving Microarray Patches (B12@DMAPs) with adequate mechanical properties to effectively penetrate the stratum corneum barrier, allowing drug deposition into the skin structure. Ex vivo penetration and permeability studies noted an effective drug presence within the dermal skin layers; in vitro compatibility studies in representative cell skin cell lines such as L929 fibroblasts and HaCaT keratinocytes ensured their safe use. The in vivo efficacy of the selected prototype was tested in a delayed-type hypersensitivity murine model that mimics an inflammatory skin process. Several findings such as a reduction of MPO-related photon emission in a bioluminescence study, protection against histological damage, and decrease of inflammatory cytokines levels point out the effectivity of B12@DMAPs to downregulate the skin inflammatory environment. Overall, B12@DMAPs offer a cost-effective translational alternative for improving patients' skin healthcare.
Collapse
Affiliation(s)
- Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain; Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen. Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Miquel Martínez-Navarrete
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - Rosa Maria Giner
- Department of Pharmacology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - Maria Carmen Recio
- Department of Pharmacology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| | - Helder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen. Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical, Health and Social Care Innovations, De Montfort University, The Gateway LE1 9BH, Leicester, United Kingdom.
| | - Ana Melero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100 Burjassot, Valencia, Spain
| |
Collapse
|
6
|
Lim EY, Kim GD. Particulate Matter-Induced Emerging Health Effects Associated with Oxidative Stress and Inflammation. Antioxidants (Basel) 2024; 13:1256. [PMID: 39456509 PMCID: PMC11505051 DOI: 10.3390/antiox13101256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Environmental pollution continues to increase with industrial development and has become a threat to human health. Atmospheric particulate matter (PM) was designated as a Group 1 carcinogen by the International Agency for Research on Cancer in 2013 and is an emerging global environmental risk factor that is a major cause of death related to cardiovascular and respiratory diseases. PM is a complex composed of highly reactive organic matter, chemicals, and metal components, which mainly cause excessive production of reactive oxygen species (ROS) that can lead to DNA and cell damage, endoplasmic reticulum stress, inflammatory responses, atherosclerosis, and airway remodeling, contributing to an increased susceptibility to and the exacerbation of various diseases and infections. PM has various effects on human health depending on the particle size, physical and chemical characteristics, source, and exposure period. PM smaller than 5 μm can penetrate and accumulate in the alveoli and circulatory system, causing harmful effects on the respiratory system, cardiovascular system, skin, and brain. In this review, we describe the relationship and mechanism of ROS-mediated cell damage, oxidative stress, and inflammatory responses caused by PM and the health effects on major organs, as well as comprehensively discuss the harmfulness of PM.
Collapse
Affiliation(s)
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea;
| |
Collapse
|
7
|
Zhao H, Zhao H, Tang Y, Li M, Cai Y, Xiao X, He F, Huang H, Zhang Y, Li J. Skin-permeable gold nanoparticles with modifications azelamide monoethanolamine ameliorate inflammatory skin diseases. Biomark Res 2024; 12:118. [PMID: 39385245 PMCID: PMC11465885 DOI: 10.1186/s40364-024-00663-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Traditional topical drug delivery for treating inflammatory skin diseases suffers from poor skin penetration and long-term side effects. Metal nanoparticles show promising application in topical drug delivery for inflammatory skin diseases. METHODS Here, we synthesized a new type of nanoparticles, azelamide monoethanolamine-functionalized gold nanoparticles (Au-MEA NPs), based on citrate-capped gold nanoparticles (Au-CA NPs) via the ligand exchange method. The physical and chemical properties of Au-CA NPs and Au-MEA NPs were characterized. In vivo studies were performed using imiquimod-induced psoriasis and LL37-induced rosacea animal models, respectively. For in vitro studies, a model of cellular inflammation was established using HaCaT cells stimulated with TNF-α. In addition, proteomics, gelatin zymography, and other techniques were used to investigate the possible therapeutic mechanisms of the Au-MEA NPs. RESULTS We found that Au-MEA NPs exhibited better stability and permeation properties compared to conventional Au-CA NPs. Transcutaneously administered Au-MEA NPs exerted potent therapeutic efficacy against both rosacea-like and psoriasiform skin dermatitis in vivo without overt signs of toxicity. Mechanistically, Au-MEA NPs reduced the production of pro-inflammatory mediators in keratinocytes by promoting SOD activity and inhibiting the activity of MMP9. CONCLUSION Au-MEA NPs have the potential to be a topical nanomedicine for the effective and safe treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- He Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Han Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Mengfan Li
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, 410082, P. R. China
| | - Yisheng Cai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xin Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Fanping He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hongwen Huang
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, 410082, P. R. China.
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
8
|
Liu Y, Zhang W, Wang H, Liu H, Yu Q, Luo X, Feng X, Yang P. Fine particulate matter potentiates Th17-cell pathogenicity in experimental autoimmune uveitis via ferroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116979. [PMID: 39232294 DOI: 10.1016/j.ecoenv.2024.116979] [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: 04/24/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
The effect of fine particulate matter (PM2.5) on the development of uveitis remains unclear. Therefore, this study was designed to investigate the role of PM2.5 in experimental autoimmune uveitis (EAU) and its potential mechanism. Our results showed that PM2.5 could exacerbate the activity of EAU, as evidenced by severer clinical and pathological changes, correlated with elevated Th17 cells frequency and IL-17A expression. Proteomic analysis revealed ferroptosis was the most significant pathway. In vivo, the levels of Fe2+, ROS, lipid ROS, and malondialdehyde, as well as the expression of TFRC, HMOX1, FTH1, and FTL1 in CD4+ T cells were increased, while GSH/GSSG ratio and the expression of ACSL1 and GPX4 were decreased after PM2.5 exposure. In vitro, the expression of TFRC and HMOX1 were increased, while the expression FTH1, FTL1, ACSL1, and GPX4 were decreased after PM2.5 exposure. Ferrostatin-1 effectively alleviated PM2.5-induced intraocular inflammation and suppressed the frequency of Th17 cells. These results suggest that PM2.5 could aggravate intraocular inflammation and immune response in EAU mice through ferroptosis. Ferroptosis could be a potential marker for the prevention and treatment of uveitis.
Collapse
Affiliation(s)
- Yaning Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wanyun Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmiao Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiuyue Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang Luo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaojie Feng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
9
|
Lee JS, Lee Y, Jang S, Oh JH, Lee DH, Cho S. Pregnane X receptor reduces particulate matter-induced type 17 inflammation in atopic dermatitis. Front Immunol 2024; 15:1415350. [PMID: 39399487 PMCID: PMC11467722 DOI: 10.3389/fimmu.2024.1415350] [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: 04/10/2024] [Accepted: 08/23/2024] [Indexed: 10/15/2024] Open
Abstract
Background Epidemiological evidence suggests that particulate matter (PM) exposure can trigger or worsen atopic dermatitis (AD); however, the underlying mechanisms remain unclear. Recently, pregnane X receptor (PXR), a xenobiotic receptor, was reported to be related to skin inflammation in AD. Objectives This study aimed to explore the effects of PM on AD and investigate the role of PXR in PM-exposed AD. Methods In vivo and in vitro AD-like models were employed, using BALB/c mice, immortalized human keratinocytes (HaCaT), and mouse CD4 + T cells. Results Topical application of PM significantly increased dermatitis score and skin thickness in AD-like mice. PM treatment increased the mRNA and protein levels of type 17 inflammatory mediators, including interleukin (IL)-17A, IL-23A, IL-1β, and IL-6, in AD-like mice and human keratinocytes. PM also activated PXR signaling, and PXR knockdown exacerbated PM-induced type 17 inflammation in human keratinocytes and mouse CD4 + T cells. In contrast, PXR activation by rifampicin (a human PXR agonist) reduced PM-induced type 17 inflammation. Mechanistically, PXR activation led to a pronounced inhibition of the nuclear factor kappa B (NF-κB) pathway. Conclusion In summary, PM exposure induces type 17 inflammation and PXR activation in AD. PXR activation reduces PM-induced type 17 inflammation by suppressing the NF-κB signaling pathway. Thus, PXR represents a promising therapeutic target for controlling the PM-induced AD aggravation.
Collapse
Affiliation(s)
- Ji Su Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Youngae Lee
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Sunhyae Jang
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging and Hair Research, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jang-Hee Oh
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
- Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Soyun Cho
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Department of Dermatology, Seoul Metropolitan Government – Seoul National University (SMG-SNU) Boramae Medical Center, Seoul, Republic of Korea
| |
Collapse
|
10
|
Paik K, Na JI, Huh CH, Shin JW. Particulate Matter and Its Molecular Effects on Skin: Implications for Various Skin Diseases. Int J Mol Sci 2024; 25:9888. [PMID: 39337376 PMCID: PMC11432173 DOI: 10.3390/ijms25189888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Particulate matter (PM) is a harmful air pollutant composed of chemicals and metals which affects human health by penetrating both the respiratory system and skin, causing oxidative stress and inflammation. This review investigates the association between PM and skin disease, focusing on the underlying molecular mechanisms and specific disease pathways involved. Studies have shown that PM exposure is positively associated with skin diseases such as atopic dermatitis, psoriasis, acne, and skin aging. PM-induced oxidative stress damages lipids, proteins, and DNA, impairing cellular functions and triggering inflammatory responses through pathways like aryl hydrocarbon receptor (AhR), NF-κB, and MAPK. This leads to increased production of inflammatory cytokines and exacerbates skin conditions. PM exposure exacerbates AD by triggering inflammation and barrier disruption. It disrupts keratinocyte differentiation and increases pro-inflammatory cytokines in psoriasis. In acne, it increases sebum production and inflammatory biomarkers. It accelerates skin aging by degrading ECM proteins and increasing MMP-1 and COX2. In conclusion, PM compromises skin health by penetrating skin barriers, inducing oxidative stress and inflammation through mechanisms like ROS generation and activation of key pathways, leading to cellular damage, apoptosis, and autophagy. This highlights the need for protective measures and targeted treatments to mitigate PM-induced skin damage.
Collapse
Affiliation(s)
- Kyungho Paik
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung-Im Na
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chang-Hun Huh
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung-Won Shin
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
11
|
Herath HMUL, Piao MJ, Kang KA, Fernando PDSM, Kang HK, Koh YS, Hyun JW. The inhibitory effect of chlorogenic acid on oxidative stress and apoptosis induced by PM 2.5 in HaCaT keratinocytes. J Biochem Mol Toxicol 2024; 38:e23806. [PMID: 39148258 DOI: 10.1002/jbt.23806] [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: 12/10/2023] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 08/17/2024]
Abstract
Exposure to fine particulate matter with an aerodynamic diameter of less than 2.5 μm (PM2.5) can cause oxidative damage and apoptosis in the human skin. Chlorogenic acid (CGA) is a bioactive polyphenolic compound with antioxidant, antifungal, and antiviral properties. The objective of this study was to identify the ameliorating impact of CGA that might protect human HaCaT cells against PM2.5. CGA significantly scavenged the reactive oxygen species (ROS) generated by PM2.5, attenuated oxidative cellular/organelle damage, mitochondrial membrane depolarization, and suppressed cytochrome c release into the cytosol. The application of CGA led to a reduction in the expression levels of Bcl-2-associated X protein, caspase-9, and caspase-3, while simultaneously increasing the expression of B-cell lymphoma 2. In addition, CGA was able to reverse the decrease in cell viability caused by PM2.5 via the inhibition of extracellular signal-regulated kinase (ERK). This effect was further confirmed by the use of the mitogen-activated protein kinase kinase inhibitor, which acted upstream of ERK. In conclusion, CGA protected keratinocytes from mitochondrial damage and apoptosis via ameliorating PM2.5-induced oxidative stress and ERK activation.
Collapse
Affiliation(s)
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Pincha Devage Sameera Madushan Fernando
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Hee Kyoung Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Young Sang Koh
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
12
|
Fernando PDSM, Piao MJ, Herath HMUL, Kang KA, Hyun CL, Kim ET, Koh YS, Hyun JW. Hyperoside reduced particulate matter 2.5-induced endoplasmic reticulum stress and senescence in skin cells. Toxicol In Vitro 2024; 99:105870. [PMID: 38848825 DOI: 10.1016/j.tiv.2024.105870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Particulate matter 2.5 (PM2.5) causes skin aging, inflammation, and impaired skin homeostasis. Hyperoside, a flavanol glycoside, has been proposed to reduce the risk of diseases caused by oxidative stress. This study evaluated the cytoprotective potential of hyperoside against PM2.5-induced skin cell damage. Cultured human HaCaT keratinocytes were pretreated with hyperoside and treated with PM2.5. Initially, the cytoprotective and antioxidant ability of hyperoside against PM2.5 was evaluated. Western blotting was further employed to investigate endoplasmic reticulum (ER) stress and cellular senescence and for evaluation of cell cycle regulation-related proteins. Hyperoside inhibited PM2.5-mediated ER stress as well as mitochondrial damage. Colony formation assessment confirmed that PM2.5-impaired cell proliferation was restored by hyperoside. Moreover, hyperoside reduced the activation of PM2.5-induced ER stress-related proteins, such as protein kinase R-like ER kinase, cleaved activating transcription factor 6, and inositol-requiring enzyme 1. Hyperoside promoted cell cycle progression in the G0/G1 phase by upregulating the PM2.5-impaired cell cycle regulatory proteins. Hyperoside significantly reduced the expression of PM2.5-induced senescence-associated β-galactosidase and matrix metalloproteinases (MMPs), such as MMP-1 and MMP-9. Overall, hyperoside ameliorated PM2.5-impaired cell proliferation, ER stress, and cellular senescence, offering potential therapeutic implications for mitigating the adverse effects of environmental pollutants on skin health.
Collapse
Affiliation(s)
- Pincha Devage Sameera Madushan Fernando
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea; Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei Jing Piao
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea; Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | - Kyoung Ah Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea; Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Chang Lim Hyun
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Eui Tae Kim
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Young Sang Koh
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jin Won Hyun
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea; Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
13
|
Zhen AX, Kang KA, Piao MJ, Madushan Fernando PDS, Lakmini Herath HMU, Hyun JW. Protective effects of astaxanthin on particulate matter 2.5‑induced senescence in HaCaT keratinocytes via maintenance of redox homeostasis. Exp Ther Med 2024; 28:275. [PMID: 38800049 PMCID: PMC11117106 DOI: 10.3892/etm.2024.12563] [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: 11/21/2023] [Accepted: 04/11/2024] [Indexed: 05/29/2024] Open
Abstract
Particulate matter 2.5 (PM2.5) imposes a heavy burden on the skin and respiratory system of human beings, causing side effects such as aging, inflammation and cancer. Astaxanthin (ATX) is a well-known antioxidant widely used for its anti-inflammatory and anti-aging properties. However, few studies have investigated the protective effects of ATX against PM2.5-induced senescence in HaCaT cells. In the present study, the levels of reactive oxygen species (ROS) and antioxidant enzymes were measured after treatment with PM2.5. The results revealed that PM2.5 generated excessive ROS and reduced the translocation of nuclear factor erythroid 2-related factor 2 (NRF2), subsequently reducing the expression of antioxidant enzymes. However, pretreatment with ATX reversed the ROS levels as well as the expression of antioxidant enzymes. In addition, ATX protected cells from PM2.5-induced DNA damage and rescued PM2.5-induced cell cycle arrest. The levels of senescence-associated phenotype markers, such as interleukin-1β, matrix metalloproteinases, and β-galactosidase, were increased by exposure to PM2.5, however these effects were reversed by ATX. After interfering with NRF2 mRNA expression and exposing cells to PM2.5, the levels of ROS and β-galactosidase were higher compared with siControl RNA cells exposed to PM2.5. However, ATX inhibited ROS and β-galactosidase levels in both the siControl RNA and the siNRF2 RNA groups. Thus, ATX protects HaCaT keratinocytes from PM2.5-induced senescence by partially inhibiting excessive ROS generation via the NRF2 signaling pathway.
Collapse
Affiliation(s)
- Ao Xuan Zhen
- Department of Biochemistry, College of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Jin Won Hyun
- Department of Biochemistry, College of Medicine and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
14
|
Kim JH, Hwang KH, Kim SH, Kim HJ, Kim JM, Lee MY, Cha SK, Lee J. Particulate Matter-Induced Neurotoxicity: Unveiling the Role of NOX4-Mediated ROS Production and Mitochondrial Dysfunction in Neuronal Apoptosis. Int J Mol Sci 2024; 25:6116. [PMID: 38892302 PMCID: PMC11172693 DOI: 10.3390/ijms25116116] [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: 04/26/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Urban air pollution, a significant environmental hazard, is linked to adverse health outcomes and increased mortality across various diseases. This study investigates the neurotoxic effects of particulate matter (PM), specifically PM2.5 and PM10, by examining their role in inducing oxidative stress and subsequent neuronal cell death. We highlight the novel finding that PM increases mitochondrial ROS production via stimulating NOX4 activity, not through its expression level in Neuro-2A cells. Additionally, PMs provoke ROS production via increasing the expression and activity of NOX2 in SH-SY5Y human neuroblastoma cells, implying differential regulation of NOX proteins. This increase in mitochondrial ROS triggers the opening of the mitochondrial permeability transition pore (mPTP), leading to apoptosis through key mediators, including caspase3, BAX, and Bcl2. Notably, the voltage-dependent anion-selective channel 1 (VDAC1) increases at 1 µg/mL of PM2.5, while PM10 triggers an increase from 10 µg/mL. At the same concentration (100 µg/mL), PM2.5 causes 1.4 times higher ROS production and 2.4 times higher NOX4 activity than PM10. The cytotoxic effects induced by PMs were alleviated by NOX inhibitors GKT137831 and Apocynin. In SH-SY5Y cells, both PM types increase ROS and NOX2 levels, leading to cell death, which Apocynin rescues. Variability in NADPH oxidase sources underscores the complexity of PM-induced neurotoxicity. Our findings highlight NOX4-driven ROS and mitochondrial dysfunction, suggesting a potential therapeutic approach for mitigating PM-induced neurotoxicity.
Collapse
Affiliation(s)
- Ji-Hee Kim
- Department of Occupational Therapy, Soonchunhyang University, Asan-si 31538, Republic of Korea;
| | - Kyu-Hee Hwang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Seong-Heon Kim
- Department of Environmental and Energy Engineering, Yonsei University, Wonju 26493, Republic of Korea;
| | - Hi-Ju Kim
- Department of Psychiatry, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| | - Jung-Min Kim
- Department of Medical Science, Soonchunhyang University, Asan-si 31538, Republic of Korea; (J.-M.K.); (M.-Y.L.)
| | - Mi-Young Lee
- Department of Medical Science, Soonchunhyang University, Asan-si 31538, Republic of Korea; (J.-M.K.); (M.-Y.L.)
- Department of Medical Biotechnology, Soonchunhyang University, Asan-si 31538, Republic of Korea
| | - Seung-Kuy Cha
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Jinhee Lee
- Department of Psychiatry, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
| |
Collapse
|
15
|
Dong Y, Zhang Y, Li F, Tang B, Lv D, Wang H, Luo S. GKT137831 in combination with adipose-derived stem cells alleviates high glucose-induced inflammaging and improves diabetic wound healing. J Leukoc Biol 2024; 115:882-892. [PMID: 37774495 DOI: 10.1093/jleuko/qiad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) have been proven to promote healing in diabetic wounds, which are one of the most serious chronic refractory wounds. However, reactive oxygen species (ROS) induced by high glucose (HG) lead to oxidative stress and aging in ADSCs, which limits the therapeutic effect of ADSCs. In this study, we investigated the role of GKT137831, a NOX1/4 inhibitor that can reduce ROS production, in protecting ADSCs from hyperglycemia and in diabetic wound healing. In vitro, ROS levels and NOX4 expression were increased after HG treatment of ADSCs, while the oxidative stress marker malondialdehyde was increased; mitochondrial membrane potential was decreased; inflammatory aging-related indicators such as p16, p21, matrix metalloproteinase-1 (MMP1), MMP3, interleukin-6, and β-galactosidase were increased; and migration was weakened. In vivo, we constructed a diabetic mouse wound model and found that the combination of ADSCs and GKT137831 synergistically promoted the 21-day wound healing rate, increased the expression of collagen and hydroxyproline, increased the number of blood vessels and the expression of CD31, and reduced the expression of interleukin-6, MMP1, MMP3, and p21. These results suggest that GKT137831 could protect ADSCs from oxidative stress and aging induced by HG and enhance the therapeutic effect of ADSCs on diabetic wounds.
Collapse
Affiliation(s)
- Yunxian Dong
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, No. 466 Middle Xingang Road, Guangzhou, Guangdong Province 510317, China
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Youliang Zhang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, No. 466 Middle Xingang Road, Guangzhou, Guangdong Province 510317, China
| | - Fangwei Li
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, No. 466 Middle Xingang Road, Guangzhou, Guangdong Province 510317, China
| | - Bing Tang
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, Guangdong Province 510080, China
| | - Dongming Lv
- Department of Burn Surgery, First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, Guangdong Province 510080, China
| | - Haibin Wang
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, No. 466 Middle Xingang Road, Guangzhou, Guangdong Province 510317, China
| | - Shengkang Luo
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, No. 466 Middle Xingang Road, Guangzhou, Guangdong Province 510317, China
| |
Collapse
|
16
|
Kang KA, Piao MJ, Fernando PDSM, Herath HMUL, Yi JM, Choi YH, Hyun YM, Zhang K, Park CO, Hyun JW. Particulate matter stimulates the NADPH oxidase system via AhR-mediated epigenetic modifications. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123675. [PMID: 38447650 DOI: 10.1016/j.envpol.2024.123675] [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: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Stimulation of human keratinocytes with particulate matter 2.5 (PM2.5) elicits complex signaling events, including a rise in the generation of reactive oxygen species (ROS). However, the mechanisms underlying PM2.5-induced ROS production remain unknown. Here, we show that PM2.5-induced ROS production in human keratinocytes is mediated via the NADPH oxidase (NOXs) system and the Ca2+ signaling pathway. PM2.5 treatment increased the expression of NOX1, NOX4, and a calcium-sensitive NOX, dual oxidase 1 (DUOX1), in human epidermal keratinocyte cell line. PM2.5 bound to aryl hydrocarbon receptor (AhR), and this complex bound to promoter regions of NOX1 and DUOX1, suggesting that AhR acted as a transcription factor of NOX1 and DUOX1. PM2.5 increased the transcription of DUOX1 via epigenetic modification. Moreover, a link between DNA demethylase and histone methyltransferase with the promoter regions of DUOX1 led to an elevation in the expression of DUOX1 mRNA. Interestingly, PM2.5 increased NOX4 expression and promoted the interaction of NOX4 and Ca2+ channels within the cytoplasmic membrane or endoplasmic reticulum, leading to Ca2+ release. The increase in intracellular Ca2+ concentration activated DUOX1, responsible for ROS production. Our findings provide evidence for a PM2.5-mediated ROS-generating system network, in which increased NOX1, NOX4, and DUOX1 expression serves as a ROS signal through AhR and Ca2+ activation.
Collapse
Affiliation(s)
- Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, 63243, South Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, 63243, South Korea
| | | | | | - Joo Mi Yi
- Department of Microbiology and Immunology, Inje University College of Medicine, Busan, 47392, South Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan, 47340, South Korea
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Kelun Zhang
- Department of Dermatology, and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Chang Ook Park
- Department of Dermatology, and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju, 63243, South Korea.
| |
Collapse
|
17
|
Zhao H, Zhao H, Li M, Tang Y, Xiao X, Cai Y, He F, Huang H, Zhang Y, Li J. Twin defect-rich Pt ultrathin nanowire nanozymes alleviate inflammatory skin diseases by scavenging reactive oxygen species. Redox Biol 2024; 70:103055. [PMID: 38290385 PMCID: PMC10844124 DOI: 10.1016/j.redox.2024.103055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
Nanozymes with superior antioxidant properties offer new hope for treating oxidative stress-related inflammatory skin diseases. However, lacking sufficient catalytic activity or having complex material designs limit the application of current metallic nanozymes in inflammatory skin diseases. Here, we report a simple and effective twin-defect platinum nanowires (Pt NWs) enzyme with multiple mimetic enzymes and broad-spectrum ROS scavenging capability for the treatment of inflammatory skin diseases in mice (including psoriasis and rosacea). Pt NWs with simultaneous superoxide dismutase, glutathione peroxidase and catalase mimetic enzyme properties exhibit cytoprotective effects against ROS-mediated damage at extremely low doses and significantly improve treatment outcomes in psoriasis- and rosacea-like mice. Meanwhile, these ultrasmall sizes of Pt NWs allow the nanomaterials to effectively penetrate the skin and do not produce significant biotoxicity. Therefore, Pt NWs have potential applications in treating diseases related to oxidative stress or inflammation.
Collapse
Affiliation(s)
- He Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Han Zhao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Mengfan Li
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, 410082, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xin Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yisheng Cai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Fanping He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hongwen Huang
- College of Materials Science and Engineering, Hunan University, Changsha, Hunan, 410082, China
| | - Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
18
|
Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Koh YS, Kang HK, Choi YH, Hyun JW. Protective Effect of Fermented Sea Tangle Extract on Skin Cell Damage Caused by Particulate Matter. Int J Med Sci 2024; 21:937-948. [PMID: 38617009 PMCID: PMC11008479 DOI: 10.7150/ijms.93034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/25/2024] [Indexed: 04/16/2024] Open
Abstract
The skin is directly exposed to atmospheric pollutants, especially particulate matter 2.5 (PM2.5) in the air, which poses significant harm to skin health. However, limited research has been performed to identify molecules that can confer resistance to such substances. Herein, we analyzed the effect of fermented sea tangle (FST) extract on PM2.5-induced human HaCaT keratinocyte damage. Results showed that FST extract, at concentrations less than 800 μg/mL, exhibited non-significant toxicity to cells and concentration-dependent inhibition of PM2.5-induced reactive oxygen species (ROS) production. PM2.5 induced oxidative stress by stimulating ROS, resulting in DNA damage, lipid peroxidation, and protein carbonylation, which were inhibited by the FST extract. FST extract significantly suppressed the increase in calcium level and apoptosis caused by PM2.5 treatment and significantly restored the reduced cell viability. Mitochondrial membrane depolarization occurred due to PM2.5 treatment, however, FST extract recovered mitochondrial membrane polarization. PM2.5 inhibited the expression of the anti-apoptotic protein Bcl-2, and induced the expression of pro-apoptotic proteins Bax and Bim, the apoptosis initiator caspase-9, as well as the executor caspase-3, however, FST extract effectively protected the changes in the levels of these proteins caused by PM2.5. Interestingly, pan-caspase inhibitor Z-VAD-FMK treatment enhanced the anti-apoptotic effect of FST extract in PM2.5-treated cells. Our results indicate that FST extract prevents PM2.5-induced cell damage via inhibition of mitochondria-mediated apoptosis in human keratinocytes. Accordingly, FST extract could be included in skin care products to protect cells against the harmful effects of PM2.5.
Collapse
Affiliation(s)
- Mei Jing Piao
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung Ah Kang
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Young Sang Koh
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Hee Kyoung Kang
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Yung Hyun Choi
- College of Oriental Medicine, Dongeui University, Busan 47340, Republic of Korea
| | - Jin Won Hyun
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
19
|
Wei J, Liu J, Wang H, Wen K, Ni X, Lin Y, Huang J, You X, Lei Z, Li J, Shen H, Lin Y. Nanoplastic propels diet-induced NAFL to NASH via ER-mitochondrial tether-controlled redox switch. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133142. [PMID: 38061129 DOI: 10.1016/j.jhazmat.2023.133142] [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: 08/29/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) is multifactorial that lifestyle, genetic, and environmental factors contribute to its onset and progression, thereby posing a challenge for therapeutic intervention. Nanoplastic (NP) is emerged as a novel environmental metabolism disruptor but the etiopathogenesis remains largely unknown. In this study, C57BL/6 J mice were fed with normal chow diet (NCD) and high-fat diet (HFD) containing 70 nm polystyrene microspheres (NP). We found that dietary-derived NP adsorbed proteins and agglomerated during the in vivo transportation, enabling diet-induced hepatic steatosis to NASH. Mechanistically, NP promoted liver steatosis by upregulating Fatp2. Furthermore, NP stabilized the Ip3r1, and facilitated ER-mitochondria contacts (MAMs) assembly in the hepatocytes, resulting in mitochondrial Ca2+ overload and redox imbalance. The redox-sensitive Nrf2 was decreased in the liver of NP-exposed mice, which positively regulated miR26a via direct binding to its promoter region [-970 bp to -847 bp and -318 bp to -176 bp]. NP decreased miR26a simultaneously upregulated 10 genes involved in MAMs formation, lipid uptake, inflammation, and fibrosis. Moreover, miR26a inhibition elevated MAMs-tether Vdac1, which promoted the nucleus translocation of NF-κB P65 and Keap1 and functionally inactivated Nrf2, leading to a vicious cycle. Hepatocyte-specific overexpressing miR26a effectively restored ER-mitochondria miscommunication and ameliorated NASH phenotype in NP-exposed and Keap1-overexpressed mice on HFD. The hepatic MAM-tethers/Nrf2/miR26a feedback loop is an essential metabolic switch from simple steatosis to NASH and a promising therapeutic target for oxidative stress-associated liver damage and NASH.
Collapse
Affiliation(s)
- Jie Wei
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jintao Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Huan Wang
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Kai Wen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Xiuye Ni
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Yilong Lin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jingru Huang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiang You
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Zhao Lei
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Juan Li
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Heqing Shen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| | - Yi Lin
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| |
Collapse
|
20
|
Tang J, Diao P, Pan W, Li L, Xiong L. The cross-linking between DNA damage, oxidative stress and epidermal barrier in keratinocytes after exposure to particulate matters and carbon black. Exp Dermatol 2024; 33:e15048. [PMID: 38439204 DOI: 10.1111/exd.15048] [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: 04/18/2023] [Revised: 10/07/2023] [Accepted: 12/08/2023] [Indexed: 03/06/2024]
Abstract
As the largest organ, the skin provides the first line of defence against environmental pollutants. Different pollutants have varied damage to the skin due to their own physical-chemical properties. A previous epidemiological study by our team revealed that eczema was positively correlated with different air pollutants. However, the mechanism of action from different pollutants on the skin is less known. In this work, the differences among the genotoxicity, intracellular reactive oxygen species, and barrier-related parameters caused by two kinds of air pollutants, that is, S1650b and carbon black (CB) were investigated by Western blot, TUNEL, comet assay and RNA-sequences. The results indicated that both S1650b and CB caused DNA damage of keratinocytes. With the content of lipophilic polycyclic aromatic hydrocarbons (PAH), S1650b leaked into the keratinocytes easily, which activated the aromatic hydrocarbon receptor (AhR) in keratinocytes, leading to worse damage to barrier-related proteins than CB. And CB-induced higher intracellular ROS than S1650b due to the smaller size which make it enter the keratinocytes easier. RNA-sequencing results revealed that S1650b and CB both caused DNA damage of keratinocytes, and the intervention of S1650b significantly upregulated AhR, cytochrome oxidase A1 and B1 (CYP1A1 and CYP1B1) genes, while the results showed oppositely after CB intervention. The mechanism of keratinocyte damage caused by different air particle pollutants in this study will help to expand our understanding on the air pollutant-associated skin disease at cell levels.
Collapse
Affiliation(s)
- Jie Tang
- Cosmetics Safety and Efficacy Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Engineering Technology Research Center of Cosmetic, Chengdu, China
- NMPA Key Laboratory for Human Evaluation and Big Data of Cosmetics, Sichuan University, Chengdu, China
| | - Ping Diao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Weixi Pan
- Analytical and Metrical Center of Sichuan Province, Chengdu, China
| | - Li Li
- Cosmetics Safety and Efficacy Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Engineering Technology Research Center of Cosmetic, Chengdu, China
- NMPA Key Laboratory for Human Evaluation and Big Data of Cosmetics, Sichuan University, Chengdu, China
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Lidan Xiong
- Cosmetics Safety and Efficacy Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Engineering Technology Research Center of Cosmetic, Chengdu, China
- NMPA Key Laboratory for Human Evaluation and Big Data of Cosmetics, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Roh YJ, Choi YH, Shin SH, Lee MK, Won YJ, Lee JH, Cho BS, Park KY, Seo SJ. Adipose tissue-derived exosomes alleviate particulate matter-induced inflammatory response and skin barrier damage in atopic dermatitis-like triple-cell model. PLoS One 2024; 19:e0292050. [PMID: 38241278 PMCID: PMC10798485 DOI: 10.1371/journal.pone.0292050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 09/11/2023] [Indexed: 01/21/2024] Open
Abstract
Recently, particulate matter (PM) has been shown to exacerbate atopic dermatitis (AD) by inducing an inflammatory response. Meanwhile, several studies revealed that exosomes derived from adipose tissue-derived mesenchymal stem cells promote wound healing and alleviate inflammation via their regenerative and immunomodulatory capacities. Our study aimed to investigate the effects of human adipose tissue-derived mesenchymal stem cell-derived (ASC)-exosomes in PM-induced AD. An AD-like triple-cell model was established by treating human keratinocytes, dermal fibroblasts, and mast cells with polyinosinic:polycytidylic acid (Poly I:C) and interleukin 1 alpha (IL-1α). The effects of PM and ASC-exosomes on the expression of pro-inflammatory cytokines and skin barrier proteins were examined using quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. PM increased pro-inflammatory cytokines (IL-6, IL-1β, and IL-1α) and decreased the anti-inflammatory cytokine IL-10, while the mRNA expression of skin barrier proteins (loricrin and filaggrin) decreased. However, when the cells were treated with ASC-exosomes, the PM-induced effects on pro-inflammatory cytokines and skin barrier proteins were reversed. Our results confirmed that PM-induced inflammation and skin barrier damage were alleviated by ASC-exosomes in our AD-like triple-cell model. These data suggest that ASC-exosomes can serve as a therapeutic agent for PM-exacerbated AD.
Collapse
Affiliation(s)
- Yoon Jin Roh
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yong Hee Choi
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sun Hye Shin
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Mi-Kyung Lee
- Department of Laboratory Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yu Jin Won
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Jun Ho Lee
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Kui Young Park
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Seong Jun Seo
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
22
|
Lee J, Kwon J, Jo YJ, Yoon SB, Hyeon JH, Park BJ, You HJ, Youn C, Kim Y, Choi HW, Kim JS. Particulate matter 10 induces oxidative stress and apoptosis in rhesus macaques skin fibroblast. PeerJ 2023; 11:e16589. [PMID: 38130933 PMCID: PMC10734408 DOI: 10.7717/peerj.16589] [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: 08/02/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
Background Particulate matter (PM) is a major air pollutant that affects human health worldwide. PM can pass through the skin barrier, thus causing skin diseases such as heat rash, allergic reaction, infection, or inflammation. However, only a few studies have been conducted on the cytotoxic effects of PM exposure on large-scale animals. Therefore, herein, we investigated whether and how PM affects rhesus macaque skin fibroblasts. Methods Rhesus macaque skin fibroblasts were treated with various concentrations of PM10 (1, 5, 10, 50, and 100 μg/mL) and incubated for 24, 48, and 72 h. Then, cell viability assay, TUNEL assay, and qRT-PCR were performed on the treated cells. Further, the reactive oxygen species, glutathione, and cathepsin B levels were determined. The MTT assay revealed that PM10 (>50 μg/mL) proportionately reduced the cell proliferation rate. Results PM10 treatment increased TUNEL-positive cell numbers, following the pro-apoptosis-associated genes (CASP3 and BAX) and tumor suppressor gene TP53 were significantly upregulated. PM10 treatment induced reactive oxidative stress. Cathepsin B intensity was increased, whereas GSH intensity was decreased. The mRNA expression levels of antioxidant enzyme-related genes (CAT, GPX1 and GPX3) were significantly upregulated. Furthermore, PM10 reduced the mitochondrial membrane potential. The mRNA expression of mitochondrial complex genes, such as NDUFA1, NDUFA2, NDUFAC2, NDUFS4, and ATP5H were also significantly upregulated. In conclusion, these results showed that PM10 triggers apoptosis and mitochondrial damage, thus inducing ROS accumulation. These findings provide potential information on the cytotoxic effects of PM10 treatment and help to understand the mechanism of air pollution-induced skin diseases.
Collapse
Affiliation(s)
- Jiin Lee
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
- Department of Animal Science, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Jeongwoo Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Yu-Jin Jo
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Seung-Bin Yoon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Jae-Hwan Hyeon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Beom-Jin Park
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Hyeong-Ju You
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Changsic Youn
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Yejin Kim
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| | - Hyun Woo Choi
- Department of Animal Science, Jeonbuk National University, Jeonju, Jeollabuk-do, Republic of Korea
| | - Ji-Su Kim
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongup-si, Republic of Korea
| |
Collapse
|
23
|
Ortiz A, Sun H, Kluz T, Matsui MS, Carle T, Gan D, Gordon T, Gildea L, Costa M. A combination of three antioxidants decreases the impact of rural particulate pollution in Normal human keratinocytes. Int J Cosmet Sci 2023; 45:791-801. [PMID: 37602524 DOI: 10.1111/ics.12888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVE It is well established that exposure of human skin to airborne pollution, particularly in the form of particulate matter sized 2.5 μm (PM2.5 ), is associated with oxidative stress, DNA damage and inflammation, leading to premature signs of skin aging. Because much of the damage results from oxidative stress, we examined the effects of a topical composition containing three antioxidants in an in vitro model system to assess the potential for amelioration of premature aging. The use of multiple antioxidants was of interest based on the typical composition of therapeutic skincare products. It is important to determine the efficacy of multiple antioxidants together and develop a short-term assay for larger scale efficacy testing. METHODS Normal human epidermal keratinocytes were exposed to a rural-derived source of PM2.5 in the presence and absence of an antioxidant mixture of resveratrol, niacinamide and GHK peptide. Endpoints related to inflammation, premature aging and carcinogenicity were monitored after 5 h of exposure and included IL-6, CXCL10, MMP-1 and NRF2. Differentially expressed genes were monitored by RNA-seq. RESULTS Pre-treatment of keratinocytes with the antioxidant preparation in the absence of PM2.5 reduced baseline levels of MMP-1, IL-6 and CYP1A1 and reduced PM2.5 -induced increases in all four endpoints, MMP-1, IL-6, CXCL10 and CYP1A1. Antioxidants significantly increased NRF2 protein in the presence of PM2.5 , indicating a protective response. RNA-seq interrogation of antioxidant-treated cells further showed increased expression of NRF2 inducible genes. The expression of CYP1A1 and genes related to aryl hydrocarbon activation were induced by PM2.5 and suppressed by antioxidants. CONCLUSIONS Specific signalling pathways known to be correlated with skin inflammation and aging were examined based on their suitability for use in efficacy testing for the prevention of skin damage due to ambient hydrocarbon pollution. Endpoints examined after only 5 h of exposure provide a useful method amenable to high through-put screening. The results obtained reinforce the concept that a multiple antioxidant preparation, topically applied, may reduce pro-inflammatory signalling and cellular damage and thereby reduce premature skin aging due to exposure to rural-derived airborne pollution.
Collapse
Affiliation(s)
- Angelica Ortiz
- Environmental Medicine, NYU Grossman School of Medicine, New York University, New York City, New York, USA
| | - Hong Sun
- Environmental Medicine, NYU Grossman School of Medicine, New York University, New York City, New York, USA
| | - Thomas Kluz
- Environmental Medicine, NYU Grossman School of Medicine, New York University, New York City, New York, USA
| | - Mary S Matsui
- Mary Kay Inc, Lewisville, Texas, USA
- Skin Biology Research Group, Teaneck, New Jersey, USA
| | | | - David Gan
- Mary Kay Inc, Lewisville, Texas, USA
| | - Terry Gordon
- Environmental Medicine, NYU Grossman School of Medicine, New York University, New York City, New York, USA
| | | | - Max Costa
- Environmental Medicine, NYU Grossman School of Medicine, New York University, New York City, New York, USA
| |
Collapse
|
24
|
Kim J, Kim Y, Song Y, Kim TJ, Lee SH, Kim HJ. Indoor particulate matter induces epigenetic changes in companion atopic dogs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115544. [PMID: 37827097 DOI: 10.1016/j.ecoenv.2023.115544] [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: 07/25/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/14/2023]
Abstract
The prevalence of atopic dermatitis (AD) is increasing and environmental factors are receiving attention as contributing causes. Indoor air pollutants (IAPs), especially particulate matter (PM) can alter epigenetic markers, DNA methylation (DNAm). Although DNAm-mediated epigenetic changes have been reported to modulate the pathogenesis of AD, their role at high risk of exposure to PM is still unclear. The study investigated the effects of exposure to IAPs in the development of AD and epigenetic changes through DNAm in companion atopic dogs that share indoor environment with their owners. Dogs were divided into two groups: AD (n = 47) and controls (n = 21). The IAPs concentration in each household was measured for 48 h, and a questionnaire on the residential environment was completed in all dogs. Eighteen dogs with AD and 12 healthy dogs were selected for DNAm analysis. In addition, clinical and immunological evaluations were conducted. The concentrations of PM2.5, PM10, and volatile organic compounds (VOCs) were significantly higher in the AD group. Moreover, there were more significant methylation differences in the LDLRAD4, KHSRP, and CTDSP2 genes in connection with PM10 in AD group compared to the controls. The degree of methylation of the LDLRAD4 and CTDSP2 genes was also correlated with related protein productions. The present study revealed that exposure to high indoor PM can cause epigenetic development of AD by methylation of the LDLRAD4, KHSRP, and CTDSP2 genes in dogs. Under the concept of "One Health," improving indoor environments should be considered to prevent the development of AD.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea; BK 21 project team, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea
| | - Yeji Kim
- Department of Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea; BK 21 project team, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea
| | - Yunji Song
- Department of Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea; BK 21 project team, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea
| | - Tae Jung Kim
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea
| | - Seung-Hwa Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul 05505, South Korea
| | - Ha-Jung Kim
- Department of Internal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea; BK 21 project team, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
25
|
Pan Z, Dai Y, Akar-Ghibril N, Simpson J, Ren H, Zhang L, Hou Y, Wen X, Chang C, Tang R, Sun JL. Impact of Air Pollution on Atopic Dermatitis: A Comprehensive Review. Clin Rev Allergy Immunol 2023; 65:121-135. [PMID: 36853525 DOI: 10.1007/s12016-022-08957-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 03/01/2023]
Abstract
Air pollution is associated with multiple health problems worldwide, contributing to increased morbidity and mortality. Atopic dermatitis (AD) is a common allergic disease, and increasing evidence has revealed a role of air pollution in the development of atopic dermatitis. Air pollutants are derived from several sources, including harmful gases such as nitrogen dioxide (NO2), sulfur dioxide (SO2), and carbon monoxide (CO), as well as particulate matter (PM) of various sizes, and bioaerosols. Possible mechanisms linking air pollution to atopic dermatitis include damage to the skin barrier through oxidative stress, increased water loss, physicochemical injury, and an effect on skin microflora. Furthermore, oxidative stress triggers immune dysregulation, leading to enhanced sensitization to allergens. There have been multiple studies focusing on the association between various types of air pollutants and atopic dermatitis. Since there are many confounders in the current research, such as climate, synergistic effects of mixed pollutants, and diversity of study population, it is not surprising that inconsistencies exist between different studies regarding AD and air pollution. Still, it is generally accepted that air pollution is a risk factor for AD. Future studies should focus on how air pollution leads to AD as well as effective intervention measures.
Collapse
Affiliation(s)
- Zhouxian Pan
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yimin Dai
- Eight-Year Clinical Medicine System, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Nicole Akar-Ghibril
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA
| | - Jessica Simpson
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA
| | - Huali Ren
- Department of Allergy, Beijing Electric Power Hospital of State Grid Company of China, Electric Power Teaching Hospital of Capital Medical University, Beijing, 100073, China
| | - Lishan Zhang
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yibo Hou
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xueyi Wen
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Christopher Chang
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children's Hospital, Memorial Healthcare System, Hollywood, FL, 33021, USA.
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, 95616, USA.
| | - Rui Tang
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Jin-Lyu Sun
- Allergy Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Allergy Department, Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
26
|
Lee S, Ma X, Lee W. Association between exposure to external airborne agents and autoimmune disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 263:115334. [PMID: 37567098 DOI: 10.1016/j.ecoenv.2023.115334] [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: 05/26/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
The etiology of autoimmune disease pathogeneses remains obscure, and the impact of general environmental or occupational exposure to external airborne agents (EAA) on autoimmune diseases remains understudied. This study was conducted to elucidate the association between exposure to EAA and the risk of autoimmune diseases according to exposure type. From the NHIS-NSC (2002-2019), 17,984,963 person-years were included in the data analysis. Autoimmune diseases were categorized based on the InterLymph classification. We estimated the incidence and rate ratio of autoimmune diseases according to the EAA exposure. Association between exposure and autoimmune diseases was investigated using logistic regression analysis, adjusted for potential confounders. Of the 1,082,879 participants, 86,376 (8.0%) were diagnosed with autoimmune diseases. Among these, 208 (14.1%) experienced severe exposure to EAA. Total EAA exposure was significantly associated with any autoimmune disease (OR: 1.29, 95% CI: 1.11-1.49) and organ-specific diseases (OR: 1.28, 95% CI: 1.08-1.53). Inorganic dust exposure was associated with organ-specific diseases (OR, 1.38; 95% CI: 1.01-1.81). Exposure to other dust was significantly associated with any autoimmune disease (OR: 1.35, 95% CI: 1.10-1.66), connective tissue diseases (OR: 1.43, 95% CI: 1.03-1.99), and organ-specific diseases (OR: 1.28, 95% CI: 1.00-1.65). Exposure to EAA was predominantly related to psoriasis, rheumatoid arthritis (RA), and type 1 diabetes (T1DM). We found that exposure to EAA is a potential risk factor for autoimmune diseases, especially psoriasis, RA, and T1DM. Our findings provide insight into the role of exposure to severe airborne agents in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Seunghyun Lee
- Department of Occupational and Environmental Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Wanhyung Lee
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Kim D, Yun J, Roh E, Shin HS, Kim JE. Higenamine Reduces Fine-Dust-Induced Matrix Metalloproteinase (MMP)-1 in Human Keratinocytes. PLANTS (BASEL, SWITZERLAND) 2023; 12:2479. [PMID: 37447040 DOI: 10.3390/plants12132479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/09/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023]
Abstract
Environmental pollutants such as fine dust are increasingly linked to premature skin aging. In this study, we investigated the protective effects of higenamine, a natural plant alkaloid, against fine-dust-induced skin aging in human keratinocytes (HaCaT cells). We found that higenamine significantly attenuated fine-dust-induced expression of matrix metalloproteinase-1 (MMP-1), a key enzyme involved in collagen degradation. Furthermore, higenamine was found to modulate fine-dust-induced AP-1 and NF-κB transactivation, which are crucial factors for MMP-1 transcription. Higenamine also impeded fine-dust-induced phosphorylation in specific pathways related to AP-1 and NF-κB activation, and effectively alleviated reactive oxygen species (ROS) production, a key factor in oxidative stress caused by fine dust exposure. These results suggest that higenamine exerts protective effects against fine-dust-induced skin aging, primarily through its MMP-1 inhibitory properties and ability to mitigate ROS-induced oxidative damage. Our data highlight the potential of higenamine as an effective ingredient in skincare products designed to combat environmental skin damage.
Collapse
Affiliation(s)
- DongHyeon Kim
- Department of Food Science and Biotechnology, Dongguk University-Seoul, Goyang-si 10326, Republic of Korea
- Department of Food Science and Technology, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
| | - JeaHyeok Yun
- Department of Food Science and Technology, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
| | - Eunmiri Roh
- Department of Cosmetic Science, Kwangju Women's University, Gwangju 62396, Republic of Korea
| | - Han-Seung Shin
- Department of Food Science and Biotechnology, Dongguk University-Seoul, Goyang-si 10326, Republic of Korea
| | - Jong-Eun Kim
- Department of Food Science and Technology, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
| |
Collapse
|
28
|
Zhen AX, Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Cho SJ, Hyun JW. 3-Bromo-4,5-dihydroxybenzaldehyde Protects Keratinocytes from Particulate Matter 2.5-Induced Damages. Antioxidants (Basel) 2023; 12:1307. [PMID: 37372037 DOI: 10.3390/antiox12061307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence can be activated by several stimuli, including ultraviolet radiation and air pollutants. This study aimed to evaluate the protective effect of marine algae compound 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on particulate matter 2.5 (PM2.5)-induced skin cell damage in vitro and in vivo. The human HaCaT keratinocyte was pre-treated with 3-BDB and then with PM2.5. PM2.5-induced reactive oxygen species (ROS) generation, lipid peroxidation, mitochondrial dysfunction, DNA damage, cell cycle arrest, apoptotic protein expression, and cellular senescence were measured using confocal microscopy, flow cytometry, and Western blot. The present study exhibited PM2.5-generated ROS, DNA damage, inflammation, and senescence. However, 3-BDB ameliorated PM2.5-induced ROS generation, mitochondria dysfunction, and DNA damage. Furthermore, 3-BDB reversed the PM2.5-induced cell cycle arrest and apoptosis, reduced cellular inflammation, and mitigated cellular senescence in vitro and in vivo. Moreover, the mitogen-activated protein kinase signaling pathway and activator protein 1 activated by PM2.5 were inhibited by 3-BDB. Thus, 3-BDB suppressed skin damage induced by PM2.5.
Collapse
Affiliation(s)
- Ao-Xuan Zhen
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei-Jing Piao
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung-Ah Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Suk-Ju Cho
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
29
|
Khoa ND, Li S, Phuong NL, Kuga K, Yabuuchi H, Kan-O K, Matsumoto K, Ito K. Computational fluid-particle dynamics modeling of ultrafine to coarse particles deposition in the human respiratory system, down to the terminal bronchiole. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 237:107589. [PMID: 37167881 DOI: 10.1016/j.cmpb.2023.107589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/27/2023] [Accepted: 05/05/2023] [Indexed: 05/13/2023]
Abstract
BACKGROUND AND OBJECTIVES Suspended respirable airborne particles are associated with human health risks and especially particles within the range of ultrafine (< 0.1 μm) or fine (< 2.5 μm) have a high possibility of penetrating the lung region, which is concerned to be closely related to the bronchial or alveoli tissue dosimetry. Nature complex structure of the respiratory system requires much effort to explore and comprehend the flow and the inhaled particle dynamics for precise health risk assessment. Therefore, this study applied the computational fluid-particle dynamics (CFPD) method to elucidate the deposition characteristics of ultrafine-to-coarse particles in the human respiratory tract from nostrils to the 16th generation of terminal bronchi. METHODS The realistic bronchi up to the 8th generation are precisely and perfectly generated from computed tomography (CT) images, and an artificial model compensates for the 9th-16th bronchioles. Herein, the steady airflow is simulated at constant breathing flow rates of 7.5, 15, and 30 L/min, reproducing human resting-intense activity. Then, trajectories of the particle size ranging from 0.002 - 10 μm are tracked using a discrete phase model. RESULTS Here, we report reliable results of airflow patterns and particle deposition efficiency in the human respiratory system validated against experimental data. The individual-related focal point of ultrafine and fine particles deposition rates was actualized at the 8th generation; whilst the hot-spot of the deposited coarse particles was found in the 6th generation. Lobar deposition characterizes the dominance of coarse particles deposited in the right lower lobe, whereas the left upper-lower and right lower lobes simultaneously occupy high deposition rates for ultrafine particles. Finally, the results indicate a higher deposition in the right lung compared to its counterpart. CONCLUSIONS From the results, the developed realistic human respiratory system down to the terminal bronchiole in this study, in coupling with the CFPD method, delivers the accurate prediction of a wide range of particles in terms of particle dosimetry and visualization of site-specific in the consecutive respiratory system. In addition, the series of CFPD analyses and their results are to offer in-depth information on particle behavior in human bronchioles, which may benefit health risk assessment or drug delivery studies.
Collapse
Affiliation(s)
- Nguyen Dang Khoa
- Interdisciplinary Graduate School of Engineering Sciences, Kyushu University, 6-1, Kasuga-Koen, Kasuga, Fukuoka 816-8580, Japan.
| | - Sixiao Li
- Interdisciplinary Graduate School of Engineering Sciences, Kyushu University, 6-1, Kasuga-Koen, Kasuga, Fukuoka 816-8580, Japan
| | - Nguyen Lu Phuong
- Faculty of Environment, University of Natural Resources and Environment, Ho Chi Minh, Viet Nam
| | - Kazuki Kuga
- Faculty of Engineering Sciences, Kyushu University, Fukuoka, Japan
| | - Hidetake Yabuuchi
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiko Kan-O
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Matsumoto
- Division of Respirology, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Kazuhide Ito
- Faculty of Engineering Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
30
|
Bae IA, Ha JW, Boo YC. Chlorogenic Acid, a Component of Oenanthe javanica (Blume) DC., Attenuates Oxidative Damage and Prostaglandin E2 Production Due to Particulate Matter 10 in HaCaT Keratinocytes. COSMETICS 2023. [DOI: 10.3390/cosmetics10020060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Oenanthe javanica (OJ) is a perennial herb that grows wildly or is cultivated in Asia, and it is used as food or in traditional medicine. The antioxidant and anti-inflammatory effects of OJ-derived materials have been extensively explored previously, but their effects on the cytotoxicity of air pollution are currently unknown. Therefore, the present study aimed to evaluate the effect of the hot water extract of OJ on atmospheric particulate matter 10 (PM10)-induced cytotoxicity and oxidative damage in human HaCaT keratinocytes, and to identify its active ingredient and mechanism of action. When the hot water extract of OJ was divided into methylene chloride, ethyl acetate (EA), n-butanol (BA), and water fractions, caffeic acid was enriched in the EA fraction and chlorogenic acid was enriched in the BA fraction. PM10 increased reactive oxygen species (ROS) production, lipid peroxidation, protein carbonylation, and inflammatory prostaglandin (PG) E2 production in cells. The BA fraction reduced the PM10-induced ROS production in cells more effectively than the total extract and other solvent fractions. Chlorogenic acid was more effective in reducing ROS levels than caffeic acid and N-acetyl cysteine (NAC). Chlorogenic acid attenuated the increase in lipid peroxidation and the PG E2 production of cells due to PM10 exposure. Of the genes involved in PG E2 production, phospholipase A2 group IVA (PLA2G4A), Prostaglandin-endoperoxide synthase 1 (PTGS1), and 2 (PTGS2) were transcriptionally up-regulated by PM10, whereas phospholipase A2 group IIA (PLA2G2A) was down-regulated and prostaglandin E synthetase 1 (PTGES1) and 2 (PTGES2) were a little altered. The PM10-induced increase in PLA2G4A mRNA was alleviated by chlorogenic acid and NAC. Accordingly, PM10 increased the expression levels of cytosolic phospholipase A2 (cPLA2) protein and its phosphorylated form, which were attenuated by chlorogenic acid and NAC. Thus, chlorogenic acid may attenuate the PM10-induced PG E2 production through the suppression of PLA2G4A mRNA and cPLA2 protein expressions. This study suggests that chlorogenic acid contained in OJ extract may help alleviate the oxidative damage to and inflammatory responses of the skin cells due to exposure to air pollutants.
Collapse
Affiliation(s)
- In Ah Bae
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Jae Won Ha
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Yong Chool Boo
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
31
|
Paik SJ, Kim DJ, Jung SK. Preventive Effect of Pharmaceutical Phytochemicals Targeting the Src Family of Protein Tyrosine Kinases and Aryl Hydrocarbon Receptor on Environmental Stress-Induced Skin Disease. Int J Mol Sci 2023; 24:ijms24065953. [PMID: 36983027 PMCID: PMC10056297 DOI: 10.3390/ijms24065953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The skin protects our body; however, it is directly exposed to the environment and is stimulated by various external factors. Among the various environmental factors that can threaten skin health, the effects of ultraviolet (UV) and particulate matter (PM) are considered the most notable. Repetitive exposure to ultraviolet and particulate matter can cause chronic skin diseases such as skin inflammation, photoaging, and skin cancer. The abnormal activation of the Src family of protein tyrosine kinases (SFKs) and the aryl hydrocarbon receptor (AhR) in response to UV and/or PM exposure are involved in the development and aggravation of skin diseases. Phytochemicals, chemical compounds of natural plants, exert preventive effects on skin diseases through the regulation of various signaling pathways. Therefore, this review aims to highlight the efficacy of phytochemicals as potential nutraceuticals and pharmaceutical materials for the treatment of skin diseases, primarily by targeting SFK and AhR, and to explore the underlying mechanisms of action. Future studies are essential to validate the clinical potential for the prevention and treatment of skin diseases.
Collapse
Affiliation(s)
- So Jeong Paik
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dong Joon Kim
- Department of Microbiology, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Sung Keun Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Research Institute of Tailored Food Technology, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
32
|
Feng JX, Liu L, Wang HY, Zhang J, Li XP. A soluble TLR5 is involved in PBLs activation and antibacterial immunity via TLR5M-MyD88-signaling pathway in tongue sole Cynoglossus semilaevis. Int J Biol Macromol 2023; 230:123208. [PMID: 36634796 DOI: 10.1016/j.ijbiomac.2023.123208] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
In higher vertebrates, there is only a membranal TLR5 (TLR5M), which is crucial for host defense against microbes via MyD88 signaling pathway. In teleost, both TLR5M and soluble TLR5 (TLR5S) are identified, whereas the antibacterial mechanism of TLR5S is largely unknown. In this study, we studied the immune antibacterial mechanism of Cynoglossus semilaevis TLR5S homologue (named CsTLR5S). CsTLR5S, a 71.1 kDa protein, consists of 649 amino acid residues and shares 41.7 %-57.8 % overall sequence identities with teleost TLR5S homologues. CsTLR5S contains a single extracellular domain (ECD) composed of 12 leucine-rich repeats. CsTLR5S expression was constitutively identified and upregulated by bacterial infection in tissues. In vitro recombinant CsTLR5S (rCsTLR5S) could interact with bacteria and tongue sole rTLR5M (rCsTLR5M). Furthermore, rCsTLR5S could bind to the membranal CsTLR5M of peripheral blood leukocytes (PBLs), which led to enhancing the activity and the antibacterial role of PBLs via Myd88-NF-κB pathway. In vivo rCsTLR5S could activate the Myd88-NF-κB pathway, facilitate the release of proinflammatory cytokines, and enhance the host antibacterial response against Vibrio harveyi. Moreover, the knockdown of CsTLR5M or the Myd88 inhibitor could significantly suppress the antibacterial effect of rCsTLR5S. Collectively, our findings added important insights into the TLR5S immune antibacterial property in a TLR5M-MyD88-dependent manner.
Collapse
Affiliation(s)
- Ji-Xing Feng
- School of Ocean, Yantai University, Yantai, China
| | - Ling Liu
- School of Ocean, Yantai University, Yantai, China
| | - Hong-Ye Wang
- School of Ocean, Yantai University, Yantai, China
| | - Jian Zhang
- School of Ocean, Yantai University, Yantai, China.
| | - Xue-Peng Li
- School of Ocean, Yantai University, Yantai, China.
| |
Collapse
|
33
|
Microbiota, Oxidative Stress, and Skin Cancer: An Unexpected Triangle. Antioxidants (Basel) 2023; 12:antiox12030546. [PMID: 36978794 PMCID: PMC10045429 DOI: 10.3390/antiox12030546] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Mounting evidence indicates that the microbiota, the unique combination of micro-organisms residing in a specific environment, plays an essential role in the development of a wide range of human diseases, including skin cancer. Moreover, a persistent imbalance of microbial community, named dysbiosis, can also be associated with oxidative stress, a well-known emerging force involved in the pathogenesis of several human diseases, including cutaneous malignancies. Although their interplay has been somewhat suggested, the connection between microbiota, oxidative stress, and skin cancer is a largely unexplored field. In the present review, we discuss the current knowledge on these topics, suggesting potential therapeutic strategies.
Collapse
|
34
|
Pecchillo Cimmino T, Ammendola R, Cattaneo F, Esposito G. NOX Dependent ROS Generation and Cell Metabolism. Int J Mol Sci 2023; 24:ijms24032086. [PMID: 36768405 PMCID: PMC9916913 DOI: 10.3390/ijms24032086] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Reactive oxygen species (ROS) represent a group of high reactive molecules with dualistic natures since they can induce cytotoxicity or regulate cellular physiology. Among the ROS, the superoxide anion radical (O2·-) is a key redox signaling molecule prominently generated by the NADPH oxidase (NOX) enzyme family and by the mitochondrial electron transport chain. Notably, altered redox balance and deregulated redox signaling are recognized hallmarks of cancer and are involved in malignant progression and resistance to drugs treatment. Since oxidative stress and metabolism of cancer cells are strictly intertwined, in this review, we focus on the emerging roles of NOX enzymes as important modulators of metabolic reprogramming in cancer. The NOX family includes seven isoforms with different activation mechanisms, widely expressed in several tissues. In particular, we dissect the contribute of NOX1, NOX2, and NOX4 enzymes in the modulation of cellular metabolism and highlight their potential role as a new therapeutic target for tumor metabolism rewiring.
Collapse
Affiliation(s)
- Tiziana Pecchillo Cimmino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
| | - Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: (F.C.); (G.E.)
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore S.c.a.r.l., 80131 Naples, Italy
- Correspondence: (F.C.); (G.E.)
| |
Collapse
|
35
|
Jeong YJ, Kim CU, Lee KS, Kim JH, Park SY, Jeong AY, Lee JB, Kim DJ, Park YJ, Lee MS. Pseudomonas stutzeri PM101005 inhaled with atmospheric particulate matter induces lung damage through inflammatory responses. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 317:120741. [PMID: 36435285 DOI: 10.1016/j.envpol.2022.120741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 06/16/2023]
Abstract
Atmospheric particulate matter (PM) contains a mixture of chemical and biological elements that pose threat to human health by increasing susceptibility to respiratory diseases. Although the identification of the microorganisms composing the PM has been assessed, their immunological impacts are still questionable. Here, we examined the mechanisms responsible for the pathogenicity of Pseudomonas stutzeri PM101005 (PMPS), a bacterium isolated from fine dust, in lung epithelial cells, alveolar cells, and macrophages. Relative to its comparative strain Pseudomonas stutzeri (PS), infections with PMPS induced higher production of inflammatory cytokines and chemokines, mediated by the activation of NF-κB and MAPK signaling pathways. Additionally, with three-dimensional (3D) airway spheroids which mimic the human bronchial epithelium, we confirmed that PMPS infections lead to relatively higher induction of pro-inflammatory cytokines than PM infections. Consistent results were observed in murine models as the infections with PMPS provoked greater inflammatory responses than the infections with PS. These PMPS-induced responses were mediated by the signaling pathways of the Toll-like receptors (TLRs), which regulated PMPS infection and played an important role in the expression of the antibiotic peptide β-defensin 3 (BD3) that suppressed PMPS proliferation. Moreover, PM pretreatment enhanced inflammatory responses and tissue damage of PMPS, while reducing BD3 expression. Overall, these results indicate that PM-isolated PMPS induce TLR-mediated inflammatory responses in lung tissues, and contributes to the understanding of the etiology of PM-induced respiratory damage.
Collapse
Affiliation(s)
- Yu-Jin Jeong
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Kyung-Soo Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Ji Hyung Kim
- Department of Food Science and Biotechnology, Gachon University, Seongnam, 13120, Republic of Korea
| | - Seo Young Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Ahn Young Jeong
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea; Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Jun Bong Lee
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Kangwon, 24341, Republic of Korea
| | - Doo-Jin Kim
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea; Infectious Disease Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea
| | - Young-Jun Park
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 125 Gwahak-ro, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 127 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
36
|
Arooj M, Rehman A, Hyun CL, Rafique A, Kang HK, Hyun JW, Koh YS. Diesel exhaust particulate matter impairs Toll-like receptor signaling and host defense against staphylococcal cutaneous infection in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114443. [PMID: 38321662 DOI: 10.1016/j.ecoenv.2022.114443] [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: 09/22/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
Air pollution is an emerging cause of mortality, affecting nearly 5 million people each year. Exposure to diesel exhaust fine particulate matter (PM2.5) aggravates respiratory and skin conditions. However, its impact on the protective immunity of the skin remains poorly understood. This study aimed to investigate the underlying molecular mechanism for adverse effects of PM2.5 on the host protective immunity using in vitro cell and in vivo mouse model. Intracellular translocation of Toll-like receptor 9 (TLR9) and CpG-DNA internalization were assessed in dendritic cells without or with PM2.5 treatment using immunofluorescence staining. Cytokine and nitric oxide production were measured in dendritic cells and macrophages without or with PM2.5 treatment. NF-κB and MAPK signaling was determined using western blotting. Skin disease severity, bacterial loads, and cytokine production were assessed in cutaneous Staphylococcus aureus (S. aureus) infection mouse model. PM2.5 interfered with TLR9 activation by inhibiting both TLR9 trafficking to early endosomes and CpG-DNA internalization via clathrin-mediated endocytosis. In addition, exposure to PM2.5 inhibited various TLR-mediated nitric oxide and cytokine production as well as MAPK and NF-κB signaling. PM2.5 rendered mice more susceptible to staphylococcal skin infections. Our results suggest that exposure to PM impairs TLR signaling and dampens the host defense against staphylococcal skin infections. Our data provide a novel perspective into the impact of PM on protective immunity which is paramount to revealing air pollutant-mediated toxicity on the host immunity.
Collapse
Affiliation(s)
- Madeeha Arooj
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Abdur Rehman
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Chang Lim Hyun
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Asma Rafique
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Hee-Kyoung Kang
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Jin Won Hyun
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea
| | - Young-Sang Koh
- College of Medicine, and Jeju Research Center for Natural Medicine, Jeju National University, 102 Jejudaehakno, Jeju 63243, South Korea.
| |
Collapse
|
37
|
Lee KY, Ho SC, Sun WL, Feng PH, Lin CW, Chen KY, Chuang HC, Tseng CH, Chen TT, Wu SM. Lnc-IL7R alleviates PM 2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol Toxicol 2022; 38:1097-1120. [PMID: 35303175 DOI: 10.1007/s10565-022-09709-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Long-term exposure to PM2.5 (particulate matter with an aerodynamic diameter of ≤ 2.5 μm) is associated with pulmonary injury and emphysema in patients with chronic obstructive pulmonary disease (COPD). We investigated mechanisms through which the long noncoding RNA lnc-IL7R contributes to cellular damage by inducing oxidative stress in COPD patients exposed to PM2.5. METHODS Associations of serum lnc-IL7R levels with lung function, emphysema, and previous PM2.5 exposure in COPD patients were analyzed. Reactive oxygen species and lnc-IL7R levels were measured in PM2.5-treated cells. The levels of lnc-IL7R and cellular senescence-associated genes, namely p16INK4a and p21CIP1/WAF1, were determined through lung tissue section staining. The effects of p16INK4a or p21CIP1/WAF1 regulation were examined by performing lnc-IL7R overexpression and knockdown assays. The functions of lnc-IL7R-mediated cell proliferation, cell cycle, senescence, colony formation, and apoptosis were examined in cells treated with PM2.5. Chromatin immunoprecipitation assays were conducted to investigate the epigenetic regulation of p21CIP1/WAF1. RESULTS Lnc-IL7R levels decreased in COPD patients and were negatively correlated with emphysema or PM2.5 exposure. Lnc-IL7R levels were upregulated in normal lung epithelial cells but not in COPD cells exposed to PM2.5. Lower lnc-IL7R expression in PM2.5-treated cells induced p16INK4a and p21CIP1/WAF1 expression by increasing oxidative stress. Higher lnc-IL7R expression protected against cellular senescence and apoptosis, whereas lower lnc-IL7R expression augmented injury in PM2.5-treated cells. Lnc-IL7R and the enhancer of zeste homolog 2 (EZH2) synergistically suppressed p21CIP1/WAF1 expression through epigenetic modulation. CONCLUSION Lnc-IL7R attenuates PM2.5-mediated p21CIP1/WAF1 expression through EZH2 recruitment, and its dysfunction may augment cellular injury in COPD.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
38
|
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: 48] [Impact Index Per Article: 24.0] [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
|
39
|
Yan X, Tsuji G, Hashimoto-Hachiya A, Furue M. Galactomyces Ferment Filtrate Potentiates an Anti-Inflammaging System in Keratinocytes. J Clin Med 2022; 11:6338. [PMID: 36362566 PMCID: PMC9657190 DOI: 10.3390/jcm11216338] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 12/24/2023] Open
Abstract
Skincare products play a crucial role in preventing the dry skin induced by various causes. Certain ingredients can help to improve the efficacy of skincare products. Galactomyces ferment filtrate (GFF) is such a functional ingredient. Its use originated from the empirical observation that the hands of sake brewers who deal with yeast fermentation retain a beautiful and youthful appearance. Consequently, skincare products based on GFF are widely used throughout the world. Recent studies have demonstrated that GFF activates an aryl hydrocarbon receptor (AHR) and upregulates the expression of filaggrin, a pivotal endogenous source of natural moisturizing factors, in epidermal keratinocytes. It also activates nuclear factor erythroid-2-related factor 2 (NRF2), the antioxidative master transcription factor, and exhibits potent antioxidative activity against oxidative stress induced by ultraviolet irradiation and proinflammatory cytokines, which also accelerate inflammaging. GFF-mediated NRF2 activation downregulates the expression of CDKN2A, which is known to be overexpressed in senescent keratinocytes. Moreover, GFF enhances epidermal terminal differentiation by upregulating the expression of caspase-14, claudin-1, and claudin-4. It also promotes the synthesis of the antiinflammatory cytokine IL-37 and downregulates the expression of proallergic cytokine IL-33 in keratinocytes. In addition, GFF downregulates the expression of the CXCL14 and IL6R genes, which are involved in inflammaging. These beneficial properties might underpin the potent barrier-protecting and anti-inflammaging effects of GFF-containing skin formulae.
Collapse
Affiliation(s)
- Xianghong Yan
- SK-II Science Communications, Kobe Innovation Center, Procter and Gamble Innovation, Kobe 651-0088, Japan
| | - Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiko Hashimoto-Hachiya
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
40
|
Kim DH, Lee H, Hwangbo H, Kim SY, Ji SY, Kim MY, Park SK, Park SH, Kim MY, Kim GY, Cheong J, Nam SW, Choi YH. Particulate matter 2.5 promotes inflammation and cellular dysfunction via reactive oxygen species/p38 MAPK pathway in primary rat corneal epithelial cells. Cutan Ocul Toxicol 2022; 41:273-284. [PMID: 36097682 DOI: 10.1080/15569527.2022.2122489] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE Numerous studies have linked particulate matter 2.5 (PM2.5) to ocular surface diseases, but few studies have been conducted on the biological effect of PM2.5 on the cornea. The objective of the present study was to evaluate the harmful effect of PM2.5 on primary rat corneal epithelial cells (RCECs) in vitro and identify the toxic mechanism involved. MATERIALS AND METHODS Primary cultured RCECs were characterized by pan-cytokeratin (CK) staining. In PM2.5-exposed RCECs, cell viability, microarray gene expression, inflammatory cytokine levels, mitochondrial damage, DNA double-strand break and signaling pathway were investigated. RESULTS Exposure to PM2.5 induced cytotoxicity and morphological changes in RCECs. In addition, PM2.5 markedly up-regulated pro-inflammatory mediators but down-regulated the wound healing-related transforming growth factor-β. Furthermore, PM2.5 promoted mitochondrial reactive oxygen species (ROS) production and mediated cellular damage to mitochondria and DNA, whereas these cellular alterations induced by PM2.5 were markedly suppressed by a potential ROS scavenger. Noteworthy, removal of ROS selectively down-regulated the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and the activation of the nuclear factor-κB (NF-κB) p65 in PM2.5-stimulated cells. Additionally, SB203580, a p38 MAPK inhibitor, markedly suppressed these PM2.5-mediated cellular dysfunctions. CONCLUSIONS Taken together, our findings show that PM2.5 can promote the ROS/p38 MAPK/NF-κB signaling pathway and lead to mitochondrial damage and DNA double-strand break, which is ultimately caused inflammation and cytotoxicity in RCECs. These findings indicate that the ROS/p38 MAPK/NF-κB signaling pathway is one mechanism involved in PM2.5-induced ocular surface disorders.
Collapse
Affiliation(s)
- Da Hye Kim
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Molecular Biology, xxxx, Busan 46241, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Convergence Medicine, xxxx, Yangsan 50612, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Biochemistry, xxxx, Busan 47227, Republic of Korea
| | - So Young Kim
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Biochemistry, xxxx, Busan 47227, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Biochemistry, xxxx, Busan 47227, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Biochemistry, xxxx, Busan 47227, Republic of Korea
| | - Seh-Kwang Park
- Research and Development Department, xxxx., Busan 47195, Republic of Korea.,xxxx, Seoul 05551, Republic of Korea
| | - Sung-Ho Park
- Research and Development Department, xxxx., Busan 47195, Republic of Korea.,xxxx, Seoul 05551, Republic of Korea
| | - Mi-Young Kim
- Research and Development Department, xxxx., Busan 47195, Republic of Korea.,xxxx, Seoul 05551, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, xxxx, Jeju 63243, Republic of Korea
| | - Jaehun Cheong
- Department of Molecular Biology, xxxx, Busan 46241, Republic of Korea
| | - Soo-Wan Nam
- Department of Smart Bio-Health, xxxx, Busan 47340, Republic of Korea.,Department of Biomedical Engineering and Biotechnology Major, Division of Applied Bioengineering, College of Engineering, xxxx, Busan 47340, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, xxxx, Busan 47340, Republic of Korea.,Department of Biochemistry, xxxx, Busan 47227, Republic of Korea.,Department of Smart Bio-Health, xxxx, Busan 47340, Republic of Korea.,Core-Facility Center for Tissue Regeneration, xxxx, Busan 47340, Republic of Korea
| |
Collapse
|
41
|
Zhang Y, Li Y, Zhou L, Yuan X, Wang Y, Deng Q, Deng Z, Xu S, Wang Q, Xie H, Li J. Nav1.8 in keratinocytes contributes to ROS-mediated inflammation in inflammatory skin diseases. Redox Biol 2022; 55:102427. [PMID: 35952475 PMCID: PMC9372634 DOI: 10.1016/j.redox.2022.102427] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/21/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS)-activated proinflammatory signals in keratinocytes play a crucial role in the immunoregulation of inflammatory skin diseases, including rosacea and psoriasis. Nav1.8 is a voltage-gated sodium ion channel, and its abnormal expression in the epidermal layer contributes to pain hypersensitivity in the skin. However, whether and how epidermal Nav1.8 is involved in skin immunoregulation remains unclear. This study was performed to identify the therapeutic role of Nav1.8 in inflammatory skin disorders. We found that Nav1.8 expression was significantly upregulated in the epidermis of rosacea and psoriasis skin lesions. Nav1.8 knockdown ameliorated skin inflammation in LL37-and imiquimod-induced inflammation mouse models. Transcriptome sequencing results indicated that Nav1.8 regulated the expression of pro-inflammatory mediators (IL1β and IL6) in keratinocytes, thereby contributing to immune infiltration in inflammatory skin disorders. In vitro, tumor necrosis factor alpha (TNFα), a cytokine that drives the development of various inflammatory skin disorders, increased Nav1.8 expression in keratinocytes. Knockdown of Nav1.8 eliminated excess ROS production, thereby attenuating the TNFα-induced production of inflammatory mediators; however, a Nav1.8 blocker did not have the same effect. Mechanistically, Nav1.8 reduced superoxide dismutase 2 (SOD2) activity by directly binding to SOD2 to prevent its deacetylation and mitochondrial localization, subsequently inducing ROS accumulation. Collectively, our study describes a central role for Nav1.8 in regulating pro-inflammatory responses in the skin and indicates a novel therapeutic strategy for rosacea and psoriasis.
Collapse
Affiliation(s)
- Yiya Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yangfan Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Lei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Yuan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Yaling Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Qing Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Qian Wang
- Hunan Binsis Biotechnology Co., Ltd, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
42
|
Fitoussi R, Faure MO, Beauchef G, Achard S. Human skin responses to environmental pollutants: A review of current scientific models. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 306:119316. [PMID: 35469928 DOI: 10.1016/j.envpol.2022.119316] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
Whatever the exposure route, chemical, physical and biological pollutants modify the whole organism response, leading to nerve, cardiac, respiratory, reproductive, and skin system pathologies. Skin acts as a barrier for preventing pollutant modifications. This review aims to present the available scientific models, which help investigate the impact of pollution on the skin. The research question was "Which experimental models illustrate the impact of pollution on the skin in humans?" The review covered a period of 10 years following a PECO statement on in vitro, ex vivo, in vivo and in silico models. Of 582 retrieved articles, 118 articles were eligible. In oral and inhalation routes, dermal exposure had an important impact at both local and systemic levels. Healthy skin models included primary cells, cell lines, co-cultures, reconstructed human epidermis, and skin explants. In silico models estimated skin exposure and permeability. All pollutants affected the skin by altering elasticity, thickness, the structure of epidermal barrier strength, and dermal extracellular integrity. Some specific models concerned wound healing or the skin aging process. Underlying mechanisms were an exacerbated inflammatory skin reaction with the modulation of several cytokines and oxidative stress responses, ending with apoptosis. Pathological skin models revealed the consequences of environmental pollutants on psoriasis, atopic dermatitis, and tumour development. Finally, scientific models were used for evaluating the safety and efficacy of potential skin formulations in preventing the skin aging process or skin irritation after repeated contact. The review gives an overview of scientific skin models used to assess the effects of pollutants. Chemical and physical pollutants were mainly represented while biological contaminants were little studied. In future developments, cell hypoxia and microbiota models may be considered as more representative of clinical situations. Models considering humidity and temperature variations may reflect the impact of these changes.
Collapse
Affiliation(s)
| | - Marie-Odile Faure
- Scientific Consulting For You, 266 avenue Daumesnil, 75012, PARIS, France
| | | | - Sophie Achard
- HERA Team (Health Environmental Risk Assessment), INSERM UMR1153, CRESS-INRAE, Université Paris Cité, Faculté de Pharmacie, 4 avenue de l'Observatoire, 75270 CEDEX 06, PARIS, France.
| |
Collapse
|
43
|
Hesperidin Protects Human HaCaT Keratinocytes from Particulate Matter 2.5-Induced Apoptosis via the Inhibition of Oxidative Stress and Autophagy. Antioxidants (Basel) 2022; 11:antiox11071363. [PMID: 35883854 PMCID: PMC9312010 DOI: 10.3390/antiox11071363] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Numerous epidemiological studies have reported that particulate matter 2.5 (PM2.5) causes skin aging and skin inflammation and impairs skin homeostasis. Hesperidin, a bioflavonoid that is abundant in citrus species, reportedly has anti-inflammatory properties. In this study, we evaluated the cytoprotective effect of hesperidin against PM2.5-mediated damage in a human skin cell line (HaCaT). Hesperidin reduced PM2.5-induced intracellular reactive oxygen species (ROS) generation and oxidative cellular/organelle damage. PM2.5 increased the proportion of acridine orange-positive cells, levels of autophagy-related proteins, beclin-1 and microtubule-associated protein light chain 3, and apoptosis-related proteins, B-cell lymphoma-2-associated X protein, cleaved caspase-3, and cleaved caspase-9. However, hesperidin ameliorated PM2.5-induced autophagy and apoptosis. PM2.5 promoted cellular apoptosis via mitogen-activated protein kinase (MAPK) activation by promoting the phosphorylation of extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38. The MAPK inhibitors U0126, SP600125, and SB203580 along with hesperidin exerted a protective effect against PM2.5-induced cellular apoptosis. Furthermore, hesperidin restored PM2.5-mediated reduction in cell viability via Akt activation; this was also confirmed using LY294002 (a phosphoinositide 3-kinase inhibitor). Overall, hesperidin shows therapeutic potential against PM2.5-induced skin damage by mitigating excessive ROS accumulation, autophagy, and apoptosis.
Collapse
|
44
|
Lin CM, Huang TH, Chi MC, Guo SE, Lee CW, Hwang SL, Shi CS. N-acetylcysteine alleviates fine particulate matter (PM2.5)-induced lung injury by attenuation of ROS-mediated recruitment of neutrophils and Ly6C high monocytes and lung inflammation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113632. [PMID: 35594827 DOI: 10.1016/j.ecoenv.2022.113632] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Exposure to particulate matter (PM) may contribute to lung inflammation and injury. The therapeutic effect of N-acetylcysteine (NAC), a well-known antioxidant, with regards to the prevention and treatment of fine PM (PM2.5)-induced lung injury is poorly understood. This study aimed to determine the effect of PM2.5 on the recruitment of neutrophils and Ly6Chigh monocytes into lung alveoli and the production of proinflammatory proteins by stimulating the generation of reactive oxygen species (ROS), and to investigate the therapeutic effect of NAC on PM2.5-induced lung injury. METHODS C57BL/6 mice were exposed to a single administration of PM2.5 (200 μg/100 μl/mouse) or phosphate-buffered saline (control) via intratracheal instillation. The mice were injected intratracheally via a microsprayer aerosolizer with NAC (20 or 40 mg/kg) 1 h before PM2.5 instillation and 24 h after PM2.5 instillation. Total protein, VEGF, IL-6, and TNF-α in bronchoalveolar lavage fluid (BALF) were measured. Oxidative stress was evaluated by determining levels of malondialdehyde (MDA) and nitrite in BALF. Flow cytometric analysis was used to identify and quantify neutrophils and Ly6Chigh and Ly6Clow monocyte subsets. RESULTS Neutrophil count, total protein, and VEGF content in BALF significantly increased after PM2.5 exposure and reached the highest level on day 2. Increased levels of TNF-alpha, IL-6, nitrite, and MDA in BALF were also noted. Flow cytometric analysis showed increased recruitment of neutrophils and Ly6Chigh, but not Ly6Clow monocytes, into lung alveoli. Treatment with NAC via the intratracheal spray significantly attenuated the recruitment of neutrophils and Ly6Chigh monocytes into lung alveoli in PM2.5-treated mice in a dose-dependent manner. Furthermore, NAC significantly attenuated the production of total protein, VEGF, nitrite, and MDA in the mice with PM2.5-induced lung injury in a dose-dependent manner. CONCLUSION PM2.5-induced lung injury caused by the generation of oxidative stress led to the recruitment of neutrophils and Ly6Chigh monocytes, and production of inflammatory proteins. NAC treatment alleviated PM2.5-induced lung injury by attenuating the ROS-mediated recruitment of neutrophils and Ly6Chigh monocytes and lung inflammation.
Collapse
Affiliation(s)
- Chieh-Mo Lin
- Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chang Gung Medical Foundation, Puzi City, Chiayi County, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Department of Nursing, Chang Gung University of Science and Technology, Chiayi Campus, Puzi City, Chiayi County, Taiwan
| | - Tzu-Hsiung Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
| | - Miao-Ching Chi
- Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chang Gung Medical Foundation, Puzi City, Chiayi County, Taiwan; Chronic Disease and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; Department of Respiratory Care, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Su-Er Guo
- Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chang Gung Medical Foundation, Puzi City, Chiayi County, Taiwan; Chronic Disease and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; Department of Nursing and Graduate Institute of Nursing, College of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Chiang-Wen Lee
- Department of Safety Health and Environmental Engineering, Ming Chi University of Technology, New Taipei City, Taiwan; Department of Nursing, Division of Basic Medical Sciences, Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan
| | - Su-Lun Hwang
- Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Chang Gung Medical Foundation, Puzi City, Chiayi County, Taiwan; Chronic Disease and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan; Department of Nursing and Graduate Institute of Nursing, College of Nursing, Chang Gung University of Science and Technology, Puzi City, Chiayi County, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan; Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County, Taiwan.
| |
Collapse
|
45
|
Li XP, Sun JQ, Sui ZH, Zhang J, Feng JX. Membrane orthologs of TLR5 of tongue sole Cynoglossus semilaevis: Expression patterns, signaling pathway and antibacterial property. FISH & SHELLFISH IMMUNOLOGY 2022; 126:131-140. [PMID: 35618170 DOI: 10.1016/j.fsi.2022.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/05/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Mammalian toll-like receptor 5 (TLR5) is crucial for recognizing bacterial flagellin and initiating the inflammatory signaling cascades via myeloid differentiation factor 88 (MyD88) signaling pathway, which plays vital roles in innate immune against pathogenic bacteria. Herein, we reported the signaling pathway and antibacterial property of tongue sole (Cynoglossus semilaevis) membrane forms of TLR5 (i.e. CsTLR5M1and CsTLR5M2). CsTLR5M1/M2 contain 936 and 885 amino acid residues respectively. CsTLR5M1 shares 86.7% overall sequence identities with CsTLR5M2. CsTLR5M1/M2 possess the same extracellular domain (ECD) and transmembrane domain (TMD), but the different toll-interleukin-1 receptor (TIR) domain. CsTLR5M1/M2 expression occurred constitutively in multiple tissues and regulated by bacterial stimulation. Recombinant CsTLR5M1/M2 (rCsTLR5M) could bind to flagellin and Gram-negative/positive bacteria, which could suppress bacterial growth. Stimulation of the CsTLR5M pathway by flagellin resulted in increased expression of MyD88-dependent signaling molecules and inflammatory cytokines. Blocking rCsTLR5M by antibody markedly reduced the phagocytosis and ROS production of peripheral blood leukocytes (PBLs), which in turn in vivo promoted the dissemination of bacteria. Overall, these observations add new insights into the signaling pathway and immune function of teleost TLR5M.
Collapse
Affiliation(s)
- Xue-Peng Li
- School of Ocean, Yantai University, Yantai, China.
| | - Jia-Qi Sun
- School of Ocean, Yantai University, Yantai, China
| | - Zhi-Hai Sui
- School of Life Science, Linyi University, Linyi, China
| | - Jian Zhang
- School of Ocean, Yantai University, Yantai, China
| | - Ji-Xing Feng
- School of Ocean, Yantai University, Yantai, China
| |
Collapse
|
46
|
Lee E, Ahn H, Park S, Kim G, Kim H, Noh MG, Kim Y, Yeon JS, Park H. Staphylococcus epidermidis WF2R11 Suppresses PM 2.5-Mediated Activation of the Aryl Hydrocarbon Receptor in HaCaT Keratinocytes. Probiotics Antimicrob Proteins 2022; 14:915-933. [PMID: 35727505 PMCID: PMC9474527 DOI: 10.1007/s12602-022-09922-8] [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] [Accepted: 01/21/2022] [Indexed: 11/09/2022]
Abstract
The skin supports a diverse microbiome whose imbalance is related to skin inflammation and diseases. Exposure to fine particulate matter (PM2.5), a major air pollutant, can adversely affect the skin microbiota equilibrium. In this study, the effect and mechanism of PM2.5 exposure in HaCaT keratinocytes were investigated. PM2.5 stimulated the aryl hydrocarbon receptor (AhR) to produce reactive oxygen species (ROS) in HaCaT cells, leading to mitochondrial dysfunction and intrinsic mitochondrial apoptosis. We observed that the culture medium derived from a particular skin microbe, Staphylococcus epidermidis WF2R11, remarkably reduced oxidative stress in HaCaT cells caused by PM2.5-mediated activation of the AhR pathway. Staphylococcus epidermidis WF2R11 also exhibited inhibition of ROS-induced inflammatory cytokine secretion. Herein, we demonstrated that S. epidermidis WF2R11 could act as a suppressor of AhRs, affect cell proliferation, and inhibit apoptosis. Our results highlight the importance of the clinical application of skin microbiome interventions in the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Eulgi Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyeok Ahn
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Shinyoung Park
- Genome and Company, Pangyo-ro 253, Bundang-gu, Seoungnam-si, Gyeonggi-do, 13486, Republic of Korea
| | - Gihyeon Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hyun Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Myung-Giun Noh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Yunjae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jae-Sung Yeon
- Genome and Company, Pangyo-ro 253, Bundang-gu, Seoungnam-si, Gyeonggi-do, 13486, Republic of Korea
| | - Hansoo Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea. .,Genome and Company, Pangyo-ro 253, Bundang-gu, Seoungnam-si, Gyeonggi-do, 13486, Republic of Korea.
| |
Collapse
|
47
|
Grishanova AY, Perepechaeva ML. Aryl Hydrocarbon Receptor in Oxidative Stress as a Double Agent and Its Biological and Therapeutic Significance. Int J Mol Sci 2022; 23:6719. [PMID: 35743162 PMCID: PMC9224361 DOI: 10.3390/ijms23126719] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) has long been implicated in the induction of a battery of genes involved in the metabolism of xenobiotics and endogenous compounds. AhR is a ligand-activated transcription factor necessary for the launch of transcriptional responses important in health and disease. In past decades, evidence has accumulated that AhR is associated with the cellular response to oxidative stress, and this property of AhR must be taken into account during investigations into a mechanism of action of xenobiotics that is able to activate AhR or that is susceptible to metabolic activation by enzymes encoded by the genes that are under the control of AhR. In this review, we examine various mechanisms by which AhR takes part in the oxidative-stress response, including antioxidant and prooxidant enzymes and cytochrome P450. We also show that AhR, as a participant in the redox balance and as a modulator of redox signals, is being increasingly studied as a target for a new class of therapeutic compounds and as an explanation for the pathogenesis of some disorders.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Timakova Str. 2, 630117 Novosibirsk, Russia;
| |
Collapse
|
48
|
Bae IA, Ha JW, Choi JY, Boo YC. Antioxidant Effects of Korean Propolis in HaCaT Keratinocytes Exposed to Particulate Matter 10. Antioxidants (Basel) 2022; 11:antiox11040781. [PMID: 35453466 PMCID: PMC9032284 DOI: 10.3390/antiox11040781] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Air pollution causes oxidative stress that leads to inflammatory diseases and premature aging of the skin. The purpose of this study was to examine the antioxidant effect of Korean propolis on oxidative stress in human epidermal HaCaT keratinocytes exposed to particulate matter with a diameter of less than 10 μm (PM10). The total ethanol extract of propolis was solvent-fractionated with water and methylene chloride to divide into a hydrophilic fraction and a lipophilic fraction. The lipophilic fraction of propolis was slightly more cytotoxic, and the hydrophilic fraction was much less cytotoxic than the total extract. The hydrophilic fraction did not affect the viability of cells exposed to PM10, but the total propolis extract and the lipophilic fraction aggravated the toxicity of PM10. The total extract and hydrophilic fraction inhibited PM10-induced ROS production and lipid peroxidation in a concentration-dependent manner, whereas the lipophilic fraction did not show such effects. High-performance liquid chromatography with photodiode array detection (HPLC-DAD) analysis showed that the hydrophilic fraction contained phenylpropanoids, such as caffeic acid, p-coumaric acid, and ferulic acid, whereas the lipophilic faction contained caffeic acid phenethyl ester (CAPE). The former three compounds inhibited PM10-induced ROS production, lipid peroxidation, and/or glutathione oxidation, and ferulic acid was the most effective among them, but CAPE exhibited cytotoxicity and aggravated the toxicity of PM10. This study suggests that Korean propolis, when properly purified, has the potential to be used as a cosmetic material that helps to alleviate the skin toxicity of air pollutants.
Collapse
Affiliation(s)
- In Ah Bae
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea; (I.A.B.); (J.W.H.); (J.Y.C.)
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
| | - Jae Won Ha
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea; (I.A.B.); (J.W.H.); (J.Y.C.)
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
| | - Joon Yong Choi
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea; (I.A.B.); (J.W.H.); (J.Y.C.)
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
| | - Yong Chool Boo
- Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea; (I.A.B.); (J.W.H.); (J.Y.C.)
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
- Correspondence: ; Tel.: +82-53-420-4946
| |
Collapse
|
49
|
Ni Q, Zhang P, Li Q, Han Z. Oxidative Stress and Gut Microbiome in Inflammatory Skin Diseases. Front Cell Dev Biol 2022; 10:849985. [PMID: 35321240 PMCID: PMC8937033 DOI: 10.3389/fcell.2022.849985] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress plays a dominant role in inflammatory skin diseases. Emerging evidence has shown that the close interaction occurred between oxidative stress and the gut microbiome. Overall, in this review, we have summarized the impact of oxidative stress and gut microbiome during the progression and treatment for inflammatory skin diseases, the interactions between gut dysbiosis and redox imbalance, and discussed the potential possible role of oxidative stress in the gut-skin axis. In addition, we have also elucidated the promising gut microbiome/redox-targeted therapeutic strategies for inflammatory skin diseases.
Collapse
Affiliation(s)
- Qingrong Ni
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Ping Zhang
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Qiang Li
- Department of Dermatology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
| | - Zheyi Han
- Department of Gastroenterology, Air Force Medical Center, Fourth Military Medical University, Beijing, China
- *Correspondence: Zheyi Han,
| |
Collapse
|
50
|
Zhu S, Li X, Dang B, Wu F, Wang C, Lin C. Lycium Barbarum polysaccharide protects HaCaT cells from PM2.5-induced apoptosis via inhibiting oxidative stress, ER stress and autophagy. Redox Rep 2022; 27:32-44. [PMID: 35130817 PMCID: PMC8843200 DOI: 10.1080/13510002.2022.2036507] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Objectives: Lycium barbarum polysaccharide (LBP) is a natural polysaccharide extracted from Lycium barbarum that has anti-inflammatory, anti-apoptotic and anti-aging effects, and plays a role in the prevention and treatment of various diseases. In this study, we investigated the therapeutic effect of LBP on particulate matter 2.5 (PM2.5)-induced skin damage. Methods: Cell viability was analyzed by MTT and LDH assays. Apoptosis was analyzed by Annexin V-FITC/PI staining. Oxidative stress/damage were assessed by intracellular ROS levels, MDA content and SOD activity. The intracellular protein expression was analyzed by Western blot. Mitochondrial damage was assayed by mitochondrial membrane potential with JC-1 probe. LC3-GFP adenovirus was transfected into HaCaT cells to analyze intracellular autophagosome levels. Results: In PM2.5-treated HaCaT cells, LBP pretreatment reduced PM2.5-induced cytotoxicity, ameliorated cell morphology and reduced cell apoptosis. LBP also inhibited the expression levels of GRP78 and CHOP, reduced the conversion of LC3I to LC3II, inhibited Bax protein and activated Bcl-2 protein. Furthermore, LBP inhibited PM2.5-induced mitochondrial autophagy (mitophagy) and mitochondrial damage. PM2.5-induced autophagy was regulated by endoplasmic reticulum (ER) stress. Conclusion: LBP protects skin cells from PM2.5-induced cytotoxicity by regulating the oxidative stress-ER stress-autophagy-apoptosis signaling axis, revealing that LBP has a great potential for the skin protection.
Collapse
Affiliation(s)
- Sen Zhu
- School of Life Sciences, Lanzhou University, Lanzhou, People's Republic of China
| | - Xuan Li
- Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Bingrong Dang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People's Republic of China
| | - Fen Wu
- School of Life Sciences, Lanzhou University, Lanzhou, People's Republic of China
| | - Chunming Wang
- School of Life Sciences, Lanzhou University, Lanzhou, People's Republic of China
| | - Changjun Lin
- School of Life Sciences, Lanzhou University, Lanzhou, People's Republic of China
| |
Collapse
|